Cancer-Derived Extracellular Vesicle-Associated MicroRNAs in Intercellular Communication: One Cell’s Trash Is Another Cell’s Treasure
Abstract
:1. Introduction
2. miRNA Biogenesis
3. Physiopathology of Extracellular Vesicles (EVs)
3.1. Classification of EVs
3.2. EV Biogenesis
3.3. miRNA Sorting in EVs
4. Extracellular Vesicles (EVs)-Associated miRNAs as Modulators of Tumor Microenvironment (TME)
4.1. EV-Associated miRNAs in the Modulation of Vascular Permeability
4.2. Extracellular Matrix Remodeling via miRNA-Containing EVs
4.3. miRNAs in EVs Drive Immune Modulation during Metastasis Formation
4.4. Dormancy in the Metastatic Niche Is Induced by miRNAs in Cancer-Associated EVs
5. Translational Potential of Extracellular Vesicles (EVs)-Associated miRNAs
5.1. miRNA-Containing Extracellular Vesicles as Cancer Biomarkers in the Clinical Setting
5.2. Therapeutic Applications of EV-Associated miRNAs
6. Final Considerations
Supplementary Materials
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
- Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
- Lee, R.C.; Ambros, V. An extensive class of small RNAs in Caenorhabditis elegans. Science. 2001, 294, 862–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lau, N.C.; Lim, L.P.; Weinstein, E.G.; Bartel, D.P. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 2001, 294, 858–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Treiber, T.; Treiber, N.; Meister, G. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat. Rev. Mol. Cell Biol. 2019, 20, 5–20. [Google Scholar] [CrossRef] [PubMed]
- Kozomara, A.; Griffiths-Jones, S. MiRBase: Annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014, 42, D68–D73. [Google Scholar] [CrossRef] [Green Version]
- Friedman, R.C.; Farh, K.K.H.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef] [Green Version]
- Croce, C.M. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009, 10, 704–714. [Google Scholar] [CrossRef]
- Bracken, C.P.; Scott, H.S.; Goodall, G.J. A network-biology perspective of microRNA function and dysfunction in cancer. Nat. Rev. Genet. 2016, 17, 719–732. [Google Scholar] [CrossRef]
- Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.; Kim, M.; Han, J.; Yeom, K.H.; Lee, S.; Baek, S.H.; Kim, V.N. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004, 23, 4051–4060. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef] [PubMed]
- Han, J.; Lee, Y.; Yeom, K.H.; Kim, Y.K.; Jin, H.; Kim, V.N. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004, 18, 3016–3027. [Google Scholar] [CrossRef] [Green Version]
- Lund, E.; Güttinger, S.; Calado, A.; Dahlberg, J.E.; Kutay, U. Nuclear Export of MicroRNA Precursors. Science 2004, 303, 95–98. [Google Scholar] [CrossRef] [Green Version]
- Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
- Meijer, H.A.; Smith, E.M.; Bushell, M. Regulation of miRNA strand selection: Follow the leader? Biochem. Soc. Trans. 2014, 42, 1135–1140. [Google Scholar] [CrossRef]
- Meijer, H.A.; Kong, Y.W.; Lu, W.T.; Wilczynska, A.; Spriggs, R.V.; Robinson, S.W.; Godfrey, J.D.; Willis, A.E.; Bushell, M. Translational repression and eIF4A2 activity are critical for microRNA-mediated gene regulation. Science 2013, 340, 82–85. [Google Scholar] [CrossRef]
- Guo, H.; Ingolia, N.T.; Weissman, J.S.; Bartel, D.P. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 2010, 466, 835–840. [Google Scholar] [CrossRef] [Green Version]
- Gebert, L.F.R.; MacRae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef]
- Tkach, M.; Théry, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lane, R.E.; Korbie, D.; Hill, M.M.; Trau, M. Extracellular vesicles as circulating cancer biomarkers: opportunities and challenges. Clin. Transl. Med. 2018, 7, 14. [Google Scholar] [CrossRef] [PubMed]
- Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [PubMed]
- Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Ann. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
- Hirsch, J.G.; Fedorko, M.E.; Cohn, Z.A. Vesicle fusion and formation at the surface of pinocytic vacuoles in macrophages. J. Cell Biol. 1968, 38, 629–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. USA 2012, 109, 2110–2116. [Google Scholar] [CrossRef] [Green Version]
- Exosome RNA Administrator. Available online: https://www.exosome-rna.com/divide-conquer-developing-new-class-of-cancer-therapeutics-to-disrupt-cell-to-cell-communication/ (accessed on 26 September 2019).
- Zappulli, V.; Pagh Friis, K.; Fitzpatrick, Z.; Maguire, C.A.; Breakefield, X.O. Extracellular vesicles and intercellular communication within the nervous system. J. Clin. Investig. 2016, 126, 1198–1207. [Google Scholar] [CrossRef] [Green Version]
- Pitt, J.M.; Kroemer, G.; Zitvogel, L. Extracellular vesicles: Masters of intercellular communication and potential clinical interventions. J. Clin. Investig. 2016, 126, 1139–1143. [Google Scholar] [CrossRef] [Green Version]
- Lobb, R.J.; Lima, L.G.; Möller, A. Exosomes: Key mediators of metastasis and pre-metastatic niche formation. Semin. Cell Dev. Biol. 2017, 67, 3–10. [Google Scholar] [CrossRef] [Green Version]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef] [PubMed]
- Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2019, 8, 516–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef]
- Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, 968–977. [Google Scholar] [CrossRef] [Green Version]
- Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009, 19, 43–51. [Google Scholar] [CrossRef]
- Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [Green Version]
- Muralidharan-Chari, V.; Clancy, J.W.; Sedgwick, A.; D’Souza-Schorey, C. Microvesicles: Mediators of extracellular communication during cancer progression. J. Cell Sci. 2010, 123, 1603–1611. [Google Scholar] [CrossRef] [Green Version]
- Melentijevic, I.; Toth, M.L.; Arnold, M.L.; Guasp, R.J.; Harinath, G.; Nguyen, K.C.; Taub, D.; Parker, J.A.; Neri, C.; Gabel, C.V.; et al. C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature 2017, 542, 367–371. [Google Scholar] [CrossRef] [Green Version]
- Ma, L.; Li, Y.; Peng, J.; Wu, D.; Zhao, X.; Cui, Y.; Chen, L.; Yan, X.; Du, Y.; Yu, L. Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res. 2015, 25, 24–38. [Google Scholar] [CrossRef] [Green Version]
- Minciacchi, V.R.; Spinelli, C.; Reis-Sobreiro, M.; Cavallini, L.; You, S.; Zandian, M.; Li, X.; Mishra, R.; Chiarugi, P.; Adam, R.M.; et al. MYC mediates large oncosome-induced fibroblast reprogramming in prostate cancer. Cancer Res. 2017, 77, 2306–2317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zijlstra, A.; Di Vizio, D. Size matters in nanoscale communication. Nat. Cell Biol. 2018, 20, 228–230. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gould, S.J.; Raposo, G. As we wait: Coping with an imperfect nomenclature for extracellular vesicles. J. Extracell. Vesicles 2013. [Google Scholar] [CrossRef] [PubMed]
- Jackson, C.E.; Scruggs, B.S.; Schaffer, J.E.; Hanson, P.I. Effects of Inhibiting VPS4 Support a General Role for ESCRTs in Extracellular Vesicle Biogenesis. Biophys. J. 2017, 113, 1342–1352. [Google Scholar] [CrossRef] [Green Version]
- Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
- Stein, J.M.; Luzio, J.P. 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, 381–386. [Google Scholar] [CrossRef]
- Schmidt, O.; Teis, D. The ESCRT machinery. Curr. Biol. 2012, 22, 116–120. [Google Scholar] [CrossRef] [Green Version]
- Möbius, W.; Ohno-Iwashita, Y.; Van Donselaar, E.G.; Oorschot, V.M.J.; Shimada, Y.; Fujimoto, T.; Heijnen, H.F.G.; Geuze, H.J.; Slot, J.W. Exosomes as mediators of platinum resistance in ovarian cancer. J. Histochem. Cytochem. 2002, 8, 11917–11936. [Google Scholar]
- Hsu, C.; Morohashi, Y.; Yoshimura, S.I.; Manrique-Hoyos, N.; Jung, S.Y.; Lauterbach, M.A.; Bakhti, M.; Grønborg, M.; Möbius, W.; Rhee, J.S.; et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 2010, 189, 223–232. [Google Scholar] [CrossRef]
- Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoshino, D.; Kirkbride, K.C.; Costello, K.; Clark, E.S.; Sinha, S.; Grega-Larson, N.; Tyska, M.J.; Weaver, A.M. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013, 5, 1159–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, W.J.; Lev, S. Tethering the assembly of SNARE complexes. Trends Cell Biol. 2014, 24, 35–43. [Google Scholar] [CrossRef] [PubMed]
- Südhof, T.C.; Rothman, J.E. Membrane fusion: Grappling with SNARE and SM proteins. Science 2009, 323, 474–477. [Google Scholar]
- Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Verweij, F.J.; Bebelman, M.P.; Jimenez, C.R.; Garcia-Vallejo, J.J.; Janssen, H.; Neefjes, J.; Knol, J.C.; de Goeij-de Haas, R.; Piersma, S.R.; Baglio, S.R.; et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J. Cell Biol. 2018, 217, 1129–1142. [Google Scholar] [CrossRef]
- Cocucci, E.; Racchetti, G.; Podini, P.; Meldolesi, J. Enlargeosome traffic: Exocytosis triggered by various signals is followed by endocytosis, membrane shedding or both. Traffic 2007, 8, 742–757. [Google Scholar] [CrossRef]
- Kobayashi, T.; Okamoto, H.; Yamada, J.I.; Setaka, M.; Kwan, T. Vesiculation of platelet plasma membranes. Dilauroylglycerophosphocholine-induced shedding of a platelet plasma membrane fraction enriched in acetylcholinesterase activity. BBA - Biomembr. 1984, 778, 210–218. [Google Scholar] [CrossRef]
- Tricarico, C.; Clancy, J.; D’Souza-Schorey, C. Biology and biogenesis of shed microvesicles. Small GTPases 2017, 8, 220–232. [Google Scholar] [CrossRef] [Green Version]
- Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
- Hinger, S.A.; Cha, D.J.; Franklin, J.L.; Higginbotham, J.N.; Dou, Y.; Ping, J.; Shu, L.; Prasad, N.; Levy, S.; Zhang, B.; et al. Diverse Long RNAs Are Differentially Sorted into Extracellular Vesicles Secreted by Colorectal Cancer Cells. Cell Rep. 2018, 25, 715–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Li, C.; Zhang, L.; Wang, X. Extracellular vesicles as carriers of non-coding RNAs in liver diseases. Front. Pharmacol. 2018, 9, 415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sork, H.; Corso, G.; Krjutskov, K.; Johansson, H.J.; Nordin, J.Z.; Wiklander, O.P.B.; Lee, Y.X.F.; Westholm, J.O.; Lehtiö, J.; Wood, M.J.A.; et al. Heterogeneity and interplay of the extracellular vesicle small RNA transcriptome and proteome. Sci. Rep. 2018, 8, 10813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Preußer, C.; Hung, L.H.; Schneider, T.; Schreiner, S.; Hardt, M.; Moebus, A.; Santoso, S.; Bindereif, A. Selective release of circRNAs in platelet-derived extracellular vesicles. J. Extracell. Vesicles 2018, 7, 1424473. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhang, Q.; Zhang, J.; Li, C.; Miao, Y.R.; Lei, Q.; Li, Q.; Guo, A.Y. EVmiRNA: A database of miRNA profiling in extracellular vesicles. Nucleic Acids Res. 2019, 47, 89–93. [Google Scholar] [CrossRef] [Green Version]
- Turchinovich, A.; Drapkina, O.; Tonevitsky, A. Transcriptome of extracellular vesicles: State-of-the-art. Front. Immunol. 2019, 10, 202. [Google Scholar] [CrossRef] [Green Version]
- Guduric-Fuchs, J.; O’Connor, A.; Camp, B.; O’Neill, C.L.; Medina, R.J.; Simpson, D.A. Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genom. 2012, 13, 357. [Google Scholar] [CrossRef] [Green Version]
- Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
- Mori, M.A.; Raghavan, P.; Thomou, T.; Boucher, J.; Robida-Stubbs, S.; MacOtela, Y.; Russell, S.J.; Kirkland, J.L.; Blackwell, T.K.; Kahn, C.R. Role of microRNA processing in adipose tissue in stress defense and longevity. Cell Metab. 2012, 16, 336–347. [Google Scholar] [CrossRef] [Green Version]
- Fernández-Messina, L.; Gutiérrez-Vázquez, C.; Rivas-García, E.; Sánchez-Madrid, F.; de la Fuente, H. Immunomodulatory role of microRNAs transferred by extracellular vesicles. Biol. Cell 2015, 107, 61–77. [Google Scholar] [CrossRef] [Green Version]
- Forterre, A.; Jalabert, A.; Chikh, K.; Pesenti, S.; Euthine, V.; Granjon, A.; Errazuriz, E.; Lefai, E.; Vidal, H.; Rome, S. Myotube-derived exosomal miRNAs downregulate Sirtuin1 in myoblasts during muscle cell differentiation. Cell Cycle 2014, 13, 78–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mateescu, B.; Kowal, E.J.K.; van Balkom, B.W.M.; Bartel, S.; Bhattacharyya, S.N.; Buzás, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.N.; Das, S.; et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - An ISEV position paper. J. Extracell. Vesicles 2017, 6, 1286095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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, 17442–17452. [Google Scholar] [CrossRef] [Green Version]
- Shurtleff, M.J.; Temoche-Diaz, M.M.; Karfilis, K.V.; Ri, S.; Schekman, R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. Elife 2016, 5, 19276. [Google Scholar] [CrossRef] [PubMed]
- Santangelo, L.; Giurato, G.; Cicchini, C.; Montaldo, C.; Mancone, C.; Tarallo, R.; Battistelli, C.; Alonzi, T.; Weisz, A.; Tripodi, M. The RNA-Binding Protein SYNCRIP Is a Component of the Hepatocyte Exosomal Machinery Controlling MicroRNA Sorting. Cell Rep. 2016, 17, 799–808. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Li, C.; Zhang, Y.; Zhang, D.; Otterbein, L.E.; Jin, Y. Caveolin-1 selectively regulates microRNA sorting into microvesicles after noxious stimuli. J. Exp. Med. 2019, 216, 2202–2220. [Google Scholar] [CrossRef]
- Villarroya-Beltri, C.; Gutiérrez-Vázquez, C.; Sánchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef] [Green Version]
- Temoche-Diaz, M.M.; Shurtleff, M.J.; Nottingham, R.M.; Yao, J.; Fadadu, R.P.; Lambowitz, A.M.; Schekman, R. Distinct mechanisms of microRNA sorting into cancer cell-derived extracellular vesicle subtypes. Elife 2019, 8, 47544. [Google Scholar] [CrossRef]
- Gibbings, D.J.; Ciaudo, C.; Erhardt, M.; Voinnet, O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat. Cell Biol. 2009, 11, 1143–1149. [Google Scholar] [CrossRef]
- McKenzie, A.J.; Hoshino, D.; Hong, N.H.; Cha, D.J.; Franklin, J.L.; Coffey, R.J.; Patton, J.G.; Weaver, A.M. KRAS-MEK Signaling Controls Ago2 Sorting into Exosomes. Cell Rep. 2016, 15, 978–987. [Google Scholar] [CrossRef] [Green Version]
- Cha, D.J.; Franklin, J.L.; Dou, Y.; Liu, Q.; Higginbotham, J.N.; Beckler, M.D.; Weaver, A.M.; Vickers, K.; Prasad, N.; Levy, S.; et al. KRAS-dependent sorting of miRNA to exosomes. Elife 2015, 4, e07197. [Google Scholar] [CrossRef] [PubMed]
- Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teng, Y.; Ren, Y.; Hu, X.; Mu, J.; Samykutty, A.; Zhuang, X.; Deng, Z.; Kumar, A.; Zhang, L.; Merchant, M.L.; et al. MVP-mediated exosomal sorting of miR-193a promotes colon cancer progression. Nat. Commun. 2017, 8, 14448. [Google Scholar] [CrossRef] [PubMed]
- Statello, L.; Maugeri, M.; Garre, E.; Nawaz, M.; Wahlgren, J.; Papadimitriou, A.; Lundqvist, C.; Lindfors, L.; Collén, A.; Sunnerhagen, P.; et al. Identification of RNA-binding proteins in exosomes capable of interacting with different types of RNA: RBP-facilitated transport of RNAs into exosomes. PLoS ONE 2018, 13, e0195969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wyman, S.K.; Knouf, E.C.; Parkin, R.K.; Fritz, B.R.; Lin, D.W.; Dennis, L.M.; Krouse, M.A.; Webster, P.J.; Tewari, M. Post-transcriptional generation of miRNA variants by multiple nucleotidyl transferases contributes to miRNA transcriptome complexity. Genome Res. 2011, 21, 1450–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koppers-Lalic, D.; Hackenberg, M.; Bijnsdorp, I.V.; van Eijndhoven, M.A.J.; Sadek, P.; Sie, D.; Zini, N.; Middeldorp, J.M.; Ylstra, B.; de Menezes, R.X.; et al. Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Rep. 2014, 8, 1649–1658. [Google Scholar] [CrossRef] [Green Version]
- Tosar, J.P.; Gámbaro, F.; Sanguinetti, J.; Bonilla, B.; Witwer, K.W.; Cayota, A. Assessment of small RNA sorting into different extracellular fractions revealed by high-throughput sequencing of breast cell lines. Nucleic Acids Res. 2015, 43, 5601–5616. [Google Scholar] [CrossRef] [Green Version]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
- Pucci, F.; Garris, C.; Lai, C.P.; Newton, A.; Pfirschke, C.; Engblom, C.; Alvarez, D.; Sprachman, M.; Evavold, C.; Magnuson, A.; et al. SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions. Science 2016, 352, 242–246. [Google Scholar] [CrossRef] [Green Version]
- Zomer, A.; Maynard, C.; Verweij, F.J.; Kamermans, A.; Schäfer, R.; Beerling, E.; Schiffelers, R.M.; De Wit, E.; Berenguer, J.; Ellenbroek, S.I.J.; et al. In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 2015, 161, 1046–1057. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.T.; Huang, C.C.; You, H.L.; Chou, F.F.; Hu, C.C.A.; Chao, F.P.; Chen, C.M.; Cheng, J.T. Overexpression of tumor susceptibility gene TSG101 in human papillary thyroid carcinomas. Oncogene 2002, 21, 4830–4837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koliopanos, A.; Friess, H.; Kleeff, J.; Shi, X.; Liao, Q.; Pecker, I.; Vlodavsky, I.; Zimmermann, A.; Büchler, M.W. Heparanase expression in primary and metastatic pancreatic cancer. Cancer Res. 2001, 61, 4655–4659. [Google Scholar] [PubMed]
- Imjeti, N.S.; Menck, K.; Egea-Jimenez, A.L.; Lecointre, C.; Lembo, F.; Bouguenina, H.; Badache, A.; Ghossoub, R.; David, G.; Roche, S.; et al. Syntenin mediates SRC function in exosomal cell-to-cell communication. Proc. Natl. Acad. Sci. USA 2017, 114, 12495–12500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.H.; Chennakrishnaiah, S.; Audemard, E.; Montermini, L.; Meehan, B.; Rak, J. Oncogenic ras-driven cancer cell vesiculation leads to emission of double-stranded DNA capable of interacting with target cells. Biochem. Biophys. Res. Commun. 2014, 451, 295–301. [Google Scholar] [CrossRef] [Green Version]
- Wei, Y.; Wang, D.; Jin, F.; Bian, Z.; Li, L.; Liang, H.; Li, M.; Shi, L.; Pan, C.; Zhu, D.; et al. Pyruvate kinase type M2 promotes tumour cell exosome release via phosphorylating synaptosome-associated protein 23. Nat. Commun. 2017, 8, 14041. [Google Scholar] [CrossRef] [Green Version]
- Farahani, M.; Rubbi, C.; Liu, L.; Slupsky, J.R.; Kalakonda, N. CLL exosomes modulate the transcriptome and behaviour of recipient stromal cells and are selectively enriched in MIR-202-3p. PLoS ONE 2015, 10, 141429. [Google Scholar] [CrossRef] [Green Version]
- Ostenfeld, M.S.; Jeppesen, D.K.; Laurberg, J.R.; Boysen, A.T.; Bramsen, J.B.; Primdal-Bengtson, B.; Hendrix, A.; Lamy, P.; Dagnaes-Hansen, F.; Rasmussen, M.H.; et al. Cellular disposal of miR23b by RAB27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Res. 2014, 74, 5758–5771. [Google Scholar] [CrossRef] [Green Version]
- Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Twyman-Saint Victor, C.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [Green Version]
- Bayraktar, R.; Van Roosbroeck, K.; Calin, G.A. Cell-to-cell communication: microRNAs as hormones. Mol. Oncol. 2017, 11, 1673–1686. [Google Scholar] [CrossRef] [Green Version]
- Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef] [Green Version]
- Fang, J.H.; Zhang, Z.J.; Shang, L.R.; Luo, Y.W.; Lin, Y.F.; Yuan, Y.; Zhuang, S.M. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018, 68, 1459–1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-Secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- King, H.W.; Michael, M.Z.; Gleadle, J.M. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 2012, 12, 421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3758. [Google Scholar] [CrossRef] [PubMed]
- Tadokoro, H.; Umezu, T.; Ohyashiki, K.; Hirano, T.; Ohyashiki, J.H. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J. Biol. Chem. 2013, 288, 34343–34351. [Google Scholar] [CrossRef] [Green Version]
- Lin, X.J.; Fang, J.H.; Yang, X.J.; Zhang, C.; Yuan, Y.; Zheng, L.; Zhuang, S.M. Hepatocellular Carcinoma Cell-Secreted Exosomal MicroRNA-210 Promotes Angiogenesis In Vitro and In Vivo. Mol. Ther. - Nucleic Acids 2018, 11, 243–252. [Google Scholar] [CrossRef] [Green Version]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Pang, W.; Su, J.; Wang, Y.; Feng, H.; Dai, X.; Yuan, Y.; Chen, X.; Yao, W. Pancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblasts. Cancer Sci. 2015, 106, 1362–1369. [Google Scholar] [CrossRef]
- Rana, S.; Malinowska, K.; Zöller, M. Exosomal tumor microRNA modulates premetastatic organ cells. Neoplasia (United States) 2013, 15, 281–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baroni, S.; Romero-Cordoba, S.; Plantamura, I.; Dugo, M.; D’Ippolito, E.; Cataldo, A.; Cosentino, G.; Angeloni, V.; Rossini, A.; Daidone, M.G.; et al. Exosome-mediated delivery of miR-9 induces cancer-Associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 2016, 7, e2312. [Google Scholar] [CrossRef] [PubMed]
- Nouraee, N.; Van Roosbroeck, K.; Vasei, M.; Semnani, S.; Samaei, N.M.; Naghshvar, F.; Omidi, A.A.; Calin, G.A.; Mowla, S.J. Expression, Tissue Distribution and Function of miR-21 in Esophageal Squamous Cell Carcinoma. PLoS ONE 2013, 8, e73009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donnarumma, E.; Fiore, D.; Nappa, M.; Roscigno, G.; Adamo, A.; Iaboni, M.; Russo, V.; Affinito, A.; Puoti, I.; Quintavalle, C.; et al. Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 2017, 8, 19592–19608. [Google Scholar] [CrossRef] [Green Version]
- Au Yeung, C.L.; Co, N.N.; Tsuruga, T.; Yeung, T.L.; Kwan, S.Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.K.; et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016, 7, 11150. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Zhao, C.; Shi, H.; Zhang, B.; Zhang, L.; Zhang, X.; Wang, S.; Wu, X.; Yang, T.; Huang, F.; et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br. J. Cancer 2014, 110, 1199–1210. [Google Scholar] [CrossRef]
- Ma, P.; Pan, Y.; Li, W.; Sun, C.; Liu, J.; Xu, T.; Shu, Y. Extracellular vesicles-mediated noncoding RNAs transfer in cancer. J. Hematol. Oncol. 2017, 10, 57. [Google Scholar] [CrossRef] [Green Version]
- Alfarouk, K.O.; Verduzco, D.; Rauch, C.; Muddathir, A.K.; Bashir, A.H.H.; Elhassan, G.O.; Ibrahim, M.E.; Orozco, J.D.P.; Cardone, R.A.; Reshkin, S.J.; et al. Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question. Oncoscience 2014, 1, 777–802. [Google Scholar] [CrossRef]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
- Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.F.; Li, S.; Chin, A.R.; et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [Green Version]
- La Shu, S.; Yang, Y.; Allen, C.L.; Maguire, O.; Minderman, H.; Sen, A.; Ciesielski, M.J.; Collins, K.A.; Bush, P.J.; Singh, P.; et al. Metabolic reprogramming of stromal fibroblasts by melanoma exosome microRNA favours a pre-metastatic microenvironment. Sci. Rep. 2018, 8, 12905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, Y.; Liu, S.; Yin, S.; Niu, W.; Xiong, W.; Tan, M.; Li, G.; Zhou, M. The reverse Warburg effect is likely to be an Achilles’ heel of cancer that can be exploited for cancer therapy. Oncotarget 2017, 8, 57813–57825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munn, L.L. Cancer and inflammation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Challagundla, K.B.; Wise, P.M.; Neviani, P.; Chava, H.; Murtadha, M.; Xu, T.; Kennedy, R.; Ivan, C.; Zhang, X.; Vannini, I.; et al. Exosome-Mediated Transfer of microRNAs Within the Tumor Microenvironment and Neuroblastoma Resistance to Chemotherapy. J. Natl. Cancer Inst. 2015, 107, djv135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, Y.L.; Hung, J.Y.; Chang, W.A.; Jian, S.F.; Lin, Y.S.; Pan, Y.C.; Wu, C.Y.; Kuo, P.L. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol. Ther. 2018, 26, 568–581. [Google Scholar] [CrossRef] [Green Version]
- Takano, Y.; Masuda, T.; Iinuma, H.; Yamaguchi, R.; Sato, K.; Tobo, T.; Hirata, H.; Kuroda, Y.; Nambara, S.; Hayashi, N.; et al. Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget 2017, 8, 78598–78613. [Google Scholar] [CrossRef] [Green Version]
- Abels, E.R.; Maas, S.L.N.; Nieland, L.; Wei, Z.; Cheah, P.S.; Tai, E.; Kolsteeg, C.-J.J.; Dusoswa, S.A.; Ting, D.T.; Hickman, S.; et al. Glioblastoma-Associated Microglia Reprogramming Is Mediated by Functional Transfer of Extracellular miR-21. Cell Rep. 2019, 28, 3105–3119.e7. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Chen, J.; Su, F.F.; Yu, B.; Su, F.F.; Lin, L.; Liu, Y.; Huang, J.D.; Song, E.; M., Y.; et al. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 2011, 10, 117. [Google Scholar] [CrossRef] [Green Version]
- DeNardo, D.G.; Johansson, M.; Coussens, L.M. Immune cells as mediators of solid tumor metastasis. Cancer Metastasis Rev. 2008, 27, 11–18. [Google Scholar] [CrossRef]
- Kitamura, T.; Qian, B.Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef]
- Ye, S.B.; Zhang, H.; Cai, T.T.; Liu, Y.N.; Ni, J.J.; He, J.; Peng, J.Y.; Chen, Q.Y.; Mo, H.Y.; Jun-Cui; et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016, 240, 329–340. [Google Scholar] [CrossRef] [PubMed]
- Ye, S.B.; Li, Z.L.; Luo, D.H.; Huang, B.J.; Chen, Y.S.; Zhang, X.S.; Cui, J.; Zeng, Y.X.; Li, J. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget 2014, 5, 5439–5452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, Y.; Cai, X.; Chen, X.; Liang, H.; Zhang, Y.; Li, J.; Wang, Z.; Chen, X.; Zhang, W.; Yokoyama, S.; et al. Tumor-secreted miR-214 induces regulatory T cells: A major link between immune evasion and tumor growth. Cell Res. 2014, 24, 1164–1180. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.; Chen, J.; Zhou, L.; Chen, W.; Ding, G.; Cao, L. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell. Immunol. 2014, 292, 65–69. [Google Scholar] [CrossRef]
- Ding, G.; Zhou, L.; Qian, Y.; Fu, M.; Chen, J.J.; Chen, J.J.; Xiang, J.; Wu, Z.; Jiang, G.; Cao, L. Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget 2015, 6, 29877–29888. [Google Scholar] [CrossRef] [Green Version]
- He, W.A.; Calore, F.; Londhe, P.; Canella, A.; Guttridge, D.C.; Croce, C.M. Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc. Natl. Acad. Sci. USA 2014, 111, 4525–4529. [Google Scholar] [CrossRef] [Green Version]
- Bliss, S.A.; Sinha, G.; Sandiford, O.A.; Williams, L.M.; Engelberth, D.J.; Guiro, K.; Isenalumhe, L.L.; Greco, S.J.; Ayer, S.; Bryan, M.; et al. Mesenchymal stem cell-derived exosomes stimulate cycling quiescence and early breast cancer dormancy in bone marrow. Cancer Res. 2016, 76, 5832–5844. [Google Scholar] [CrossRef] [Green Version]
- Lim, P.K.; Bliss, S.A.; Patel, S.A.; Taborga, M.; Dave, M.A.; Gregory, L.A.; Greco, S.J.; Bryan, M.; Patel, P.S.; Rameshwar, P. Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 2011, 71, 1550–1560. [Google Scholar] [CrossRef] [Green Version]
- Chiarini, F.; Lonetti, A.; Evangelisti, C.C.; Buontempo, F.; Orsini, E.; Evangelisti, C.C.; Cappellini, A.; Neri, L.M.; McCubrey, J.A.; Martelli, A.M. Advances in understanding the acute lymphoblastic leukemia bone marrow microenvironment: From biology to therapeutic targeting. Biochim. Biophys. Acta - Mol. Cell Res. 2016, 1863, 449–463. [Google Scholar] [CrossRef]
- Moses, B.S.; Evans, R.; Slone, W.L.; Piktel, D.; Martinez, I.; Craig, M.D.; Gibson, L.F. Bone marrow microenvironment niche regulates miR-221/222 in acute lymphoblastic Leukemia. Mol. Cancer Res. 2016, 14, 909–919. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Nguyen, L.X.T.; Li, L.; Zhao, D.; Kumar, B.; Wu, H.; Lin, A.; Pellicano, F.; Hopcroft, L.; Su, Y.L.; et al. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat. Med. 2018, 24, 450–462. [Google Scholar] [CrossRef] [PubMed]
- WHO (World Health Organization). Biomarkers in Risk Assessment: Validity and Validation (EHC 222); World Health Organization: Geneva, Switzerland, 2001. [Google Scholar]
- Goossens, N.; Nakagawa, S.; Sun, X.; Hoshida, Y. Cancer biomarker discovery and validation. Transl. Cancer Res. 2015, 4, 256–269. [Google Scholar] [PubMed]
- Pardini, B.; Sabo, A.A.; Birolo, G.; Calin, G.A. Noncoding RNAs in Extracellular Fluids as Cancer Biomarkers: The New Frontier of Liquid Biopsies. Cancers (Basel) 2019, 11, 1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rossi, G.; Ignatiadis, M. Promises and pitfalls of using liquid biopsy for precision medicine. Cancer Res. 2019, 79, 2798–2804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, Z.; Fan, J.; Hsu, Y.M.S.; Lyon, C.J.; Ning, B.; Hu, T.Y. Extracellular vesicles as cancer liquid biopsies: From discovery, validation, to clinical application. Lab Chip 2019, 19, 1114–1140. [Google Scholar] [CrossRef] [PubMed]
- Cortez, M.A.; Calin, G.A. MicroRNA identification in plasma and serum: A new tool to diagnose and monitor diseases. Expert Opin. Biol. Ther. 2009, 9, 703–711. [Google Scholar] [CrossRef]
- Park, N.J.; Zhou, H.; Elashoff, D.; Henson, B.S.; Kastratovic, D.A.; Abemayor, E.; Wong, D.T. Salivary microRNA: Discovery, characterization, and clinical utility for oral cancer detection. Clin. Cancer Res. 2009, 15, 5473–5477. [Google Scholar] [CrossRef] [Green Version]
- Chiabotto, G.; Gai, C.; Deregibus, M.C.; Camussi, G. Salivary extracellular vesicle-associated exRNA as cancer biomarker. Cancers (Basel) 2019, 11, 891. [Google Scholar] [CrossRef] [Green Version]
- Hanke, M.; Hoefig, K.; Merz, H.; Feller, A.C.; Kausch, I.; Jocham, D.; Warnecke, J.M.; Sczakiel, G. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol. Oncol. 2010, 28, 655–661. [Google Scholar] [CrossRef]
- Liu, Y.R.; Ortiz-Bonilla, C.J.; Lee, Y.F. Extracellular vesicles in bladder cancer: Biomarkers and beyond. Int. J. Mol. Sci. 2018, 19, 2822. [Google Scholar] [CrossRef] [Green Version]
- Ren, A.; Dong, Y.; Tsoi, H.; Yu, J. Detection of miRNA as non-invasive biomarkers of colorectal cancer. Int. J. Mol. Sci. 2015, 16, 2810–2823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zorofchian, S.; Iqbal, F.; Rao, M.; Aung, P.P.; Esquenazi, Y.; Ballester, L.Y. Circulating tumour DNA, microRNA and metabolites in cerebrospinal fluid as biomarkers for central nervous system malignancies. J. Clin. Pathol. 2019, 72, 271–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, G.K.; Khan, M.A.; Bhardwaj, A.; Srivastava, S.K.; Zubair, H.; Patton, M.C.; Singh, S.; Khushman, M.; Singh, A.P. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br. J. Cancer 2017, 116, 609–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Momen-Heravi, F.; Bala, S. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogramme monocytes via NF-κB pathway. Oncotarget 2018, 9, 34838–34854. [Google Scholar] [CrossRef] [Green Version]
- Shen, M.; Dong, C.; Ruan, X.; Yan, W.; Cao, M.; Pizzo, D.; Wu, X.; Yang, L.; Liu, L.; Ren, X.; et al. Chemotherapy-induced extracellular vesicle miRNAs promote breast cancer stemness by targeting OneCUT2. Cancer Res. 2019, 79, 3608–3621. [Google Scholar] [CrossRef] [PubMed]
- Bouvy, C.; Wannez, A.; Laloy, J.; Chatelain, C.; Dogné, J.M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leuk. Res. 2017, 62, 70–76. [Google Scholar] [CrossRef]
- Corcoran, C.; Rani, S.; O’Brien, K.; O’Neill, A.; Prencipe, M.; Sheikh, R.; Webb, G.; McDermott, R.; Watson, W.; Crown, J.; et al. Docetaxel-Resistance in Prostate Cancer: Evaluating Associated Phenotypic Changes and Potential for Resistance Transfer via Exosomes. PLoS ONE 2012, 7, 50999. [Google Scholar] [CrossRef]
- Crow, J.; Atay, S.; Banskota, S.; Artale, B.; Schmitt, S.; Godwin, A.K. Exosomes as mediators of platinum resistance in ovarian cancer. Oncotarget 2017, 8, 11917–11936. [Google Scholar] [CrossRef] [Green Version]
- Blower, P.E.; Chung, J.H.; Verducci, J.S.; Lin, S.; Park, J.K.; Dai, Z.; Liu, C.G.; Schmittgen, T.D.; Reinhold, W.C.; Croce, C.M.; et al. MicroRNAs modulate the chemosensitivity of tumor cells. Mol. Cancer Ther. 2008, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Rooj, A.K.; Mineo, M.; Godlewski, J. MicroRNA and extracellular vesicles in glioblastoma: small but powerful. Brain Tumor Pathol. 2016, 33, 77–88. [Google Scholar] [CrossRef] [Green Version]
- Fornari, F.; Pollutri, D.; Patrizi, C.; La Bella, T.; Marinelli, S.; Casadei Gardini, A.; Marisi, G.; Baron Toaldo, M.; Baglioni, M.; Salvatore, V.; et al. In hepatocellular carcinoma miR-221 modulates sorafenib resistance through inhibition of caspase-3–mediated apoptosis. Clin. Cancer Res. 2017, 23, 3953–3965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koga, Y.; Yasunaga, M.; Moriya, Y.; Akasu, T.; Fujita, S.; Yamamoto, S.; Matsumura, Y. Exosome can prevent RNase from degrading microRNA in feces. J. Gastrointest. Oncol. 2011, 2, 215–21522. [Google Scholar] [PubMed]
- Cheng, L.; Sharples, R.A.; Scicluna, B.J.; Hill, A.F. Exosomes provide a protective and enriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood. J. Extracell. Vesicles 2014. [Google Scholar] [CrossRef] [PubMed]
- Ge, Q.; Zhou, Y.; Lu, J.; Bai, Y.; Xie, X.; Lu, Z. MiRNA in plasma exosome is stable under different storage conditions. Molecules 2014, 19, 1568–1575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Neill, C.P.; Gilligan, K.E.; Dwyer, R.M. Role of extracellular vesicles (EVs) in cell stress response and resistance to cancer therapy. Cancers (Basel). 2019, 11, 136. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Z.; Zhang, D.; Lee, H.; Menon, A.A.; Wu, J.; Hu, K.; Jin, Y. Macrophage-derived apoptotic bodies promote the proliferation of the recipient cells via shuttling microRNA-221/222. J. Leukoc. Biol. 2017, 101, 1349–1359. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Zhang, D.; Zhu, Z.; Dela Cruz, C.S.; Jin, Y. Epithelial cell-derived microvesicles activate macrophages and promote inflammation via microvesicle-containing microRNAs. Sci. Rep. 2016, 12, 35250. [Google Scholar] [CrossRef] [Green Version]
- Saleh, A.F.; Lázaro-Ibáñez, E.; Forsgard, M.A.M.; Shatnyeva, O.; Osteikoetxea, X.; Karlsson, F.; Heath, N.; Ingelsten, M.; Rose, J.; Harris, J.; et al. Extracellular vesicles induce minimal hepatotoxicity and immunogenicity. Nanoscale 2019, 11, 6990–7001. [Google Scholar] [CrossRef]
- Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Exosomes derived from natural killer cells exert therapeutic effect in melanoma. Theranostics 2017. [Google Scholar] [CrossRef]
- Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
- Hadla, M.; Palazzolo, S.; Corona, G.; Caligiuri, I.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse models. Nanomedicine 2016, 11, 2431–2441. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fareh, M.; Almairac, F.; Turchi, L.; Burel-Vandenbos, F.; Paquis, P.; Fontaine, D.; Lacas-Gervais, S.; Junier, M.P.; Chneiweiss, H.; Virolle, T. Cell-based therapy using miR-302-367 expressing cells represses glioblastoma growth. Cell Death Dis. 2017. [Google Scholar] [CrossRef]
- Munoz, J.L.; Bliss, S.A.; Greco, S.J.; Ramkissoon, S.H.; Ligon, K.L.; Rameshwar, P. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol. Ther. - Nucleic Acids 2013. [Google Scholar] [CrossRef] [PubMed]
- Sil, S.; Dagur, R.S.; Liao, K.; Peeples, E.S.; Hu, G.; Periyasamy, P.; Buch, S. Strategies for the use of Extracellular Vesicles for the Delivery of Therapeutics. J. Neuroimmune Pharmacol. 2019. [Google Scholar] [CrossRef] [PubMed]
- Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 195–204. [Google Scholar] [CrossRef]
- Bellavia, D.; Raimondo, S.; Calabrese, G.; Forte, S.; Cristaldi, M.; Patinella, A.; Memeo, L.; Manno, M.; Raccosta, S.; Diana, P.; et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous Leukemia cell growth. Theranostics 2017, 7, 1333–1345. [Google Scholar] [CrossRef]
- Van Dommelen, S.M.; Vader, P.; Lakhal, S.; Kooijmans, S.A.A.; Van Solinge, W.W.; Wood, M.J.A.; Schiffelers, R.M. Microvesicles and exosomes: Opportunities for cell-derived membrane vesicles in drug delivery. J. Control. Release 2012, 161, 635–644. [Google Scholar] [CrossRef]
- Escudier, B.; Dorval, T.; Chaput, N.; André, F.; Caby, M.P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of the first phase 1 clinical trial. J. Transl. Med. 2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morse, M.A.; Garst, J.; Osada, T.; Khan, S.; Hobeika, A.; Clay, T.M.; Valente, N.; Shreeniwas, R.; Sutton, M.A.; Delcayre, A.; et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J. Transl. Med. 2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neviani, P.; Wise, P.M.; Murtadha, M.; Liu, C.W.; Wu, C.H.; Jong, A.Y.; Seeger, R.C.; Fabbri, M. Natural killer–derived exosomal miR-186 inhibits neuroblastoma growth and immune escape mechanisms. Cancer Res. 2019, 79, 1151–1164. [Google Scholar] [CrossRef]
- Lugini, L.; Cecchetti, S.; Huber, V.; Luciani, F.; Macchia, G.; Spadaro, F.; Paris, L.; Abalsamo, L.; Colone, M.; Molinari, A.; et al. Immune Surveillance Properties of Human NK Cell-Derived Exosomes. J. Immunol. 2012, 189, 2833–2842. [Google Scholar] [CrossRef] [Green Version]
- Jong, A.Y.; Wu, C.H.; Li, J.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J. Extracell. Vesicles 2017. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.-C.; Lee, K.-M.; Kim, D.-W.; Heo, D.S. Elevated TGF-β1 Secretion and Down-Modulation of NKG2D Underlies Impaired NK Cytotoxicity in Cancer Patients. J. Immunol. 2004, 172, 7335–7340. [Google Scholar] [CrossRef]
- Castriconi, R.; Cantoni, C.; Della Chiesa, M.; Vitale, M.; Marcenaro, E.; Conte, R.; Biassoni, R.; Bottino, C.; Moretta, L.; Moretta, A. Transforming growth factor β1 inhibits expression of NKP30 and NKG2d receptors: Consequences for the NK-mediated killing of dendritic cells. Proc. Natl. Acad. Sci. USA 2003, 100, 4120–4125. [Google Scholar] [CrossRef] [Green Version]
- Friese, M.A.; Wischhusen, J.; Wick, W.; Weiler, M.; Eisele, G.; Steinle, A.; Weller, M. RNA interference targeting transforming growth factor-β enhances NKG2D-mediated antiglioma immune response, inhibits glioma cell migration and invasiveness, and abrogates tumorigenicity in vivo. Cancer Res. 2004, 64, 7596–7603. [Google Scholar] [CrossRef] [Green Version]
- Beg, M.S.; Brenner, A.J.; Sachdev, J.; Borad, M.; Kang, Y.K.; Stoudemire, J.; Smith, S.; Bader, A.G.; Kim, S.; Hong, D.S. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investig. New Drugs 2017, 35, 180–188. [Google Scholar] [CrossRef]
- Bader, A.G. MiR-34—A microRNA replacement therapy is headed to the clinic. Front. Genet. 2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ji, Q.; Hao, X.; Zhang, M.; Tang, W.; Meng, Y.; Li, L.; Xiang, D.; DeSano, J.T.; Bommer, G.T.; Fan, D.; et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE 2009, 4, e6816. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Fu, H.; Tie, Y.; Hu, Z.; Kong, W.; Wu, Y.; Zheng, X. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett. 2009, 275, 44–53. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Kelnar, K.; Liu, B.; Chen, X.; Calhoun-Davis, T.; Li, H.; Patrawala, L.; Yan, H.; Jeter, C.; Honorio, S.; et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 2011, 17, 211–216. [Google Scholar] [CrossRef] [Green Version]
- Di Martino, M.T.; Leone, E.; Amodio, N.; Foresta, U.; Lionetti, M.; Pitari, M.R.; Gallo Cantafio, M.E.; Gullà, A.; Conforti, F.; Morelli, E.; et al. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: In vitro and in vivo evidence. Clin. Cancer Res. 2012, 18, 6260–6270. [Google Scholar] [CrossRef] [Green Version]
- Zhao, J.; Kelnar, K.; Bader, A.G. In-depth analysis shows synergy between erlotinib and miR-34a. PLoS ONE 2014, 9, e89105. [Google Scholar] [CrossRef]
- Wiggins, J.F.; Ruffino, L.; Kelnar, K.; Omotola, M.; Patrawala, L.; Brown, D.; Bader, A.G. Development of a lung cancer therapeutic based on the tumor suppressor microRNA-34. Cancer Res. 2010, 70, 5923–5930. [Google Scholar] [CrossRef] [Green Version]
- Craig, V.J.; Tzankov, A.; Flori, M.; Schmid, C.A.; BaDer, A.G.; Müller, A. Systemic microRNA-34a delivery induces apoptosis and abrogates growth of diffuse large B-cell lymphoma in vivo. Leukemia 2012, 26, 2421–2424. [Google Scholar] [CrossRef] [Green Version]
- Van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017. [Google Scholar] [CrossRef]
- Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic micrornas regulate cancer cell metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef] [Green Version]
- Yokoi, A.; Yoshioka, Y.; Yamamoto, Y.; Ishikawa, M.; Ikeda, S.I.; Kato, T.; Kiyono, T.; Takeshita, F.; Kajiyama, H.; Kikkawa, F.; et al. Malignant extracellular vesicles carrying MMP1 mRNA facilitate peritoneal dissemination in ovarian cancer. Nat. Commun. 2017. [Google Scholar] [CrossRef] [Green Version]
- Marleau, A.M.; Chen, C.S.; Joyce, J.A.; Tullis, R.H. Exosome removal as a therapeutic adjuvant in cancer. J. Transl. Med. 2012. [Google Scholar] [CrossRef] [Green Version]
- Christianson, H.C.; Svensson, K.J.; Van Kuppevelt, T.H.; 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. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [Green Version]
- Cocucci, E.; Gaudin, R.; Kirchhausen, T. Dynamin recruitment and membrane scission at the neck of a clathrin-coated pit. Mol. Biol. Cell 2014, 25, 3595–3609. [Google Scholar] [CrossRef]
- Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
Name of the Study | Disease | Phase | miRNA | Sample Source | NCT Identifier |
---|---|---|---|---|---|
Pathogenic Mechanisms of Cancer and Cardiovascular Diseases | All tumors | NA | exomiRNA | Blood | NCT03051191 |
U01-Biomarkers for Noninvasive and Early Detection of Pancreatic Cancer | Pancreatic cancer | NA | cfmiRNA, exomiRNA | Blood | NCT03886571 |
Evaluation of MicroRNA Expression in Blood and Cytology for Detecting Barrett’s Esophagus and Associated Neoplasia | Esophageal cancer | NA | exomiRNA | Blood, bile | NCT02464930 |
Prostasomes as Diagnostic Tool for Prostate Cancer Detection | Prostate cancer | NA | exomiRNA | Blood | NCT03694483 |
To Investigate the Diagnostic Accuracy of Exosomal microRNA in Predicting the Aggressiveness of Prostate Cancer in Chinese Patients | Prostate cancer | NA | exomiRNA | Urine | NCT03911999 |
Non-coding RNA in the Exosome of the Epithelia Ovarian Cancer | Ovarian cancer | NA | exomiRNA | Blood | NCT03738319 |
Identification and Characterization of Predictive Factors of Onset of Bone Metastases in Cancer Patients (PreMetOn) | Bone metastasis | NA | exomiRNA | Blood | NCT03895216 |
Neoadjuvant Nivolumab for Oral Cancer Combined With FDG and Anti-PD-L1 PET/CT Imaging for Response Prediction (NeoNivo) | Oral cancer | Phase I | EV-associated miRNA | Blood | NCT03843515 |
Circulating Exosome RNA in Lung Metastases of Primary High-Grade Osteosarcoma | Osteosarcoma | NA | exomiRNA | Blood | NCT03108677 |
Study of Exosomes in Monitoring Patients With Sarcoma (EXOSARC) | Sarcoma | NA | exomiRNA | Blood | NCT03800121 |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mills, J.; Capece, M.; Cocucci, E.; Tessari, A.; Palmieri, D. Cancer-Derived Extracellular Vesicle-Associated MicroRNAs in Intercellular Communication: One Cell’s Trash Is Another Cell’s Treasure. Int. J. Mol. Sci. 2019, 20, 6109. https://doi.org/10.3390/ijms20246109
Mills J, Capece M, Cocucci E, Tessari A, Palmieri D. Cancer-Derived Extracellular Vesicle-Associated MicroRNAs in Intercellular Communication: One Cell’s Trash Is Another Cell’s Treasure. International Journal of Molecular Sciences. 2019; 20(24):6109. https://doi.org/10.3390/ijms20246109
Chicago/Turabian StyleMills, Joseph, Marina Capece, Emanuele Cocucci, Anna Tessari, and Dario Palmieri. 2019. "Cancer-Derived Extracellular Vesicle-Associated MicroRNAs in Intercellular Communication: One Cell’s Trash Is Another Cell’s Treasure" International Journal of Molecular Sciences 20, no. 24: 6109. https://doi.org/10.3390/ijms20246109