Exosome-Mediated Signaling in Epithelial to Mesenchymal Transition and Tumor Progression
Abstract
:1. Introduction
2. Exosome Heterogeneity and Cargo Composition
3. EMT Associated with Tumor Progression: The Role of Exosomes
3.1. Exosomes in Tumor Niche
3.2. Exosomes in Migration, Invasion, and Metastasis
4. Conclusions and Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- 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]
- Thierry, A.R.; El Messaoudi, S.; Gahan, P.B.; Anker, P.; Stroun, M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef] [Green Version]
- Mittelbrunn, M.; Sanchez-Madrid, F. Intercellular communication: Diverse structures for exchange of genetic information. Nat. Rev. Mol. Cell Biol. 2012, 13, 328–335. [Google Scholar] [CrossRef]
- Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [Green Version]
- Bakhti, M.; Winter, C.; Simons, M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 2011, 286, 787–796. [Google Scholar] [CrossRef] [PubMed]
- Fruhbeis, C.; Frohlich, D.; Kuo, W.P.; Amphornrat, J.; Thilemann, S.; Saab, A.S.; Kirchhoff, F.; Mobius, W.; Goebbels, S.; Nave, K.A.; et al. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 2013, 11, e1001604. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Verrilli, M.A.; Picou, F.; Court, F.A. Schwann cell-derived exosomes enhance axonal regeneration in the peripheral nervous system. Glia 2013, 61, 1795–1806. [Google Scholar] [CrossRef] [PubMed]
- Guay, C.; Regazzi, R. Exosomes as new players in metabolic organ cross-talk. Diabetes Obes. Metab. 2017, 19 (Suppl. 1), 137–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Logozzi, M.; De Milito, A.; Lugini, L.; Borghi, M.; Calabro, L.; Spada, M.; Perdicchio, M.; Marino, M.L.; Federici, C.; Iessi, E.; et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS ONE 2009, 4, e5219. [Google Scholar] [CrossRef]
- Peinado, H.; Aleckovic, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; Garcia-Santos, G.; Ghajar, C.; 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] [PubMed] [Green Version]
- Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Thery, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; Degeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E.; et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef]
- Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Thery, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef] [Green Version]
- Willms, E.; Johansson, H.J.; Mager, I.; Lee, Y.; Blomberg, K.E.; Sadik, M.; Alaarg, A.; Smith, C.I.; Lehtio, J.; El Andaloussi, S.; et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci. Rep. 2016, 6, 22519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ogawa, Y.; Miura, Y.; Harazono, A.; Kanai-Azuma, M.; Akimoto, Y.; Kawakami, H.; Yamaguchi, T.; Toda, T.; Endo, T.; Tsubuki, M.; et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol. Pharm. Bull. 2011, 34, 13–23. [Google Scholar] [CrossRef]
- Ogawa, Y.; Taketomi, Y.; Murakami, M.; Tsujimoto, M.; Yanoshita, R. Small RNA transcriptomes of two types of exosomes in human whole saliva determined by next generation sequencing. Biol. Pharm. Bull. 2013, 36, 66–75. [Google Scholar] [CrossRef] [PubMed]
- Lasser, C.; Shelke, G.V.; Yeri, A.; Kim, D.K.; Crescitelli, R.; Raimondo, S.; Sjostrand, M.; Gho, Y.S.; van Keuren Jensen, K.; Lotvall, J. Two distinct extracellular RNA signatures released by a single cell type identified by microarray and next-generation sequencing. RNA Biol. 2017, 14, 58–72. [Google Scholar] [CrossRef] [PubMed]
- Bobrie, A.; Colombo, M.; Krumeich, S.; Raposo, G.; Thery, C. Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation. J. Extracell. Ves. 2012, 1, 18397. [Google Scholar] [CrossRef] [PubMed]
- ExoCarta. Available online: http://www.exocarta.org (accessed on 24 December 2018).
- Vesiclepedia. Available online: http://www.microvesicles.org (accessed on 24 December 2018).
- Skog, J.; Wurdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Sena-Esteves, M.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conigliaro, A.; Fontana, S.; Raimondo, S.; Alessandro, R. Exosomes: Nanocarriers of Biological Messages. Adv. Exp. Med. Biol. 2017, 998, 23–43. [Google Scholar] [PubMed]
- Nolte-’t Hoen, E.N.; Buermans, H.P.; Waasdorp, M.; Stoorvogel, W.; Wauben, M.H.; ’t Hoen, P.A. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012, 40, 9272–9285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mittelbrunn, M.; Gutierrez-Vazquez, C.; Villarroya-Beltri, C.; Gonzalez, S.; Sanchez-Cabo, F.; Gonzalez, M.A.; Bernad, A.; Sanchez-Madrid, F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat. Commun. 2011, 2, 282. [Google Scholar] [CrossRef] [PubMed]
- Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, 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]
- Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Wurdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Liu, S.C.; Luo, X.H.; Tao, G.X.; Guan, M.; Yuan, H.; Hu, D.K. Exosomal Long Noncoding RNAs are Differentially Expressed in the Cervicovaginal Lavage Samples of Cervical Cancer Patients. J. Clin. Lab. Anal. 2016, 30, 1116–1121. [Google Scholar] [CrossRef]
- Berrondo, C.; Flax, J.; Kucherov, V.; Siebert, A.; Osinski, T.; Rosenberg, A.; Fucile, C.; Richheimer, S.; Beckham, C.J. Expression of the Long Non-Coding RNA HOTAIR Correlates with Disease Progression in Bladder Cancer and Is Contained in Bladder Cancer Patient Urinary Exosomes. PLoS ONE 2016, 11, e0147236. [Google Scholar] [CrossRef]
- Conigliaro, A.; Costa, V.; Lo Dico, A.; Saieva, L.; Buccheri, S.; Dieli, F.; Manno, M.; Raccosta, S.; Mancone, C.; Tripodi, M.; et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 2015, 14, 155. [Google Scholar] [CrossRef]
- Santos, M.F.; Rappa, G.; Karbanova, J.; Kurth, T.; Corbeil, D.; Lorico, A. VAMP-associated protein-A and oxysterol-binding protein-related protein 3 promote the entry of late endosomes into the nucleoplasmic reticulum. J. Biol. Chem. 2018, 293, 13834–13848. [Google Scholar] [CrossRef]
- Rappa, G.; Santos, M.F.; Green, T.M.; Karbanova, J.; Hassler, J.; Bai, Y.; Barsky, S.H.; Corbeil, D.; Lorico, A. Nuclear transport of cancer extracellular vesicle-derived biomaterials through nuclear envelope invagination-associated late endosomes. Oncotarget 2017, 8, 14443–14461. [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] [PubMed]
- Villarroya-Beltri, C.; Gutierrez-Vazquez, C.; Sanchez-Cabo, F.; Perez-Hernandez, D.; Vazquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sanchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef]
- Hobor, F.; Dallmann, A.; Ball, N.J.; Cicchini, C.; Battistelli, C.; Ogrodowicz, R.W.; Christodoulou, E.; Martin, S.R.; Castello, A.; Tripodi, M.; et al. A cryptic RNA-binding domain mediates Syncrip recognition and exosomal partitioning of miRNA targets. Nat. Commun. 2018, 9, 831. [Google Scholar] [CrossRef] [PubMed]
- Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef]
- Kucharzewska, P.; Christianson, H.C.; Welch, J.E.; Svensson, K.J.; Fredlund, E.; Ringner, M.; Morgelin, M.; Bourseau-Guilmain, E.; Bengzon, J.; Belting, M. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc. Natl. Acad. Sci. USA 2013, 110, 7312–7317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- French, K.C.; Antonyak, M.A.; Cerione, R.A. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Semin. Cell Dev. Biol. 2017, 67, 48–55. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [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]
- Wei, G.; Jie, Y.; Haibo, L.; Chaoneng, W.; Dong, H.; Jianbing, Z.; Junjie, G.; Leilei, M.; Hongtao, S.; Yunzeng, Z.; et al. Dendritic cells derived exosomes migration to spleen and induction of inflammation are regulated by CCR7. Sci. Rep. 2017, 7, 42996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corrado, C.; Saieva, L.; Raimondo, S.; Santoro, A.; De Leo, G.; Alessandro, R. Chronic myelogenous leukaemia exosomes modulate bone marrow microenvironment through activation of epidermal growth factor receptor. J. Cell. Mol. Med. 2016, 20, 1829–1839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taverna, S.; Pucci, M.; Giallombardo, M.; Di Bella, M.A.; Santarpia, M.; Reclusa, P.; Gil-Bazo, I.; Rolfo, C.; Alessandro, R. Amphiregulin contained in NSCLC-exosomes induces osteoclast differentiation through the activation of EGFR pathway. Sci. Rep. 2017, 7, 3170. [Google Scholar] [CrossRef]
- Raimondo, S.; Saieva, L.; Corrado, C.; Fontana, S.; Flugy, A.; Rizzo, A.; De Leo, G.; Alessandro, R. Chronic myeloid leukemia-derived exosomes promote tumor growth through an autocrine mechanism. Cell Commun. Signal. 2015, 13, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, L.; Yang, F.; Jiang, L.; Chen, Y.; Wang, K.; Xu, F.; Wei, Y.; Cao, X.; Wang, J.; Cai, Z. Exosomes with membrane-associated TGF-beta1 from gene-modified dendritic cells inhibit murine EAE independently of MHC restriction. Eur. J. Immunol. 2013, 43, 2461–2472. [Google Scholar] [CrossRef] [PubMed]
- Sakakura, H.; Mii, S.; Hagiwara, S.; Kato, T.; Yamamoto, N.; Hibi, H.; Takahashi, M.; Murakumo, Y. CD109 is a component of exosome secreted from cultured cells. Biochem. Biophys. Res. Commun. 2016, 469, 816–822. [Google Scholar] [CrossRef] [PubMed]
- Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef]
- Brabletz, T.; Jung, A.; Reu, S.; Porzner, M.; Hlubek, F.; Kunz-Schughart, L.A.; Knuechel, R.; Kirchner, T. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl. Acad. Sci. USA 2001, 98, 10356–10361. [Google Scholar] [CrossRef]
- Dahl, U.; Sjodin, A.; Larue, L.; Radice, G.L.; Cajander, S.; Takeichi, M.; Kemler, R.; Semb, H. Genetic dissection of cadherin function during nephrogenesis. Mol. Cell. Biol. 2002, 22, 1474–1487. [Google Scholar] [CrossRef]
- Perez-Moreno, M.A.; Locascio, A.; Rodrigo, I.; Dhondt, G.; Portillo, F.; Nieto, M.A.; Cano, A. A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J. Biol. Chem. 2001, 276, 27424–27431. [Google Scholar] [CrossRef]
- Nieto, M.A. The snail superfamily of zinc-finger transcription factors. Nat. Rev. Mol. Cell Biol. 2002, 3, 155–166. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Mani, S.A.; Donaher, J.L.; Ramaswamy, S.; Itzykson, R.A.; Come, C.; Savagner, P.; Gitelman, I.; Richardson, A.; Weinberg, R.A. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 2004, 117, 927–939. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, M.; Aladowicz, E.; Lanfrancone, L.; Goding, C.R. Tbx3 represses E-cadherin expression and enhances melanoma invasiveness. Cancer Res. 2008, 68, 7872–7881. [Google Scholar] [CrossRef] [PubMed]
- Peinado, H.; Olmeda, D.; Cano, A. Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat. Rev. Cancer 2007, 7, 415–428. [Google Scholar] [CrossRef] [PubMed]
- Battistelli, C.; Cicchini, C.; Santangelo, L.; Tramontano, A.; Grassi, L.; Gonzalez, F.J.; de Nonno, V.; Grassi, G.; Amicone, L.; Tripodi, M. The Snail repressor recruits EZH2 to specific genomic sites through the enrollment of the lncRNA HOTAIR in epithelial-to-mesenchymal transition. Oncogene 2017, 36, 942–955. [Google Scholar] [CrossRef]
- Battistelli, C.; Tripodi, M.; Cicchini, C. Targeting of polycombs to DNA in EMT. Oncotarget 2017, 8, 57936–57937. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.Y.; Wong, M.K.; Tan, T.Z.; Kuay, K.T.; Ng, A.H.; Chung, V.Y.; Chu, Y.S.; Matsumura, N.; Lai, H.C.; Lee, Y.F.; et al. An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase inhibitor, saracatinib (AZD0530). Cell Death Dis. 2013, 4, e915. [Google Scholar] [CrossRef] [PubMed]
- Schliekelman, M.J.; Taguchi, A.; Zhu, J.; Dai, X.; Rodriguez, J.; Celiktas, M.; Zhang, Q.; Chin, A.; Wong, C.H.; Wang, H.; et al. Molecular portraits of epithelial, mesenchymal, and hybrid States in lung adenocarcinoma and their relevance to survival. Cancer Res. 2015, 75, 1789–1800. [Google Scholar] [CrossRef] [PubMed]
- Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468. [Google Scholar] [CrossRef]
- Ruscetti, M.; Quach, B.; Dadashian, E.L.; Mulholland, D.J.; Wu, H. Tracking and Functional Characterization of Epithelial-Mesenchymal Transition and Mesenchymal Tumor Cells during Prostate Cancer Metastasis. Cancer Res. 2015, 75, 2749–2759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamashita, N.; Tokunaga, E.; Iimori, M.; Inoue, Y.; Tanaka, K.; Kitao, H.; Saeki, H.; Oki, E.; Maehara, Y. Epithelial Paradox: Clinical Significance of Coexpression of E-cadherin and Vimentin With Regard to Invasion and Metastasis of Breast Cancer. Clin. Breast Cancer 2018, 18, e1003–e1009. [Google Scholar] [CrossRef] [PubMed]
- Conigliaro, A.; Amicone, L.; Costa, V.; De Santis Puzzonia, M.; Mancone, C.; Sacchetti, B.; Cicchini, C.; Garibaldi, F.; Brenner, D.A.; Kisseleva, T.; et al. Evidence for a common progenitor of epithelial and mesenchymal components of the liver. Cell Death Differ. 2013, 20, 1116–1123. [Google Scholar] [CrossRef] [Green Version]
- Garibaldi, F.; Cicchini, C.; Conigliaro, A.; Santangelo, L.; Cozzolino, A.M.; Grassi, G.; Marchetti, A.; Tripodi, M.; Amicone, L. An epistatic mini-circuitry between the transcription factors Snail and HNF4alpha controls liver stem cell and hepatocyte features exhorting opposite regulation on stemness-inhibiting microRNAs. Cell Death Differ. 2012, 19, 937–946. [Google Scholar] [CrossRef]
- Diaz-Lopez, A.; Moreno-Bueno, G.; Cano, A. Role of microRNA in epithelial to mesenchymal transition and metastasis and clinical perspectives. Cancer Manag. Res. 2014, 6, 205–216. [Google Scholar] [PubMed]
- Costa, V.; Lo Dico, A.; Rizzo, A.; Rajata, F.; Tripodi, M.; Alessandro, R.; Conigliaro, A. MiR-675-5p supports hypoxia induced epithelial to mesenchymal transition in colon cancer cells. Oncotarget 2017, 8, 24292–24302. [Google Scholar] [CrossRef] [Green Version]
- Jolly, M.K.; Tripathi, S.C.; Jia, D.; Mooney, S.M.; Celiktas, M.; Hanash, S.M.; Mani, S.A.; Pienta, K.J.; Ben-Jacob, E.; Levine, H. Stability of the hybrid epithelial/mesenchymal phenotype. Oncotarget 2016, 7, 27067–27084. [Google Scholar] [CrossRef] [Green Version]
- Lu, M.; Jolly, M.K.; Levine, H.; Onuchic, J.N.; Ben-Jacob, E. MicroRNA-based regulation of epithelial-hybrid-mesenchymal fate determination. Proc. Natl. Acad. Sci. USA 2013, 110, 18144–18149. [Google Scholar] [CrossRef] [Green Version]
- Tian, X.J.; Zhang, H.; Xing, J. Coupled reversible and irreversible bistable switches underlying TGFbeta-induced epithelial to mesenchymal transition. Biophys. J. 2013, 105, 1079–1089. [Google Scholar] [CrossRef]
- Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef] [Green Version]
- Green, T.M.; Alpaugh, M.L.; Barsky, S.H.; Rappa, G.; Lorico, A. Breast Cancer-Derived Extracellular Vesicles: Characterization and Contribution to the Metastatic Phenotype. Biomed. Res. Int. 2015, 2015, 634865. [Google Scholar] [CrossRef]
- Cesi, G.; Walbrecq, G.; Margue, C.; Kreis, S. Transferring intercellular signals and traits between cancer cells: Extracellular vesicles as “homing pigeons”. Cell Commun. Signal. 2016, 14, 13. [Google Scholar] [CrossRef] [PubMed]
- You, Y.; Shan, Y.; Chen, J.; Yue, H.; You, B.; Shi, S.; Li, X.; Cao, X. Matrix metalloproteinase 13-containing exosomes promote nasopharyngeal carcinoma metastasis. Cancer Sci. 2015, 106, 1669–1677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zomer, A.; Maynard, C.; Verweij, F.J.; Kamermans, A.; Schafer, R.; Beerling, E.; Schiffelers, R.M.; de Wit, E.; Berenguer, J.; Ellenbroek, S.I.; et al. In Vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 2015, 161, 1046–1057. [Google Scholar] [CrossRef] [PubMed]
- Tang, M.K.; Wong, A.S. Exosomes: Emerging biomarkers and targets for ovarian cancer. Cancer Lett. 2015, 367, 26–33. [Google Scholar] [CrossRef]
- Aga, M.; Bentz, G.L.; Raffa, S.; Torrisi, M.R.; Kondo, S.; Wakisaka, N.; Yoshizaki, T.; Pagano, J.S.; Shackelford, J. Exosomal HIF1alpha supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene 2014, 33, 4613–4622. [Google Scholar] [CrossRef] [PubMed]
- Franzen, C.A.; Blackwell, R.H.; Todorovic, V.; Greco, K.A.; Foreman, K.E.; Flanigan, R.C.; Kuo, P.C.; Gupta, G.N. Urothelial cells undergo epithelial-to-mesenchymal transition after exposure to muscle invasive bladder cancer exosomes. Oncogenesis 2015, 4, e163. [Google Scholar] [CrossRef] [PubMed]
- Jeppesen, D.K.; Nawrocki, A.; Jensen, S.G.; Thorsen, K.; Whitehead, B.; Howard, K.A.; Dyrskjot, L.; Orntoft, T.F.; Larsen, M.R.; Ostenfeld, M.S. Quantitative proteomics of fractionated membrane and lumen exosome proteins from isogenic metastatic and nonmetastatic bladder cancer cells reveal differential expression of EMT factors. Proteomics 2014, 14, 699–712. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Guan, X.; Zhang, Y.; Ge, S.; Zhang, L.; Li, H.; Wang, X.; Liu, R.; Ning, T.; Deng, T.; et al. Exosomal miR-27a Derived from Gastric Cancer Cells Regulates the Transformation of Fibroblasts into Cancer-Associated Fibroblasts. Cell. Physiol. Biochem. 2018, 49, 869–883. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Yang, X.; Li, R.; Wang, L.; Gu, Y.; Zhao, Y.; Huang, K.H.; Cheng, T.; Yuan, Y.; Gao, S. Long non-coding RNA MYU promotes prostate cancer proliferation by mediating the miR-184/c-Myc axis. Oncol. Rep. 2018, 40, 2814–2825. [Google Scholar] [CrossRef]
- Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef] [PubMed]
- Ringuette Goulet, C.; Bernard, G.; Tremblay, S.; Chabaud, S.; Bolduc, S.; Pouliot, F. Exosomes Induce Fibroblast Differentiation into Cancer-Associated Fibroblasts through TGFbeta Signaling. Mol. Cancer Res. 2018, 16, 1196–1204. [Google Scholar] [CrossRef] [PubMed]
- Principe, S.; Mejia-Guerrero, S.; Ignatchenko, V.; Sinha, A.; Ignatchenko, A.; Shi, W.; Pereira, K.; Su, S.; Huang, S.H.; O’Sullivan, B.; et al. Proteomic Analysis of Cancer-Associated Fibroblasts Reveals a Paracrine Role for MFAP5 in Human Oral Tongue Squamous Cell. Carcinoma. J. Proteome Res. 2018, 17, 2045–2059. [Google Scholar] [CrossRef] [PubMed]
- Herrera, M.; Llorens, C.; Rodriguez, M.; Herrera, A.; Ramos, R.; Gil, B.; Candia, A.; Larriba, M.J.; Garre, P.; Earl, J.; et al. Differential distribution and enrichment of non-coding RNAs in exosomes from normal and Cancer-associated fibroblasts in colorectal cancer. Mol. Cancer 2018, 17, 114. [Google Scholar] [CrossRef]
- Zhang, Z.; Li, X.; Sun, W.; Yue, S.; Yang, J.; Li, J.; Ma, B.; Wang, J.; Yang, X.; Pu, M.; et al. Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Lett. 2017, 397, 33–42. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Zhao, H.; Yang, L.; Baddour, J.; Achreja, A.; Bernard, V.; Moss, T.; Marini, J.C.; Tudawe, T.; Seviour, E.G.; et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. Elife 2016, 5, e10250. [Google Scholar] [CrossRef] [PubMed]
- Muller, L.; Mitsuhashi, M.; Simms, P.; Gooding, W.E.; Whiteside, T.L. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 2016, 6, 20254. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Yang, Y.; Wang, W.; Zhang, Y.; Chen, Z.; Hao, C.; Zhang, J. Melanoma-released exosomes directly activate the mitochondrial apoptotic pathway of CD4(+) T cells through their microRNA cargo. Exp. Cell Res. 2018, 371, 364–371. [Google Scholar] [CrossRef]
- Hirschberger, S.; Hinske, L.C.; Kreth, S. MiRNAs: Dynamic regulators of immune cell functions in inflammation and cancer. Cancer Lett. 2018, 431, 11–21. [Google Scholar] [CrossRef]
- Clayton, A.; Al-Taei, S.; Webber, J.; Mason, M.D.; Tabi, Z. Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. J. Immunol. 2011, 187, 676–683. [Google Scholar] [CrossRef] [PubMed]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [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.; 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]
- Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Ma, X.; Wang, J.; Zhao, Y.; Wang, Y.; Bihl, J.C.; Chen, Y.; Jiang, C. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget 2017, 8, 36137–36148. [Google Scholar] [CrossRef] [PubMed]
- Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef]
- Petrova, V.; Annicchiarico-Petruzzelli, M.; Melino, G.; Amelio, I. The hypoxic tumour microenvironment. Oncogenesis 2018, 7, 10. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Zhou, X.; Yao, Q.; Liu, Y.; Zhang, H.; Dong, Z. HIF-1-mediated production of exosomes during hypoxia is protective in renal tubular cells. Am. J. Physiol. Ren. Physiol. 2017, 313, F906–F913. [Google Scholar] [CrossRef]
- Shao, C.; Yang, F.; Miao, S.; Liu, W.; Wang, C.; Shu, Y.; Shen, H. Role of hypoxia-induced exosomes in tumor biology. Mol. Cancer 2018, 17, 120. [Google Scholar] [CrossRef]
- Dey, N.; De, P.; Brian, L.J. Evading anti-angiogenic therapy: Resistance to anti-angiogenic therapy in solid tumors. Am. J. Transl. Res. 2015, 7, 1675–1698. [Google Scholar] [CrossRef]
- Tang, M.K.S.; Yue, P.Y.K.; Ip, P.P.; Huang, R.L.; Lai, H.C.; Cheung, A.N.Y.; Tse, K.Y.; Ngan, H.Y.S.; Wong, A.S.T. Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nat. Commun. 2018, 9, 2270. [Google Scholar] [CrossRef] [PubMed]
- Song, X.; Ding, Y.; Liu, G.; Yang, X.; Zhao, R.; Zhang, Y.; Zhao, X.; Anderson, G.J.; Nie, G. Cancer Cell-derived Exosomes Induce Mitogen-activated Protein Kinase-dependent Monocyte Survival by Transport. of Functional Receptor Tyrosine Kinases. J. Biol. Chem. 2016, 291, 8453–8464. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.A.; Barger, J.F.; Lovat, F.; Gao, M.; Otterson, G.A.; Nana-Sinkam, P. Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget 2016, 7, 54852–54866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.; Guo, P.; He, Y.; Chen, Z.; Chen, L.; Luo, Y.; Qi, L.; Liu, Y.; Wu, Q.; Cui, Y.; et al. HCC-derived exosomes elicit HCC progression and recurrence by epithelial-mesenchymal transition through MAPK/ERK signalling pathway. Cell Death Dis. 2018, 9, 513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Higginbotham, J.N.; Demory Beckler, M.; Gephart, J.D.; Franklin, J.L.; Bogatcheva, G.; Kremers, G.J.; Piston, D.W.; Ayers, G.D.; McConnell, R.E.; Tyska, M.J.; et al. Amphiregulin exosomes increase cancer cell invasion. Curr. Biol. 2011, 21, 779–786. [Google Scholar] [CrossRef] [PubMed]
- Harris, D.A.; Patel, S.H.; Gucek, M.; Hendrix, A.; Westbroek, W.; Taraska, J.W. Exosomes released from breast cancer carcinomas stimulate cell movement. PLoS ONE 2015, 10, e0117495. [Google Scholar] [CrossRef] [PubMed]
- Sung, B.H.; Ketova, T.; Hoshino, D.; Zijlstra, A.; Weaver, A.M. Directional cell movement through tissues is controlled by exosome secretion. Nat. Commun. 2015, 6, 7164. [Google Scholar] [CrossRef]
- Schillaci, O.; Fontana, S.; Monteleone, F.; Taverna, S.; Di Bella, M.A.; Di Vizio, D.; Alessandro, R. Exosomes from metastatic cancer cells transfer amoeboid phenotype to non-metastatic cells and increase endothelial permeability: Their emerging role in tumor heterogeneity. Sci. Rep. 2017, 7, 4711. [Google Scholar] [CrossRef]
- Dong, L.; Lin, W.; Qi, P.; Xu, M.D.; Wu, X.; Ni, S.; Huang, D.; Weng, W.W.; Tan, C.; Sheng, W.; et al. Circulating Long RNAs in Serum Extracellular Vesicles: Their Characterization and Potential Application as Biomarkers for Diagnosis of Colorectal Cancer. Cancer Epidemiol. Biomarkers Prev. 2016, 25, 1158–1166. [Google Scholar] [CrossRef]
- Battistelli, C.; Sabarese, G.; Santangelo, L.; Montaldo, C.; Gonzalez, F.J.; Tripodi, M.; Cicchini, C. The lncRNA HOTAIR transcription is controlled by HNF4alpha-induced chromatin topology modulation. Cell Death Differ. 2018. [Google Scholar] [CrossRef]
- Amicone, L.; Citarella, F.; Cicchini, C. Epigenetic regulation in hepatocellular carcinoma requires long noncoding RNAs. Biomed. Res. Int. 2015, 2015, 473942. [Google Scholar] [CrossRef] [PubMed]
- Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana, J.L. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012, 151, 1542–1556. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Li, W.; Zhang, X.; Wang, J.; Li, M.; Cao, C.; Tan, J.; Ma, D.; Gao, Q. TGFbeta1 in fibroblasts-derived exosomes promotes epithelial-mesenchymal transition of ovarian cancer cells. Oncotarget 2017, 8, 96035–96047. [Google Scholar]
- Zhao, X.; Wu, X.; Qian, M.; Song, Y.; Wu, D.; Zhang, W. Knockdown of TGF-beta1 expression in human umbilical cord mesenchymal stem cells reverts their exosome-mediated EMT promoting effect on lung cancer cells. Cancer Lett. 2018, 428, 34–44. [Google Scholar] [CrossRef] [PubMed]
- Ramteke, A.; Ting, H.; Agarwal, C.; Mateen, S.; Somasagara, R.; Hussain, A.; Graner, M.; Frederick, B.; Agarwal, R.; Deep, G. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 2015, 54, 554–565. [Google Scholar] [CrossRef]
- Xue, M.; Chen, W.; Xiang, A.; Wang, R.; Chen, H.; Pan, J.; Pang, H.; An, H.; Wang, X.; Hou, H.; et al. Hypoxic exosomes facilitate bladder tumor growth and development through transferring long non-coding RNA-UCA1. Mol. Cancer 2017, 16, 143. [Google Scholar] [CrossRef]
- Takahashi, K.; Yan, I.K.; Haga, H.; Patel, T. Modulation of hypoxia-signaling pathways by extracellular linc-RoR. J. Cell Sci. 2014, 127, 1585–1594. [Google Scholar] [CrossRef] [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]
- Zhou, C.F.; Ma, J.; Huang, L.; Yi, H.Y.; Zhang, Y.M.; Wu, X.G.; Yan, R.M.; Liang, L.; Zhong, M.; Yu, Y.H.; et al. Cervical squamous cell carcinoma-secreted exosomal miR-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene 2018. [Google Scholar] [CrossRef]
- Karaosmanoglu, O.; Banerjee, S.; Sivas, H. Identification of biomarkers associated with partial epithelial to mesenchymal transition in the secretome of slug over-expressing hepatocellular carcinoma cells. Cell. Oncol. 2018, 41, 439–453. [Google Scholar] [CrossRef] [PubMed]
- Grosse-Wilde, A.; Fouquier d’Herouel, A.; McIntosh, E.; Ertaylan, G.; Skupin, A.; Kuestner, R.E.; del Sol, A.; Walters, K.A.; Huang, S. Stemness of the hybrid Epithelial/Mesenchymal State in Breast Cancer and Its Association with Poor Survival. PLoS ONE 2015, 10, e0126522. [Google Scholar] [CrossRef] [PubMed]
- Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.; Pochampally, R.; Watabe, K.; Lu, Z.; Mo, Y.Y. Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Mol. Cancer 2014, 13, 256. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Yan, C.; Mu, L.; Huang, K.; Li, X.; Tao, D.; Wu, Y.; Qin, J. Fibroblast-Derived Exosomes Contribute to Chemoresistance through Priming Cancer Stem Cells in Colorectal Cancer. PLoS ONE 2015, 10, e0125625. [Google Scholar] [CrossRef] [PubMed]
- Qu, L.; Ding, J.; Chen, C.; Wu, Z.J.; Liu, B.; Gao, Y.; Chen, W.; Liu, F.; Sun, W.; Li, X.F.; et al. Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA. Cancer Cell 2016, 29, 653–668. [Google Scholar] [CrossRef]
- Bolukbasi, M.F.; Mizrak, A.; Ozdener, G.B.; Madlener, S.; Strobel, T.; Erkan, E.P.; Fan, J.B.; Breakefield, X.O.; Saydam, O. miR-1289 and “Zipcode”-like Sequence Enrich. mRNAs in Microvesicles. Mol. Ther. Nucleic Acids 2012, 1, e10. [Google Scholar] [CrossRef]
- Hirsova, P.; Ibrahim, S.H.; Krishnan, A.; Verma, V.K.; Bronk, S.F.; Werneburg, N.W.; Charlton, M.R.; Shah, V.H.; Malhi, H.; Gores, G.J. Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. Gastroenterology 2016, 150, 956–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Savina, A.; Furlan, M.; Vidal, M.; Colombo, M.I. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J. Biol. Chem. 2003, 278, 20083–20090. [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. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
Exosome-Mediated Effect | Producing Cell | Specific TDE Content and Mechanism of Action | Reference |
---|---|---|---|
Tumor cell proliferation | Prostate cancer cells | lncRNAc-Myc Upregulated (MYU)-mediated upregulation of c-Myc by competitively binding miR-184 | [82] |
Hypoxic bladder cancer cells | lncRNA-UCA (unknown mechanism) | [119] | |
Hypoxic hepatocellular carcinoma cells | lncRoR-induced hypoxic responses (by downregulation of miR-145 and upregulation of Hypoxia-inducible factor 1 (HIF1) | [120] | |
CAF from Human Oral Tongue Squamous Cell Carcinoma | MFAP5 (Microfibril Associated Protein 5)-induced activation of mitogen-activated protein kinase (MAPK) and AKT | [85] | |
CAF from Hepatocellular carcinoma | MAPK activation by negative regulation of miR-320a | [87] | |
CAF from pancreatic ductal adenocarcinomas | Snail and microRNA-146a upregulation | [125] | |
EMT and metastasis of tumor cells | Bladder cancer cells Colon cancer cell lines | RhoA/ROCK (Rho-associated protein kinase) signaling pathway activation and acquisition of migratory capacity | [79,110] |
Epstein-Barr-Virus EBV infected Nasopharyngeal carcinoma (NPC) | HIF1 upregulation | [78] | |
metastatic melanoma cells | MET induced pro-vasculogenic and metastatic effects | [11] | |
Hypoxic cancer cells | Activation of Epithelial-to-Mesenchymal Transition (EMT) genes in receiving cells | [101] and enclosed references | |
Lung cancer cells | vimentin | [105] | |
Hepatocellular carcinoma cells | MAPK/ERK (Extracellular signal-Regulated Kinase) signalling activation (unknown mechanism) | [106] | |
High aggressive breast cancer | proteins involved in metastasis and invasion | [108] | |
Prostate cancer cells | metalloproteinases induction and targeting of adherens junction proteins | [118] | |
Hypoxic bladder cancer | lncRNA-UCA1 (Urothelial Cancer Associated 1) (mechanism of action unspecified) | [119] | |
Metastatic breast cancer | miR-10b targeting HOXD10 (HomeoboxD10) | [126] | |
CAF from Human Oral Tongue Squamous Cell Carcinoma | MFAP5 activation of MAPK and AKT | [85] | |
CAF from Hepatocellular carcinoma | MAPK activation | [87] | |
CAF from breast cancer | EMT activation by miRs -21, miR-378 and miR-143 | [115] | |
CAF from ovarian cancer | TGF (Transforming Growth Factor) β1-induced EMT | [116] | |
Mesenchymal stem cells | TGFβ1 activation of Smad2/3, Akt/GSK (Glycogen synthase kinase)-3β/β-catenin, NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells), ERK (Extracellular signal-Regulated Kinase), JNK (c-Jun N-terminal kinase and p38 MAPK (mitogen-activated protein kinase) | [117] |
Tumor Microenvironment Modification | Producing Cell | Specific Content and Mechanism of Action | Reference |
---|---|---|---|
CAF activation | Gastric cancer cells | miR-27a-mediated downregulation of CSRP2 (cysteine and glycine rich protein 2) | [81] |
Bladder cancer cells | TGFβ–induced SMAD (small mothers against decapentaplegic) activation | [84] | |
Pancreatic ductal adenocarcinomas | Stellate cells activation and induction of a pro-inflammatory milieu. (unknown mechanism) | [94] | |
Prostate cancer cells | Induction of TGF-β2, TNF1α (Tumor necrosis factor1 α), IL6 (Interleukin 6), TSG101 (Tumor susceptibility gene 101), Akt, ILK1 (Integrin-linked kinase1) and β-catenin. | [118] | |
Angiogenesis and vascular permeability | Cancer Stem Cells from Hepatocellular Carcinoma | lncRNA H19-mediated VEGF (Vascular endothelial growth factor) induction | [31] |
Metastatic breast cancer | miR-105 targeting of ZO-1 | [95] | |
Glioma stem cells | miR21-mediated induction of VEGF pathway. | [97] | |
Hypoxic cancer cells | Upregulation of miR-135-b, miR-23a, miR-210, miR-494 and Wnt pathway activation. | [101] and enclosed references | |
Ovarian cancer | E-cadherin-mediated activation of β-catenin and NFκB (Nuclear Factor kappa-light-chain-enhancer of activated B cells) signaling | [103] | |
Cervical squamous cell carcinoma | miR-221-3p-mediated activation of the ERK (Extracellular signal-Regulated Kinase)/AKT pathway | [122] | |
Immunomodulation | Head and neck squamous cell carcinoma | Receptor–ligand interactions regulating gene expression in T cells | [90] |
Melanoma cells | miR-690 induction of mitochondrial apoptotic pathway in CD4+ T cells | [91] | |
Several cancer cells | miRNAs regulation | [92] and enclosed references | |
Lung adenocarcinoma, hepatocellular carcinoma, breast carcinoma | Monocyte recruitment and Generation of Tumor Associated Macrofages | [104] | |
Hypoxic lung cancer | miR-103a-mediated targeting of PTEN (Phosphatase and tensin homolog) and activation of Tumor Associated Macrofages | [120] | |
Chemoresistence and Cancer Stem Cell stimulation | CAF from colon rectal cancer | Wnt3a induction of WNT signalling activation in CSC (Cancer Stem Cells) | [127] |
CAF from breast cancer | miR-21, miR-378e and mir-143-mediated Cancer Stem Cells maintenance | [113] | |
Renal cell carcinoma | lncRNA ARSR-mediated chemoresistance via competitively binding of miR-34/miR-449. | [128] | |
Metabolism modulation | Melanoma cells | miR-155 and miR-210-mediated promotion of glycolysis and inhibition of oxidative phosphorylation. | [88] |
CAF from prostate cancer and from pancreatic cancer | Metabolites inhibiting mitochondrial oxidative phosphorylation and increasing glycolysis. | [89] |
© 2018 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
Conigliaro, A.; Cicchini, C. Exosome-Mediated Signaling in Epithelial to Mesenchymal Transition and Tumor Progression. J. Clin. Med. 2019, 8, 26. https://doi.org/10.3390/jcm8010026
Conigliaro A, Cicchini C. Exosome-Mediated Signaling in Epithelial to Mesenchymal Transition and Tumor Progression. Journal of Clinical Medicine. 2019; 8(1):26. https://doi.org/10.3390/jcm8010026
Chicago/Turabian StyleConigliaro, Alice, and Carla Cicchini. 2019. "Exosome-Mediated Signaling in Epithelial to Mesenchymal Transition and Tumor Progression" Journal of Clinical Medicine 8, no. 1: 26. https://doi.org/10.3390/jcm8010026
APA StyleConigliaro, A., & Cicchini, C. (2019). Exosome-Mediated Signaling in Epithelial to Mesenchymal Transition and Tumor Progression. Journal of Clinical Medicine, 8(1), 26. https://doi.org/10.3390/jcm8010026