393
Views
38
CrossRef citations to date
0
Altmetric
Review

Biogenesis, Membrane Trafficking, Functions, and Next Generation Nanotherapeutics Medicine of Extracellular Vesicles

ORCID Icon, ORCID Icon, ORCID Icon, & ORCID Icon
Pages 3357-3383 | Published online: 18 May 2021

References

  • van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228. doi:10.1038/nrm.2017.125
  • Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17. doi:10.1038/s41556-018-0250-9
  • Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750. doi:10.1080/20013078.2018.1535750
  • Witwer KW, Théry C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J Extracell Vesicles. 2019;8(1):1648167. doi:10.1080/20013078.2019.1648167
  • Lee C, Mitsialis SA, Aslam M, et al. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation. 2012;126(22):2601–2611. doi:10.1161/circulationaha.112.114173
  • Buzas EI, György B, Nagy G, Falus A, Gay S. Emerging role of extracellular vesicles in inflammatory diseases. Nat Rev Rheumatol. 2014;10(6):356–364. doi:10.1038/nrrheum.2014.19
  • Povero D, Eguchi A, Li H, et al. Circulating extracellular vesicles with specific proteome and liver microRNAs are potential biomarkers for liver injury in experimental fatty liver disease. PLoS One. 2014;9(12):e113651. doi:10.1371/journal.pone.0113651
  • Rabinowits G, Gerçel-Taylor C, Day JM, Taylor DD, Kloecker GH. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10(1):42–46. doi:10.3816/CLC.2009.n.006
  • Tan SS, Yin Y, Lee T, et al. Therapeutic MSC exosomes are derived from lipid raft microdomains in the plasma membrane. J Extracell Vesicles. 2013;2. doi:10.3402/jev.v2i0.22614
  • Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9(8):581–593. doi:10.1038/nri2567
  • Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36(3):301–312. doi:10.1007/s10571-016-0366-z
  • Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–383. doi:10.1083/jcb.201211138
  • Gould SJ, Raposo G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles. 2013;2(1):20389. doi:10.3402/jev.v2i0.20389
  • Trams EG, Lauter CJ, Salem J Jr, Heine U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim Biophys Acta. 1981;645(1):63–70. doi:10.1016/0005-2736(81)90512-5
  • Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412–9420. doi:10.1016/S0021-9258(18)48095-7
  • Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161–1172. doi:10.1084/jem.183.3.1161
  • Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600. doi:10.1038/nm0598-594
  • Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51. doi:10.1016/j.semcdb.2015.02.010
  • Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci. 2000;113(Pt 19):3365–3374. doi:10.1242/jcs.113.19.3365
  • Russell AE, Jun S, Sarkar S, et al. Extracellular vesicles secreted in response to cytokine exposure increase mitochondrial oxygen consumption in recipient cells. Front Cell Neurosci. 2019;13:51. doi:10.3389/fncel.2019.00051
  • Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–445.e418. doi:10.1016/j.cell.2019.02.029
  • Hsu C, Morohashi Y, Yoshimura S, et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J Cell Biol. 2010;189(2):223–232. doi:10.1083/jcb.200911018
  • Ostrowski M, Carmo NB, Krumeich S, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):19–30; sup pp 11–13. doi:10.1038/ncb2000
  • 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. doi:10.1016/j.celrep.2013.10.050
  • 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(Pt 24):5553–5565. doi:10.1242/jcs.128868
  • 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. doi:10.1083/jcb.201601025
  • Lu A, Wawro P, Morgens DW, Portela F, Bassik MC, Pfeffer SR. Genome-wide interrogation of extracellular vesicle biology using barcoded miRNAs. Elife. 2018;7. doi:10.7554/eLife.41460
  • Yeung V, Webber JP, Dunlop EA, et al. Rab35-dependent extracellular nanovesicles are required for induction of tumour supporting stroma. Nanoscale. 2018;10(18):8547–8559. doi:10.1039/c8nr02417k
  • Graves LE, Ariztia EV, Navari JR, Matzel HJ, Stack MS, Fishman DA. Proinvasive properties of ovarian cancer ascites-derived membrane vesicles. Cancer Res. 2004;64(19):7045–7049. doi:10.1158/0008-5472.Can-04-1800
  • Kim DK, Kang B, Kim OY, et al. EVpedia: an integrated database of high-throughput data for systemic analyses of extracellular vesicles. J Extracell Vesicles. 2013;2. doi:10.3402/jev.v2i0.20384
  • Savina A, Furlán M, Vidal M, Colombo MI. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J Biol Chem. 2003;278(22):20083–20090. doi:10.1074/jbc.M301642200
  • Holleman J, Marchese A. The ubiquitin ligase deltex-3l regulates endosomal sorting of the G protein-coupled receptor CXCR4. Mol Biol Cell. 2014;25(12):1892–1904. doi:10.1091/mbc.E13-10-0612
  • van Niel G, Charrin S, Simoes S, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21(4):708–721. doi:10.1016/j.devcel.2011.08.019
  • 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. doi:10.1016/j.cub.2009.09.059
  • Perez-Hernandez D, Gutiérrez-Vázquez C, Jorge I, et al. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J Biol Chem. 2013;288(17):11649–11661. doi:10.1074/jbc.M112.445304
  • Vanni I, Alama A, Grossi F, Dal Bello MG, Coco S. Exosomes: a new horizon in lung cancer. Drug Discov Today. 2017;22(6):927–936. doi:10.1016/j.drudis.2017.03.004
  • Kadota T, Yoshioka Y, Fujita Y, Kuwano K, Ochiya T. Extracellular vesicles in lung cancer-from bench to bedside. Semin Cell Dev Biol. 2017;67:39–47. doi:10.1016/j.semcdb.2017.03.001
  • Syn NL, Wang L, Chow EK, Lim CT, Goh BC. Exosomes in cancer nanomedicine and immunotherapy: prospects and challenges. Trends Biotechnol. 2017;35(7):665–676. doi:10.1016/j.tibtech.2017.03.004
  • Yang B, Chen Y, Shi J. Exosome biochemistry and advanced nanotechnology for next-generation theranostic platforms. Adv Mater. 2019;31(2):e1802896. doi:10.1002/adma.201802896
  • Zhang H, Freitas D, Kim HS, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20(3):332–343. doi:10.1038/s41556-018-0040-4
  • Zhang Q, Higginbotham JN, Jeppesen DK, et al. Transfer of functional cargo in exomeres. Cell Rep. 2019;27(3):940–954.e946. doi:10.1016/j.celrep.2019.01.009
  • Simons M, Raposo G. Exosomes–vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575–581. doi:10.1016/j.ceb.2009.03.007
  • Balaj L, Lessard R, Dai L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180. doi:10.1038/ncomms1180
  • Ashcroft BA, de Sonneville J, Yuana Y, et al. Determination of the size distribution of blood microparticles directly in plasma using atomic force microscopy and microfluidics. Biomed Microdevices. 2012;14(4):641–649. doi:10.1007/s10544-012-9642-y
  • Dalton AJ. Microvesicles and vesicles of multivesicular bodies versus “virus-like” particles. J Natl Cancer Inst. 1975;54(5):1137–1148. doi:10.1093/jnci/54.5.1137
  • Gabel V, Miglis M, Zeitzer JM. Effect of artificial dawn light on cardiovascular function, alertness, and balance in middle-aged and older adults. Sleep. 2020;43(10). doi:10.1093/sleep/zsaa082
  • Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J Transl Med. 2011;9:86. doi:10.1186/1479-5876-9-86
  • Hata T, Murakami K, Nakatani H, Yamamoto Y, Matsuda T, Aoki N. Isolation of bovine milk-derived microvesicles carrying mRNAs and microRNAs. Biochem Biophys Res Commun. 2010;396(2):528–533. doi:10.1016/j.bbrc.2010.04.135
  • Wiggins R, Glatfelter A, Kshirsagar B, Beals T. Lipid microvesicles and their association with procoagulant activity in urine and glomeruli of rabbits with nephrotoxic nephritis. Lab Invest. 1987;56(3):264–272.
  • Momen-Heravi F, Balaj L, Alian S, et al. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013;394(10):1253–1262. doi:10.1515/hsz-2013-0141
  • Livshits MA, Khomyakova E, Evtushenko EG, et al. Isolation of exosomes by differential centrifugation: theoretical analysis of a commonly used protocol. Sci Rep. 2015;5:17319. doi:10.1038/srep17319
  • Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7(3):789–804. doi:10.7150/thno.18133
  • Lobb RJ, Becker M, Wen SW, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031. doi:10.3402/jev.v4.27031
  • Koliha N, Wiencek Y, Heider U, et al. A novel multiplex bead-based platform highlights the diversity of extracellular vesicles. J Extracell Vesicles. 2016;5:29975. doi:10.3402/jev.v5.29975
  • Chen C, Skog J, Hsu CH, et al. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab Chip. 2010;10(4):505–511. doi:10.1039/b916199f
  • Brennan K, Martin K, FitzGerald SP, et al. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci Rep. 2020;10(1):1039. doi:10.1038/s41598-020-57497-7
  • Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A. 2021;1636:461773. doi:10.1016/j.chroma.2020.461773
  • Johnstone RM, Mathew A, Mason AB, Teng K. Exosome formation during maturation of mammalian and avian reticulocytes: evidence that exosome release is a major route for externalization of obsolete membrane proteins. J Cell Physiol. 1991;147(1):27–36. doi:10.1002/jcp.1041470105
  • Février B, Raposo G. Exosomes: endosomal-derived vesicles shipping extracellular messages. Curr Opin Cell Biol. 2004;16(4):415–421. doi:10.1016/j.ceb.2004.06.003
  • Wolf P. The nature and significance of platelet products in human plasma. Br J Haematol. 1967;13(3):269–288. doi:10.1111/j.1365-2141.1967.tb08741.x
  • Stein JM, Luzio JP. Ectocytosis caused by sublytic autologous complement attack on human neutrophils. The sorting of endogenous plasma-membrane proteins and lipids into shed vesicles. Biochem J. 1991;274(Pt 2):381–386. doi:10.1042/bj2740381
  • Stahl PD, Raposo G. Extracellular vesicles: exosomes and microvesicles, integrators of homeostasis. Physiology (Bethesda). 2019;34(3):169–177. doi:10.1152/physiol.00045.2018
  • Sims PJ, Faioni EM, Wiedmer T, Shattil SJ. Complement proteins C5b-9 cause release of membrane vesicles from the platelet surface that are enriched in the membrane receptor for coagulation factor Va and express prothrombinase activity. J Biol Chem. 1988;263(34):18205–18212.
  • Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193–208. doi:10.1007/s00018-017-2595-9
  • 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. doi:10.1101/gad.192351.112
  • Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–125. doi:10.1016/j.ceb.2014.05.004
  • Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: from biogenesis and secretion to biological function. Immunol Lett. 2006;107(2):102–108. doi:10.1016/j.imlet.2006.09.005
  • Mayor S, Pagano RE. Pathways of clathrin-independent endocytosis. Nat Rev Mol Cell Biol. 2007;8(8):603–612. doi:10.1038/nrm2216
  • Huotari J, Helenius A. Endosome maturation. EMBO j. 2011;30(17):3481–3500. doi:10.1038/emboj.2011.286
  • Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19(2):43–51. doi:10.1016/j.tcb.2008.11.003
  • Fader CM, Sánchez DG, Mestre MB, Colombo MI. TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways. Biochim Biophys Acta. 2009;1793(12):1901–1916. doi:10.1016/j.bbamcr.2009.09.011
  • Luzio JP, Gray SR, Bright NA. Endosome-lysosome fusion. Biochem Soc Trans. 2010;38(6):1413–1416. doi:10.1042/bst0381413
  • Liu C, Su C. Design strategies and application progress of therapeutic exosomes. Theranostics. 2019;9(4):1015–1028. doi:10.7150/thno.30853
  • Tai YL, Chen KC, Hsieh JT, Shen TL. Exosomes in cancer development and clinical applications. Cancer Sci. 2018;109(8):2364–2374. doi:10.1111/cas.13697
  • Gurunathan S, Chapman-Shimshoni D, Trajkovic S, Gerst JE. Yeast exocytic v-SNAREs confer endocytosis. Mol Biol Cell. 2000;11(10):3629–3643. doi:10.1091/mbc.11.10.3629
  • Gurunathan S, David D, Gerst JE. Dynamin and clathrin are required for the biogenesis of a distinct class of secretory vesicles in yeast. EMBO j. 2002;21(4):602–614. doi:10.1093/emboj/21.4.602
  • Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–1247. doi:10.1126/science.1153124
  • Morvan J, Rinaldi B, Friant S. Pkh1/2-dependent phosphorylation of Vps27 regulates ESCRT-I recruitment to endosomes. Mol Biol Cell. 2012;23(20):4054–4064. doi:10.1091/mbc.E12-01-0001
  • Adell MA, Vogel GF, Pakdel M, et al. Coordinated binding of Vps4 to ESCRT-III drives membrane neck constriction during MVB vesicle formation. J Cell Biol. 2014;205(1):33–49. doi:10.1083/jcb.201310114
  • Romancino DP, Paterniti G, Campos Y, et al. Identification and characterization of the nano-sized vesicles released by muscle cells. FEBS Lett. 2013;587(9):1379–1384. doi:10.1016/j.febslet.2013.03.012
  • McGough IJ, Vincent JP. Exosomes in developmental signalling. Development. 2016;143(14):2482–2493. doi:10.1242/dev.126516
  • Baietti MF, Zhang Z, Mortier E, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14(7):677–685. doi:10.1038/ncb2502
  • Theos AC, Berson JF, Theos SC, et al. Dual loss of ER export and endocytic signals with altered melanosome morphology in the silver mutation of Pmel17. Mol Biol Cell. 2006;17(8):3598–3612. doi:10.1091/mbc.e06-01-0081
  • Kajimoto T, Okada T, Miya S, Zhang L, Nakamura S. Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nat Commun. 2013;4:2712. doi:10.1038/ncomms3712
  • Yu X, Harris SL, Levine AJ. The regulation of exosome secretion: a novel function of the p53 protein. Cancer Res. 2006;66(9):4795–4801. doi:10.1158/0008-5472.Can-05-4579
  • Wang Y, Balaji V, Kaniyappan S, et al. The release and trans-synaptic transmission of Tau via exosomes. Mol Neurodegener. 2017;12(1):5. doi:10.1186/s13024-016-0143-y
  • Marsh AA, Finger EC, Mitchell DG, et al. Reduced amygdala response to fearful expressions in children and adolescents with callous-unemotional traits and disruptive behavior disorders. Am J Psychiatry. 2008;165(6):712–720. doi:10.1176/appi.ajp.2007.07071145
  • Kobayashi H, Tanaka N, Asao H, et al. Hrs, a mammalian master molecule in vesicular transport and protein sorting, suppresses the degradation of ESCRT proteins signal transducing adaptor molecule 1 and 2. J Biol Chem. 2005;280(11):10468–10477. doi:10.1074/jbc.M409969200
  • van Niel G, Porto-Carreiro I, Simoes S, Raposo G. Exosomes: a common pathway for a specialized function. J Biochem. 2006;140(1):13–21. doi:10.1093/jb/mvj128
  • Ren X, Hurley JH. VHS domains of ESCRT-0 cooperate in high-avidity binding to polyubiquitinated cargo. EMBO j. 2010;29(6):1045–1054. doi:10.1038/emboj.2010.6
  • Wollert T, Wunder C, Lippincott-Schwartz J, Hurley JH. Membrane scission by the ESCRT-III complex. Nature. 2009;458(7235):172–177. doi:10.1038/nature07836
  • Yeates EF, Tesco G. The endosome-associated deubiquitinating enzyme USP8 regulates BACE1 enzyme ubiquitination and degradation. J Biol Chem. 2016;291(30):15753–15766. doi:10.1074/jbc.M116.718023
  • Laulagnier K, Motta C, Hamdi S, et al. Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380(Pt 1):161–171. doi:10.1042/bj20031594
  • 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. doi:10.1038/ncomms4477
  • Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009;10(8):513–525. doi:10.1038/nrm2728
  • Tauro BJ, Mathias RA, Greening DW, et al. Oncogenic H-ras reprograms Madin-Darby canine kidney (MDCK) cell-derived exosomal proteins following epithelial-mesenchymal transition. Mol Cell Proteomics. 2013;12(8):2148–2159. doi:10.1074/mcp.M112.027086
  • Savina A, Vidal M, Colombo MI. The exosome pathway in K562 cells is regulated by Rab11. J Cell Sci. 2002;115(Pt 12):2505–2515.
  • Savina A, Fader CM, Damiani MT, Colombo MI. Rab11 promotes docking and fusion of multivesicular bodies in a calcium-dependent manner. Traffic. 2005;6(2):131–143. doi:10.1111/j.1600-0854.2004.00257.x
  • Bobrie A, Krumeich S, Reyal F, et al. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012;72(19):4920–4930. doi:10.1158/0008-5472.Can-12-0925
  • Zomer A, Vendrig T, Hopmans ES, van Eijndhoven M, Middeldorp JM, Pegtel DM. Exosomes: fit to deliver small RNA. Commun Integr Biol. 2010;3(5):447–450. doi:10.4161/cib.3.5.12339
  • Choi DS, Yang JS, Choi EJ, et al. The protein interaction network of extracellular vesicles derived from human colorectal cancer cells. J Proteome Res. 2012;11(2):1144–1151. doi:10.1021/pr200842h
  • Li J, Lee Y, Johansson HJ, et al. Serum-free culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles. J Extracell Vesicles. 2015;4:26883. doi:10.3402/jev.v4.26883
  • Setti M, Osti D, Richichi C, et al. Extracellular vesicle-mediated transfer of CLIC1 protein is a novel mechanism for the regulation of glioblastoma growth. Oncotarget. 2015;6(31):31413–31427. doi:10.18632/oncotarget.5105
  • Yang Q, Nanayakkara GK, Drummer C, et al. Low-intensity ultrasound-induced anti-inflammatory effects are mediated by several new mechanisms including gene induction, immunosuppressor cell promotion, and enhancement of exosome biogenesis and docking. Front Physiol. 2017;8:818. doi:10.3389/fphys.2017.00818
  • Tang X, Chang C, Guo J, et al. Tumour-secreted Hsp90α on external surface of exosomes mediates tumour - stromal cell communication via autocrine and paracrine mechanisms. Sci Rep. 2019;9(1):15108. doi:10.1038/s41598-019-51704-w
  • Giordano C, Gelsomino L, Barone I, et al. Leptin modulates exosome biogenesis in breast cancer cells: an additional mechanism in cell-to-cell communication. J Clin Med. 2019;8(7):1027. doi:10.3390/jcm8071027
  • Hitomi K, Okada R, Loo TM, Miyata K, Nakamura AJ, Takahashi A. DNA damage regulates senescence-associated extracellular vesicle release via the ceramide pathway to prevent excessive inflammatory responses. Int J Mol Sci. 2020;21(10):3720. doi:10.3390/ijms21103720
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi:10.1016/j.cell.2011.02.013
  • Roma-Rodrigues C, Raposo LR, Cabral R, Paradinha F, Baptista PV, Fernandes AR. Tumor microenvironment modulation via gold nanoparticles targeting malicious exosomes: implications for cancer diagnostics and therapy. Int J Mol Sci. 2017;18(1):162. doi:10.3390/ijms18010162
  • Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomedicine. 2021;16:1281–1312. doi:10.2147/ijn.S291956
  • Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19. doi:10.1186/s13578-019-0282-2
  • Tricarico C, Clancy J, D’Souza-Schorey C. Biology and biogenesis of shed microvesicles. Small GTPases. 2017;8(4):220–232. doi:10.1080/21541248.2016.1215283
  • Clancy JW, Sedgwick A, Rosse C, et al. Regulated delivery of molecular cargo to invasive tumour-derived microvesicles. Nat Commun. 2015;6:6919. doi:10.1038/ncomms7919
  • 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;5:14748. doi:10.1038/srep14748
  • Kozlov MM, Campelo F, Liska N, Chernomordik LV, Marrink SJ, McMahon HT. Mechanisms shaping cell membranes. Curr Opin Cell Biol. 2014;29:53–60. doi:10.1016/j.ceb.2014.03.006
  • McMahon HT, Boucrot E. Membrane curvature at a glance. J Cell Sci. 2015;128(6):1065–1070. doi:10.1242/jcs.114454
  • 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. doi:10.1038/onc.2011.636
  • Gulei D, Irimie AI, Cojocneanu-Petric R, Schultze JL, Berindan-Neagoe I. Exosomes-small players, big sound. Bioconjug Chem. 2018;29(3):635–648. doi:10.1021/acs.bioconjchem.8b00003
  • Christ L, Wenzel EM, Liestøl K, Raiborg C, Campsteijn C, Stenmark H. ALIX and ESCRT-I/II function as parallel ESCRT-III recruiters in cytokinetic abscission. J Cell Biol. 2016;212(5):499–513. doi:10.1083/jcb.201507009
  • Sedgwick AE, D’Souza-Schorey C. The biology of extracellular microvesicles. Traffic. 2018;19(5):319–327. doi:10.1111/tra.12558
  • 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. doi:10.1007/s12307-010-0049-0
  • Monteith GR, Prevarskaya N, Roberts-Thomson SJ. The calcium-cancer signalling nexus. Nat Rev Cancer. 2017;17(6):367–380. doi:10.1038/nrc.2017.18
  • 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. doi:10.1021/bi9805238
  • 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. doi:10.1210/jc.2012-2098
  • 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. doi:10.1182/blood-2004-03-1095
  • Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17(3):160–172. doi:10.1038/nrn.2015.29
  • 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 U S A. 2014;111(31):E3234–3242. doi:10.1073/pnas.1410041111
  • 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. doi:10.1016/j.ajpath.2012.07.030
  • 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. doi:10.1158/0008-5472.Can-08-3860
  • Stachowiak JC, Schmid EM, Ryan CJ, et al. Membrane bending by protein-protein crowding. Nat Cell Biol. 2012;14(9):944–949. doi:10.1038/ncb2561
  • Yang JS, Gad H, Lee SY, et al. A role for phosphatidic acid in COPI vesicle fission yields insights into Golgi maintenance. Nat Cell Biol. 2008;10(10):1146–1153. doi:10.1038/ncb1774
  • McConnell RE, Higginbotham JN, Shifrin DA Jr, Tabb DL, Coffey RJ, Tyska MJ. The enterocyte microvillus is a vesicle-generating organelle. J Cell Biol. 2009;185(7):1285–1298. doi:10.1083/jcb.200902147
  • Rilla K, Pasonen-Seppänen S, Deen AJ, et al. Hyaluronan production enhances shedding of plasma membrane-derived microvesicles. Exp Cell Res. 2013;319(13):2006–2018. doi:10.1016/j.yexcr.2013.05.021
  • Choi DS, Lee JM, Park GW, et al. Proteomic analysis of microvesicles derived from human colorectal cancer cells. J Proteome Res. 2007;6(12):4646–4655. doi:10.1021/pr070192y
  • Xu Y, Zhang Y, Wang L, et al. miR-200a targets Gelsolin: a novel mechanism regulating secretion of microvesicles in hepatocellular carcinoma cells. Oncol Rep. 2017;37(5):2711–2719. doi:10.3892/or.2017.5506
  • Taylor J, Azimi I, Monteith G, Bebawy M. Ca(2+) mediates extracellular vesicle biogenesis through alternate pathways in malignancy. J Extracell Vesicles. 2020;9(1):1734326. doi:10.1080/20013078.2020.1734326
  • Akers JC, Gonda D, Kim R, Carter BS, Chen CC. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol. 2013;113(1):1–11. doi:10.1007/s11060-013-1084-8
  • Caruso S, Poon IKH. Apoptotic cell-derived extracellular vesicles: more than just debris. Front Immunol. 2018;9:1486. doi:10.3389/fimmu.2018.01486
  • Brock CK, Wallin ST, Ruiz OE, et al. Stem cell proliferation is induced by apoptotic bodies from dying cells during epithelial tissue maintenance. Nat Commun. 2019;10(1):1044. doi:10.1038/s41467-019-09010-6
  • Collino F, Bruno S, Incarnato D, et al. AKI recovery induced by mesenchymal stromal cell-derived extracellular vesicles carrying microRNAs. J Am Soc Nephrol. 2015;26(10):2349–2360. doi:10.1681/asn.2014070710
  • Depraetere V. “Eat me” signals of apoptotic bodies. Nat Cell Biol. 2000;2(6):E104. doi:10.1038/35014098
  • Gardai SJ, McPhillips KA, Frasch SC, et al. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123(2):321–334. doi:10.1016/j.cell.2005.08.032
  • Orlando KA, Stone NL, Pittman RN. Rho kinase regulates fragmentation and phagocytosis of apoptotic cells. Exp Cell Res. 2006;312(1):5–15. doi:10.1016/j.yexcr.2005.09.012
  • Atkin-Smith GK, Tixeira R, Paone S, et al. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat Commun. 2015;6:7439. doi:10.1038/ncomms8439
  • Núñez R, Sancho-Martínez SM, Novoa JM, López-Hernández FJ. Apoptotic volume decrease as a geometric determinant for cell dismantling into apoptotic bodies. Cell Death Differ. 2010;17(11):1665–1671. doi:10.1038/cdd.2010.96
  • Maeno E, Tsubata T, Okada Y. Apoptotic volume decrease (AVD) is independent of mitochondrial dysfunction and initiator caspase activation. Cells. 2012;1(4):1156–1167. doi:10.3390/cells1041156
  • Ernest NJ, Habela CW, Sontheimer H. Cytoplasmic condensation is both necessary and sufficient to induce apoptotic cell death. J Cell Sci. 2008;121(Pt 3):290–297. doi:10.1242/jcs.017343
  • Crescitelli R, Lässer C, Szabó TG, et al. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013;2(1):20677. doi:10.3402/jev.v2i0.20677
  • Gregory CD, Dransfield I. Apoptotic tumor cell-derived extracellular vesicles as important regulators of the onco-regenerative niche. Front Immunol. 2018;9:1111. doi:10.3389/fimmu.2018.01111
  • Hauser P, Wang S, Didenko VV. Apoptotic bodies: selective detection in extracellular vesicles. Methods Mol Biol. 2017;1554:193–200. doi:10.1007/978-1-4939-6759-9_12
  • Lunavat TR, Cheng L, Kim DK, et al. Small RNA deep sequencing discriminates subsets of extracellular vesicles released by melanoma cells–evidence of unique microRNA cargos. RNA Biol. 2015;12(8):810–823. doi:10.1080/15476286.2015.1056975
  • Meldolesi J. Exosomes and ectosomes in intercellular communication. Curr Biol. 2018;28(8):R435–r444. doi:10.1016/j.cub.2018.01.059
  • Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–289. doi:10.1146/annurev-cellbio-101512-122326
  • Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25(6):364–372. doi:10.1016/j.tcb.2015.01.004
  • Palmisano G, Jensen SS, Le Bihan MC, et al. Characterization of membrane-shed microvesicles from cytokine-stimulated β-cells using proteomics strategies. Mol Cell Proteomics. 2012;11(8):230–243. doi:10.1074/mcp.M111.012732
  • Dinkla S, van Cranenbroek B, van der Heijden WA, et al. Platelet microparticles inhibit IL-17 production by regulatory T cells through P-selectin. Blood. 2016;127(16):1976–1986. doi:10.1182/blood-2015-04-640300
  • 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. doi:10.1038/ncb1725
  • Meehan B, Rak J, Di Vizio D. Oncosomes - large and small: what are they, where they came from? J Extracell Vesicles. 2016;5:33109. doi:10.3402/jev.v5.33109
  • Clancy JW, Zhang Y, Sheehan C, D’Souza-Schorey C. An ARF6-Exportin-5 axis delivers pre-miRNA cargo to tumour microvesicles. Nat Cell Biol. 2019;21(7):856–866. doi:10.1038/s41556-019-0345-y
  • Pasquet JM, Dachary-Prigent J, Nurden AT. Calcium influx is a determining factor of calpain activation and microparticle formation in platelets. Eur J Biochem. 1996;239(3):647–654. doi:10.1111/j.1432-1033.1996.0647u.x
  • 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. doi:10.18632/oncotarget.3598
  • Bertolini I, Terrasi A, Martelli C, et al. A GBM-like V-ATPase signature directs cell-cell tumor signaling and reprogramming via large oncosomes. EBioMedicine. 2019;41:225–235. doi:10.1016/j.ebiom.2019.01.051
  • Morello M, Minciacchi VR, de Candia P, et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle. 2013;12(22):3526–3536. doi:10.4161/cc.26539
  • 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. doi:10.3390/ijms17020175
  • Willms E, Cabañas C, Mäger I, Wood MJA, Vader P. Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in cancer progression. Front Immunol. 2018;9:738. doi:10.3389/fimmu.2018.00738
  • Atay S, Wilkey DW, Milhem M, Merchant M, Godwin AK. Insights into the proteome of gastrointestinal stromal tumors-derived exosomes reveals new potential diagnostic biomarkers. Mol Cell Proteomics. 2018;17(3):495–515. doi:10.1074/mcp.RA117.000267
  • Ciardiello C, Leone A, Lanuti P, et al. Large oncosomes overexpressing integrin alpha-V promote prostate cancer adhesion and invasion via AKT activation. J Exp Clin Cancer Res. 2019;38(1):317. doi:10.1186/s13046-019-1317-6
  • 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. doi:10.1074/jbc.M109.041152
  • Montecalvo A, Larregina AT, Shufesky WJ, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756–766. doi:10.1182/blood-2011-02-338004
  • Heusermann W, Hean J, Trojer D, et al. Exosomes surf on filopodia to enter cells at endocytic hot spots, traffic within endosomes, and are targeted to the ER. J Cell Biol. 2016;213(2):173–184. doi:10.1083/jcb.201506084
  • Yao Z, Qiao Y, Li X, et al. Exosomes exploit the virus entry machinery and pathway to transmit alpha interferon-induced antiviral activity. J Virol. 2018;92(24). doi:10.1128/jvi.01578-18
  • Théry C, Regnault A, Garin J, et al. Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol. 1999;147(3):599–610. doi:10.1083/jcb.147.3.599
  • Zaborowski MP, Balaj L, Breakefield XO, Lai CP. Extracellular vesicles: composition, biological relevance, and methods of study. Bioscience. 2015;65(8):783–797. doi:10.1093/biosci/biv084
  • Kalra H, Drummen GP, Mathivanan S. Focus on extracellular vesicles: introducing the next small big thing. Int J Mol Sci. 2016;17(2):170. doi:10.3390/ijms17020170
  • Ragusa M, Barbagallo C, Cirnigliaro M, et al. Asymmetric RNA distribution among cells and their secreted exosomes: biomedical meaning and considerations on diagnostic applications. Front Mol Biosci. 2017;4:66. doi:10.3389/fmolb.2017.00066
  • Shimoda M, Khokha R. Metalloproteinases in extracellular vesicles. Biochim Biophys Acta Mol Cell Res. 2017;1864(11):1989–2000. doi:10.1016/j.bbamcr.2017.05.027
  • Batista BS, Eng WS, Pilobello KT, Hendricks-Muñoz KD, Mahal LK. Identification of a conserved glycan signature for microvesicles. J Proteome Res. 2011;10(10):4624–4633. doi:10.1021/pr200434y
  • Janas T, Janas MM, Sapoń K, Janas T. Mechanisms of RNA loading into exosomes. FEBS Lett. 2015;589(13):1391–1398. doi:10.1016/j.febslet.2015.04.036
  • Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol. 2009;11(9):1143–1149. doi:10.1038/ncb1929
  • Katzmann DJ, Babst M, Emr SD. Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I. Cell. 2001;106(2):145–155. doi:10.1016/s0092-8674(01)00434-2
  • Gillooly DJ, Raiborg C, Stenmark H. Phosphatidylinositol 3-phosphate is found in microdomains of early endosomes. Histochem Cell Biol. 2003;120(6):445–453. doi:10.1007/s00418-003-0591-7
  • Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Sánchez-Madrid F, Mittelbrunn M. Sorting it out: regulation of exosome loading. Semin Cancer Biol. 2014;28:3–13. doi:10.1016/j.semcancer.2014.04.009
  • Norris A, Tammineni P, Wang S, et al. SNX-1 and RME-8 oppose the assembly of HGRS-1/ESCRT-0 degradative microdomains on endosomes. Proc Natl Acad Sci U S A. 2017;114(3):E307–e316. doi:10.1073/pnas.1612730114
  • Teo H, Perisic O, González B, Williams RL. ESCRT-II, an endosome-associated complex required for protein sorting: crystal structure and interactions with ESCRT-III and membranes. Dev Cell. 2004;7(4):559–569. doi:10.1016/j.devcel.2004.09.003
  • Frankel EB, Audhya A. ESCRT-dependent cargo sorting at multivesicular endosomes. Semin Cell Dev Biol. 2018;74:4–10. doi:10.1016/j.semcdb.2017.08.020
  • Agromayor M, Martin-Serrano J. Interaction of AMSH with ESCRT-III and deubiquitination of endosomal cargo. J Biol Chem. 2006;281(32):23083–23091. doi:10.1074/jbc.M513803200
  • Sun S, Zhou X, Corvera J, Gallick GE, Lin SH, Kuang J. ALG-2 activates the MVB sorting function of ALIX through relieving its intramolecular interaction. Cell Discov. 2015;1:15018. doi:10.1038/celldisc.2015.18
  • Dores MR, Chen B, Lin H, et al. ALIX binds a YPX(3)L motif of the GPCR PAR1 and mediates ubiquitin-independent ESCRT-III/MVB sorting. J Cell Biol. 2012;197(3):407–419. doi:10.1083/jcb.201110031
  • Iavello A, Frech VS, Gai C, Deregibus MC, Quesenberry PJ, Camussi G. Role of Alix in miRNA packaging during extracellular vesicle biogenesis. Int J Mol Med. 2016;37(4):958–966. doi:10.3892/ijmm.2016.2488
  • Friand V, David G, Zimmermann P. Syntenin and syndecan in the biogenesis of exosomes. Biol Cell. 2015;107(10):331–341. doi:10.1111/boc.201500010
  • Roucourt B, Meeussen S, Bao J, Zimmermann P, David G. Heparanase activates the syndecan-syntenin-ALIX exosome pathway. Cell Res. 2015;25(4):412–428. doi:10.1038/cr.2015.29
  • Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10(7):925–937. doi:10.1111/j.1600-0854.2009.00920.x
  • Edgar JR, Eden ER, Futter CE. Hrs- and CD63-dependent competing mechanisms make different sized endosomal intraluminal vesicles. Traffic. 2014;15(2):197–211. doi:10.1111/tra.12139
  • Villarroya-Beltri C, Baixauli F, Mittelbrunn M, et al. ISGylation controls exosome secretion by promoting lysosomal degradation of MVB proteins. Nat Commun. 2016;7:13588. doi:10.1038/ncomms13588
  • Ageta H, Ageta-Ishihara N, Hitachi K, et al. UBL3 modification influences protein sorting to small extracellular vesicles. Nat Commun. 2018;9(1):3936. doi:10.1038/s41467-018-06197-y
  • 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 U S A. 2012;109(11):4146–4151. doi:10.1073/pnas.1200448109
  • Boukouris S, Mathivanan S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin Appl. 2015;9(3–4):358–367. doi:10.1002/prca.201400114
  • 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. doi:10.3402/jev.v1i0.18397
  • Schmidt O, Teis D. The ESCRT machinery. Curr Biol. 2012;22(4):R116–120. doi:10.1016/j.cub.2012.01.028
  • Moreno-Gonzalo J, Osoro K, García U, et al. Anthelmintic effect of heather in goats experimentally infected with Trichostrongylus colubriformis. Parasitol Res. 2014;113(2):693–699. doi:10.1007/s00436-013-3697-4
  • Stoorvogel W. Resolving sorting mechanisms into exosomes. Cell Res. 2015;25(5):531–532. doi:10.1038/cr.2015.39
  • Putz U, Howitt J, Lackovic J, et al. Nedd4 family-interacting protein 1 (Ndfip1) is required for the exosomal secretion of Nedd4 family proteins. J Biol Chem. 2008;283(47):32621–32627. doi:10.1074/jbc.M804120200
  • Mackenzie K, Foot NJ, Anand S, et al. Regulation of the divalent metal ion transporter via membrane budding. Cell Discov. 2016;2:16011. doi:10.1038/celldisc.2016.11
  • Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: cargo sorting into exosomes and extracellular vesicles. Biochim Biophys Acta Proteins Proteom. 2019;1867(12):140203. doi:10.1016/j.bbapap.2019.02.005
  • Kim BY, Olzmann JA, Barsh GS, Chin LS, Li L. Spongiform neurodegeneration-associated E3 ligase Mahogunin ubiquitylates TSG101 and regulates endosomal trafficking. Mol Biol Cell. 2007;18(4):1129–1142. doi:10.1091/mbc.e06-09-0787
  • McDonald B, Martin-Serrano J. No strings attached: the ESCRT machinery in viral budding and cytokinesis. J Cell Sci. 2009;122(Pt 13):2167–2177. doi:10.1242/jcs.028308
  • Stringer DK, Piper RC. A single ubiquitin is sufficient for cargo protein entry into MVBs in the absence of ESCRT ubiquitination. J Cell Biol. 2011;192(2):229–242. doi:10.1083/jcb.201008121
  • Moreno-Gonzalo O, Fernandez-Delgado I, Sanchez-Madrid F. Post-translational add-ons mark the path in exosomal protein sorting. Cell Mol Life Sci. 2018;75(1):1–19. doi:10.1007/s00018-017-2690-y
  • Hu W, Liu C, Bi ZY, et al. Comprehensive landscape of extracellular vesicle-derived RNAs in cancer initiation, progression, metastasis and cancer immunology. Mol Cancer. 2020;19(1):102. doi:10.1186/s12943-020-01199-1
  • Koppers-Lalic D, Hackenberg M, Bijnsdorp IV, et al. Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Rep. 2014;8(6):1649–1658. doi:10.1016/j.celrep.2014.08.027
  • Hinger SA, Cha DJ, Franklin JL, et al. Diverse long RNAs are differentially sorted into extracellular vesicles secreted by colorectal cancer cells. Cell Rep. 2018;25(3):715–725.e714. doi:10.1016/j.celrep.2018.09.054
  • Squadrito ML, Baer C, Burdet F, et al. Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Rep. 2014;8(5):1432–1446. doi:10.1016/j.celrep.2014.07.035
  • 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. doi:10.1186/1471-2164-13-357
  • McKenzie AJ, Hoshino D, Hong NH, et al. KRAS-MEK signaling controls Ago2 sorting into exosomes. Cell Rep. 2016;15(5):978–987. doi:10.1016/j.celrep.2016.03.085
  • Wei JX, Lv LH, Wan YL, et al. Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology. 2015;61(4):1284–1294. doi:10.1002/hep.27660
  • Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun. 2013;4:2980. doi:10.1038/ncomms3980
  • Lee H, Li C, Zhang Y, Zhang D, Otterbein LE, Jin Y. Caveolin-1 selectively regulates microRNA sorting into microvesicles after noxious stimuli. J Exp Med. 2019;216(9):2202–2220. doi:10.1084/jem.20182313
  • Santangelo L, Giurato G, Cicchini C, et al. The RNA-binding protein SYNCRIP is a component of the hepatocyte exosomal machinery controlling microRNA sorting. Cell Rep. 2016;17(3):799–808. doi:10.1016/j.celrep.2016.09.031
  • Hagiwara K, Katsuda T, Gailhouste L, Kosaka N, Ochiya T. Commitment of Annexin A2 in recruitment of microRNAs into extracellular vesicles. FEBS Lett. 2015;589(24Pt B):4071–4078. doi:10.1016/j.febslet.2015.11.036
  • Liang J, Zhang X, He S, et al. Sphk2 RNAi nanoparticles suppress tumor growth via downregulating cancer cell derived exosomal microRNA. J Control Release. 2018;286:348–357. doi:10.1016/j.jconrel.2018.07.039
  • Mukherjee K, Ghoshal B, Ghosh S, et al. Reversible HuR-microRNA binding controls extracellular export of miR-122 and augments stress response. EMBO Rep. 2016;17(8):1184–1203. doi:10.15252/embr.201541930
  • Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010;101(10):2087–2092. doi:10.1111/j.1349-7006.2010.01650.x
  • Lu P, Li H, Li N, et al. MEX3C interacts with adaptor-related protein complex 2 and involves in miR-451a exosomal sorting. PLoS One. 2017;12(10):e0185992. doi:10.1371/journal.pone.0185992
  • Bolukbasi MF, Mizrak A, Ozdener GB, et al. miR-1289 and “Zipcode”-like sequence enrich mRNAs in microvesicles. Mol Ther Nucleic Acids. 2012;1(2):e10. doi:10.1038/mtna.2011.2
  • Wei Z, Batagov AO, Schinelli S, et al. Coding and noncoding landscape of extracellular RNA released by human glioma stem cells. Nat Commun. 2017;8(1):1145. doi:10.1038/s41467-017-01196-x
  • Russell AE, Sneider A, Witwer KW, et al. Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: an ISEV position paper arising from the ISEV membranes and EVs workshop. J Extracell Vesicles. 2019;8(1):1684862. doi:10.1080/20013078.2019.1684862
  • Prattichizzo F, Matacchione G, Giuliani A, et al. Extracellular vesicle-shuttled miRNAs: a critical appraisal of their potential as nano-diagnostics and nano-therapeutics in type 2 diabetes mellitus and its cardiovascular complications. Theranostics. 2021;11(3):1031–1045. doi:10.7150/thno.51605
  • Zuccato E, Blott EJ, Holt O, et al. Sorting of Fas ligand to secretory lysosomes is regulated by mono-ubiquitylation and phosphorylation. J Cell Sci. 2007;120(Pt 1):191–199. doi:10.1242/jcs.03315
  • Saman S, Kim W, Raya M, et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem. 2012;287(6):3842–3849. doi:10.1074/jbc.M111.277061
  • Valapala M, Thamake SI, Vishwanatha JK. A competitive hexapeptide inhibitor of annexin A2 prevents hypoxia-induced angiogenic events. J Cell Sci. 2011;124(Pt9):1453–1464. doi:10.1242/jcs.079236
  • Delacour D, Greb C, Koch A, et al. Apical sorting by galectin-3-dependent glycoprotein clustering. Traffic. 2007;8(4):379–388. doi:10.1111/j.1600-0854.2007.00539.x
  • Delacour D, Cramm-Behrens CI, Drobecq H, Le Bivic A, Naim HY, Jacob R. Requirement for galectin-3 in apical protein sorting. Curr Biol. 2006;16(4):408–414. doi:10.1016/j.cub.2005.12.046
  • Cloutier N, Tan S, Boudreau LH, et al. The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: the microparticle-associated immune complexes. EMBO Mol Med. 2013;5(2):235–249. doi:10.1002/emmm.201201846
  • Skriner K, Adolph K, Jungblut PR, Burmester GR. Association of citrullinated proteins with synovial exosomes. Arthritis Rheum. 2006;54(12):3809–3814. doi:10.1002/art.22276
  • Shen B, Wu N, Yang JM, Gould SJ. Protein targeting to exosomes/microvesicles by plasma membrane anchors. J Biol Chem. 2011;286(16):14383–14395. doi:10.1074/jbc.M110.208660
  • Surgucheva I, Sharov VS, Surguchov A. γ-Synuclein: seeding of α-synuclein aggregation and transmission between cells. Biochemistry. 2012;51(23):4743–4754. doi:10.1021/bi300478w
  • Sterzenbach U, Putz U, Low LH, Silke J, Tan SS, Howitt J. Engineered exosomes as vehicles for biologically active proteins. Mol Ther. 2017;25(6):1269–1278. doi:10.1016/j.ymthe.2017.03.030
  • Mathivanan S, Lim JW, Tauro BJ, Ji H, Moritz RL, Simpson RJ. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol Cell Proteomics. 2010;9(2):197–208. doi:10.1074/mcp.M900152-MCP200
  • Yuana Y, Sturk A, Nieuwland R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013;27(1):31–39. doi:10.1016/j.blre.2012.12.002
  • Lowry MC, Gallagher WM, O’Driscoll L. The role of exosomes in breast cancer. Clin Chem. 2015;61(12):1457–1465. doi:10.1373/clinchem.2015.240028
  • Im EJ, Lee CH, Moon PG, et al. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat Commun. 2019;10(1):1387. doi:10.1038/s41467-019-09387-4
  • Catalano M, O’Driscoll L. Inhibiting extracellular vesicles formation and release: a review of EV inhibitors. J Extracell Vesicles. 2020;9(1):1703244. doi:10.1080/20013078.2019.1703244
  • Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285(23):17442–17452. doi:10.1074/jbc.M110.107821
  • Wei Y, Li M, Cui S, et al. Shikonin inhibits the proliferation of human breast cancer cells by reducing tumor-derived exosomes. Molecules. 2016;21(6). doi:10.3390/molecules21060777
  • Burnley-Hall N, Willis G, Davis J, Rees DA, James PE. Nitrite-derived nitric oxide reduces hypoxia-inducible factor 1α-mediated extracellular vesicle production by endothelial cells. Nitric Oxide. 2017;63:1–12. doi:10.1016/j.niox.2016.12.005
  • Overmiller AM, Pierluissi JA, Wermuth PJ, et al. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes. FASEB J. 2017;31(8):3412–3424. doi:10.1096/fj.201601138RR
  • 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). doi:10.3390/ijms18051007
  • Fogli S, Neri T, Nuti E, Mattii L, Camodeca C, Rossello A. Matrix metalloproteinase inhibitors prevent the release and proteolytic activity of monocyte/macrophage-derived microparticles. Pharmacol Rep. 2019;71(3):485–490. doi:10.1016/j.pharep.2019.01.013
  • Koumangoye RB, Sakwe AM, Goodwin JS, Patel T, Ochieng J. Detachment of breast tumor cells induces rapid secretion of exosomes which subsequently mediate cellular adhesion and spreading. PLoS One. 2011;6(9):e24234. doi:10.1371/journal.pone.0024234
  • Emam SE, Ando H, Abu Lila AS, et al. A Novel strategy to increase the yield of exosomes (extracellular vesicles) for an expansion of basic research. Biol Pharm Bull. 2018;41(5):733–742. doi:10.1248/bpb.b17-00919
  • King HW, Michael MZ, Gleadle JM. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer. 2012;12:421. doi:10.1186/1471-2407-12-421
  • Hannafon BN, Carpenter KJ, Berry WL, Janknecht R, Dooley WC, Ding WQ. Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer. 2015;14:133. doi:10.1186/s12943-015-0400-7
  • Chao OS, Chang TC, Di Bella MA, et al. The HDAC6 Inhibitor Tubacin induces release of CD133(+) extracellular vesicles from cancer cells. J Cell Biochem. 2017;118(12):4414–4424. doi:10.1002/jcb.26095
  • Zhang C, Xiao X, Chen M, Aldharee H, Chen Y, Long W. Liver kinase B1 restoration promotes exosome secretion and motility of lung cancer cells. Oncol Rep. 2018;39(1):376–382. doi:10.3892/or.2017.6085
  • Nkosi D, Sun L, Duke LC, et al. Epstein-Barr virus LMP1 promotes Syntenin-1- and Hrs-induced extracellular vesicle formation for its own secretion to increase cell proliferation and migration. mBio. 2020;11(3). doi:10.1128/mBio.00589-20
  • Fan SJ, Kroeger B, Marie PP, et al. Glutamine deprivation alters the origin and function of cancer cell exosomes. EMBO j. 2020;39(16):e103009. doi:10.15252/embj.2019103009
  • Montermini L, Meehan B, Garnier D, et al. Inhibition of oncogenic epidermal growth factor receptor kinase triggers release of exosome-like extracellular vesicles and impacts their phosphoprotein and DNA content. J Biol Chem. 2015;290(40):24534–24546. doi:10.1074/jbc.M115.679217
  • Fujiwara T, Eguchi T, Sogawa C, et al. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition. Biochem Biophys Res Commun. 2018;503(3):1267–1272. doi:10.1016/j.bbrc.2018.07.035
  • Clayton A, Turkes A, Navabi H, Mason MD, Tabi Z. Induction of heat shock proteins in B-cell exosomes. J Cell Sci. 2005;118(Pt 16):3631–3638. doi:10.1242/jcs.02494
  • Lancaster GI, Febbraio MA. Exosome-dependent trafficking of HSP70: a novel secretory pathway for cellular stress proteins. J Biol Chem. 2005;280(24):23349–23355. doi:10.1074/jbc.M502017200
  • Taha EA, Ono K, Eguchi T. Roles of extracellular HSPs as biomarkers in immune surveillance and immune evasion. Int J Mol Sci. 2019;20(18). doi:10.3390/ijms20184588
  • Atienzar-Aroca S, Flores-Bellver M, Serrano-Heras G, et al. Oxidative stress in retinal pigment epithelium cells increases exosome secretion and promotes angiogenesis in endothelial cells. J Cell Mol Med. 2016;20(8):1457–1466. doi:10.1111/jcmm.12834
  • Bewicke-Copley F, Mulcahy LA, Jacobs LA, et al. Extracellular vesicles released following heat stress induce bystander effect in unstressed populations. J Extracell Vesicles. 2017;6(1):1340746. doi:10.1080/20013078.2017.1340746
  • Hedlund M, Nagaeva O, Kargl D, Baranov V, Mincheva-Nilsson L. Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS One. 2011;6(2):e16899. doi:10.1371/journal.pone.0016899
  • Collett GP, Redman CW, Sargent IL, Vatish M. Endoplasmic reticulum stress stimulates the release of extracellular vesicles carrying danger-associated molecular pattern (DAMP) molecules. Oncotarget. 2018;9(6):6707–6717. doi:10.18632/oncotarget.24158
  • Sudnitsyna J, Skverchinskaya E, Dobrylko I, Nikitina E, Gambaryan S, Mindukshev I. Microvesicle formation induced by oxidative stress in human erythrocytes. Antioxidants (Basel). 2020;9(10). doi:10.3390/antiox9100929
  • Harmati M, Gyukity-Sebestyen E, Dobra G, et al. Small extracellular vesicles convey the stress-induced adaptive responses of melanoma cells. Sci Rep. 2019;9(1):15329. doi:10.1038/s41598-019-51778-6
  • Zhu L, Zang J, Liu B, et al. Oxidative stress-induced RAC autophagy can improve the HUVEC functions by releasing exosomes. J Cell Physiol. 2020;235(10):7392–7409. doi:10.1002/jcp.29641
  • Gurunathan S, Kang MH, Jeyaraj M, Kim JH. Platinum nanoparticles enhance exosome release in human lung epithelial adenocarcinoma cancer cells (A549): oxidative stress and the ceramide pathway are key players. Int J Nanomedicine. 2021;16:515–538. doi:10.2147/ijn.S291138
  • Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38(6):754–763. doi:10.1038/aps.2017.12
  • Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606–1614. doi:10.1038/mt.2010.105
  • Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383–2390. doi:10.1016/j.biomaterials.2013.11.083
  • Wang J, Zheng Y, Zhao M. Exosome-based cancer therapy: implication for targeting cancer stem cells. Front Pharmacol. 2016;7:533. doi:10.3389/fphar.2016.00533
  • Wang J, Dong Y, Li Y, et al. Designer exosomes for active targeted chemo-photothermal synergistic tumor therapy. Adv Funct Mater. 2018;28(18):1707360. doi:10.1002/adfm.201707360
  • Hong Y, Nam G-H, Koh E, et al. Exosome as a vehicle for delivery of membrane protein therapeutics, PH20, for enhanced tumor penetration and antitumor efficacy. Adv Funct Mater. 2018;28(5):1703074. doi:10.1002/adfm.201703074
  • Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–345. doi:10.1038/nbt.1807
  • El-Andaloussi S, Lee Y, Lakhal-Littleton S, et al. Exosome-mediated delivery of siRNA in vitro and in vivo. Nat Protoc. 2012;7(12):2112–2126. doi:10.1038/nprot.2012.131
  • Bellavia D, Raimondo S, Calabrese G, et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics. 2017;7(5):1333–1345. doi:10.7150/thno.17092
  • Kojima R, Bojar D, Rizzi G, et al. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson’s disease treatment. Nat Commun. 2018;9(1):1305. doi:10.1038/s41467-018-03733-8
  • György B, Sage C, Indzhykulian AA, et al. Rescue of hearing by gene delivery to inner-ear hair cells using exosome-associated AAV. Mol Ther. 2017;25(2):379–391. doi:10.1016/j.ymthe.2016.12.010
  • Barbosa FMC, Dupin TV, Toledo MDS, et al. Extracellular vesicles released by Leishmania (Leishmania) amazonensis promote disease progression and induce the production of different cytokines in macrophages and B-1 Cells. Front Microbiol. 2018;9:3056. doi:10.3389/fmicb.2018.03056
  • Roig J, Saiz ML, Galiano A, et al. Extracellular vesicles from the helminth fasciola hepatica prevent DSS-induced acute ulcerative colitis in a T-lymphocyte independent mode. Front Microbiol. 2018;9:1036. doi:10.3389/fmicb.2018.01036
  • Eichenberger RM, Talukder MH, Field MA, et al. Characterization of Trichuris muris secreted proteins and extracellular vesicles provides new insights into host-parasite communication. J Extracell Vesicles. 2018;7(1):1428004. doi:10.1080/20013078.2018.1428004
  • Yuan D, Zhao Y, Banks WA, et al. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials. 2017;142:1–12. doi:10.1016/j.biomaterials.2017.07.011
  • Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30. doi:10.1016/j.jconrel.2015.03.033
  • Yim N, Ryu SW, Choi K, et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein-protein interaction module. Nat Commun. 2016;7:12277. doi:10.1038/ncomms12277
  • Wolfers J, Lozier A, Raposo G, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7(3):297–303. doi:10.1038/85438
  • Ouyang W, O’Garra A. IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation. Immunity. 2019;50(4):871–891. doi:10.1016/j.immuni.2019.03.020
  • Tang TT, Wang B, Wu M, et al. Extracellular vesicle-encapsulated IL-10 as novel nanotherapeutics against ischemic AKI. Sci Adv. 2020;6(33):eaaz0748. doi:10.1126/sciadv.aaz0748
  • Bruno S, Grange C, Collino F, et al. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 2012;7(3):e33115. doi:10.1371/journal.pone.0033115
  • Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–222. doi:10.1016/j.scr.2009.12.003
  • Tan X, Gong YZ, Wu P, Liao DF, Zheng XL. Mesenchymal stem cell-derived microparticles: a promising therapeutic strategy. Int J Mol Sci. 2014;15(8):14348–14363. doi:10.3390/ijms150814348
  • Yan Y, Jiang W, Tan Y, et al. hucMSC exosome-derived GPX1 is required for the recovery of hepatic oxidant injury. Mol Ther. 2017;25(2):465–479. doi:10.1016/j.ymthe.2016.11.019
  • Mardpour S, Ghanian MH, Sadeghi-Abandansari H, et al. Hydrogel-mediated sustained systemic delivery of mesenchymal stem cell-derived extracellular vesicles improves hepatic regeneration in chronic liver failure. ACS Appl Mater Interfaces. 2019;11(41):37421–37433. doi:10.1021/acsami.9b10126
  • Zhang Y, Jin X, Liang J, et al. Extracellular vesicles derived from ODN-stimulated macrophages transfer and activate Cdc42 in recipient cells and thereby increase cellular permissiveness to EV uptake. Sci Adv. 2019;5(7):eaav1564. doi:10.1126/sciadv.aav1564
  • Usman WM, Pham TC, Kwok YY, et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat Commun. 2018;9(1):2359. doi:10.1038/s41467-018-04791-8
  • 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. doi:10.1038/ncb1800
  • Shao H, Chung J, Lee K, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;6:6999. doi:10.1038/ncomms7999
  • Melo SA, Luecke LB, Kahlert C, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177–182. doi:10.1038/nature14581
  • Park J, Lin HY, Assaker JP, et al. Integrated kidney exosome analysis for the detection of kidney transplant rejection. ACS Nano. 2017;11(11):11041–11046. doi:10.1021/acsnano.7b05083
  • Meng W, He C, Hao Y, Wang L, Li L, Zhu G. Prospects and challenges of extracellular vesicle-based drug delivery system: considering cell source. Drug Deliv. 2020;27(1):585–598. doi:10.1080/10717544.2020.1748758
  • Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira de Almeida L. Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J Control Release. 2017;262:247–258. doi:10.1016/j.jconrel.2017.07.001
  • Das CK, Jena BC, Banerjee I, et al. Exosome as a novel shuttle for delivery of therapeutics across biological barriers. Mol Pharm. 2019;16(1):24–40. doi:10.1021/acs.molpharmaceut.8b00901
  • Zhang K, Zhao X, Chen X, et al. Enhanced therapeutic effects of mesenchymal stem cell-derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Appl Mater Interfaces. 2018;10(36):30081–30091. doi:10.1021/acsami.8b08449
  • Tian T, Zhang HX, He CP, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials. 2018;150:137–149. doi:10.1016/j.biomaterials.2017.10.012
  • Yamashita T, Takahashi Y, Takakura Y. Possibility of exosome-based therapeutics and challenges in production of exosomes eligible for therapeutic application. Biol Pharm Bull. 2018;41(6):835–842. doi:10.1248/bpb.b18-00133
  • Rosell A, Havervall S, von Meijenfeldt F, et al. Patients with COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality. Arterioscler Thromb Vasc Biol. 2020;41(2):Atvbaha120315547. doi:10.1161/atvbaha.120.315547