Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases
Abstract
:Simple Summary
Abstract
1. Introduction
2. Pathological Functions of Exosomes
2.1. Tumor Pathogenesis
2.1.1. Cancer Initiation
2.1.2. Tumor Angiogenesis
2.1.3. Tumor Metastasis
2.1.4. Tumor Immunity
2.1.5. Cancer Drug Resistance
Exosome Components | Cancer Type | Cell Function | Induced Mechanism | Reference |
---|---|---|---|---|
miRNA-202-3p | Chronic lymphoblastic leukemia (CLL) | Inhibits cancer initiation | Discarded by tumor cells in extracellular vesicles (EVs) | [61] |
miRNA-19b miRNA-20a | Acute myeloid leukemia (AML) | Multidrug resistance | Transfer of multidrug resistance protein-1 (MRP-1) | [172] |
miRNA-126 | Chronic myelogenous leukemia (CML) | Leukemic stem cell quiescence and leukemia growth | Not defined | [109] |
LncRNA-MONC miRNA 100HG | Acute megacaryobastic leukemia | Tumor growth | Oncogenes | [71] |
miRNA-103a | Lung cancer | Cancer progression and angiogenesis | Decreased phosphatase and tensin homolog (PTEN) and M2 polarization of protumoral macrophages | [64] |
miRNA-21 | Lung cancer | Modulates immunity, promotes angiogenesis | Increase in ligands of long terminal repeats (LTRs) in immune cells, vascular endothelial growth factor (VEGF) levels | [28,55] |
[75,132] | ||||
miRNA-21 | Ovarian cancer | Suppresses apoptosis (drug resistance) | Binding to apoptotic protease activating factor 1 (APAF1) | [175] |
miRNA-21 | Glioblastoma | Priming tumor microenvironment | Microglial cell reprograming | [176] |
miRNA-21 | Esophageal squamous cell carcinoma | Cancer cell migration and invasion | Activator of cancer-associated fibroblasts (CAFs), cancer cell migration | [177] |
miRNA-21 | Breast cancer | Tumor progression | Cancer cell stemness and epithelial-mesenchymal transition (EMT), induction of proinflammatory and pro-tumorigenic monocyte profile | [62,178] |
miRNA-9 | Breast cancer | Promotes angiogenesis tumor metastasis | Janus kinase-signal transducer and activator of transcription (JAK-STAT) activation Induction of CAFs | [78,179] |
miRNA-9-5p miRNA-195-5p miRNA-203a-3p | Breast cancer | Stimulate cancer stem-like line phenotype | Transcription factor one cut homeobox 2 (ONECUT2) | [173] |
miRNA-939 miRNA-105 | Breast cancer | Destruction of endothelial barrier | Downregulation of vascular endothelial (VE)-cadherin, tight junction protein Zonula occludens-1 (ZO-1) | [113,133] |
miRNA-105 | Breast cancer | Tumor growth | CAF mediation of metabolic reprograming | [180] |
miRNA-10b | Breast cancer | Cell invasion | Suppression target genes homeobox D10 (HOXD10) and Kruppel-like factor 4 (KLF4) | [114] |
miRNA-200 miRNA-122 | Breast cancer | Promote metastasis | Mesenchymal-to-epithelial transition (MET) regulation process, glucose metabolism reprogramming | [108,116] |
miRNA-181c | Breast cancer and metastatic brain cancer | BBB destruction Brain metastasis | Downregulation of gene phosphoinositide dependent protein kinase 1 (PDPK1) | [115] |
miRNA-221/222 | Breast cancer | Drug resistance | Reduction in expression of target genes P27 and ERa | [170] |
miRNA-222/223 | Breast cancer | Breast cancer cell dormancy in bone marrow and drug resistance | Not defined | [148] |
miRNA-143 | Breast cancer | Promotion of cancer cell stemness and EMT phenotype | Not defined | [62] |
miRNA-143 | Prostate cancer | Inhibition of cell growth | Induce death signaling between normal and cancer cells | [45] |
miRNA-203 miRNA-212-3p | Pancreatic cancer | Immune dysfunction, immune tolerance | Toll-like receptor 4 (TLR4) regulation, downregulation of regulatory factor X-associated protein (RFXAP) expression | [122,133] |
miRNA-155 | Pancreatic cancer | Chemoresistance Tumor invasion and progression | Promotion of reactive oxygen species (ROS) detoxification Reprograming of normal fibroblasts into CAFs via tumor protein P53 inducible nuclear protein 1 (TP53INP1) | [110,174] |
miRNA-21/155 | Neuroblastoma | Resistance to chemotherapy | Crosstalk with miRNA-21 Activation of toll-like receptor 8/nuclear factor Kappa B (TLR8/NFKB) and telomeric repeat binding factor 1 (TERF1) axis | [171] |
LncRNA DANCR | Glioma | Tumor progression and malignancy | Sponging miRNA-33a-5p | [73] |
miRNA-146a | Multiple myeloma | Tumor cell growth | Increased cytokine and chemokine secretion | [59] |
miRNA-24-3p | Nansopharyngeal carcinoma (NPC) | Tumor growth | Target fibroblast growth factor 11 (FGF11) to suppress T cells | [181] |
Let-7 family | Gastric cancer | Suppression of cancer initiation | Not defined | [182] |
miRNA-15b-3a | Gastric cancer | Tumor progression | Restraining dynein light chain Tctex-type 1 (DYNLT1)/caspase-3/Caspase-9 signaling pathway | [183] |
miRNA-203 | Colorectal cancer | Metastasis | Differentiation of monocytes to M2 tumor-associated macrophages | [63] |
miRNA-210 | Hepatocellular carcinoma | Angiogenesis | Inhibition of Mothers against decapentaplegic homolog 4 (SMAD4) and Signal transducer and activator of transcription 6 (STAT6) secretion by endothelial cells | [109] |
miRNA-103 | Hepatocellular carcinoma | Increase in vascular permeability | Inhibition of VE-cadherin, P120-catenin and zonula occludens 1 expression | [184] |
LncRNA-RoR | Hepatocellular cancer | Tumor growth | Sponge miRNA-145 and promote hypoxia-inducible factor (HIF) | [72] |
LncRNA-HOTAIR | Urotheral bladder cancer | Tumor initiation and progression | Sponge miRNA-205 | [68] |
LncRNA-MALATI | Cervical cancer | Tumor invasion | Modulation of epithelial-to-mesenchymal transition (EMT) | [69,70] |
2.2. Neurodegenerative Disease
3. Exosome Composition
3.1. Nucleic Acids
3.2. Proteins
3.3. Lipids
4. Applications of Exosomes in Biomedicine
4.1. Exosomes as Biomarkers
4.1.1. Exosomes for Cancer Diagnosis
4.1.2. Use of Exosomes for Molecular Diagnostics of Neurodegenerative Diseases
4.2. Use of Exosomes as Therapeutic Agents
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3136. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, B.; Caetano, J.; Barahona, F.; Lopes, R.; Carneiro, E.; Costa-Silva, B.; Joao, C. Liquid biopsies for multiple myeloma in a time of precision medicine. J. Mol. Med. 2020, 98, 513–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef] [PubMed]
- Wong, S.Q.; Dawson, S.J. Combining liquid biopsies and PET-CT for early cancer detection. Nat. Med. 2020, 26, 1010–1011. [Google Scholar] [CrossRef] [PubMed]
- Aheget, H.; Tristán-Manzano, M.; Mazini, L.; Cortijo-Gutierrez, M.; Galindo-Moreno, P.; Herrera, C.; Martin, F.; Marchal, J.A.; Benabdellah, K. Exosome: A new player in translational nanomedicine. J. Clin. Med. 2020, 9, 2380. [Google Scholar] [CrossRef]
- Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
- Tkach, M.; Thery, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
- Santucci, L.; Bruschi, M.; Del Zotto, G.; Antonini, F.; Ghiggeri, G.M.; Panfoli, I.; Candiano, G. Biological surface properties in extracellular vesicles and their effect on cargo proteins. Sci. Rep. 2019, 9, 13048. [Google Scholar] [CrossRef] [Green Version]
- Yanez-Mo, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borras, 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]
- 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. 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]
- 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] [PubMed] [Green Version]
- Valcz, G.; Buzás, E.I.; Kittel, Á.; Krenács, T.; Visnovitz, T.; Spisák, S.; Török, G.; Homolya, L.; Zsigrai, S.; Kiszler, G. En bloc release of MVB-like small extracellular vesicle clusters by colorectal carcinoma cells. J. Extracell. Vesicles 2019, 8, 1596668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Malm, T.; Loppi, S.; Kanninen, K.M. Exosomes in Alzheimer’s disease. Neurochem. Int. 2016, 97, 193–199. [Google Scholar] [CrossRef]
- Li, K.; Chen, Y.; Li, A.; Tan, C.; Liu, X. Exosomes play roles in sequential processes of tumor metastasis. Int. J. Cancer 2019, 144, 1486–1495. [Google Scholar] [CrossRef]
- Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef]
- Nabet, B.Y.; Qiu, Y.; Shabason, J.E.; Wu, T.J.; Yoon, T.; Kim, B.C.; Benci, J.L.; DeMichele, A.M.; Tchou, J.; Marcotrigiano, J. Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer. Cell 2017, 170, 352–366.e313. [Google Scholar] [CrossRef] [Green Version]
- Hill, A.F. Extracellular vesicles and neurodegenerative diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef]
- Jiang, L.; Dong, H.; Cao, H.; Ji, X.; Luan, S.; Liu, J. Exosomes in pathogenesis, diagnosis, and treatment of Alzheimer’s disease. Medical Sci. Monit. 2019, 25, 3329. [Google Scholar] [CrossRef]
- Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Garbis, S.D.; Ntzouni, M.; Margaritis, L.H.; Stefanis, L.; Vekrellis, K. Cell-produced α-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851. [Google Scholar] [CrossRef] [Green Version]
- Fevrier, B.; Vilette, D.; Archer, F.; Loew, D.; Faigle, W.; Vidal, M.; Laude, H.; Raposo, G. Cells release prions in association with exosomes. Proc. Natl. Acad. Sci. USA 2004, 101, 9683–9688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, Y.; Long, J.; Sun, Y.; Li, H.; Jiang, E.; Zeng, C.; Zhu, W. The function and clinical significance of eIF3 in cancer. Gene 2018, 673, 130–133. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-β/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Webber, J.; Steadman, R.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010, 70, 9621–9630. [Google Scholar] [CrossRef] [Green Version]
- Chowdhury, R.; Webber, J.P.; Gurney, M.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015, 6, 715–731. [Google Scholar] [CrossRef]
- Pezzuto, A.; Carico, E. Role of HIF-1 in Cancer Progression: Novel Insights. A Review. Curr. Mol. Med. 2018, 18, 343–351. [Google Scholar] [CrossRef]
- Demory Beckler, M.; Higginbotham, J.N.; Franklin, J.L.; Ham, A.J.; Halvey, P.J.; Imasuen, I.E.; Whitwell, C.; Li, M.; Liebler, D.C.; Coffey, R.J. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Mol. Cell. Proteom. 2013, 12, 343–355. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Li, C.-W.; Chan, L.-C.; Wei, Y.; Hsu, J.-M.; Xia, W.; Cha, J.-H.; Hou, J.; Hsu, J.L.; Sun, L. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018, 28, 862–864. [Google Scholar] [CrossRef] [Green Version]
- Lancaster, G.I.; Febbraio, M.A. Exosome-Dependent trafficking of HSP70 A novel secretory pathway for cellular stress proteins. J. Biol. Chem. 2005, 280, 23349–23355. [Google Scholar] [CrossRef] [Green Version]
- Clayton, A.; Turkes, A.; Navabi, H.; Mason, M.D.; Tabi, Z. Induction of heat shock proteins in B-cell exosomes. J. Cell Sci. 2005, 118, 3631–3638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, J.A.; Lee, Y.S.; Kim, S.H.; Ko, J.K.; Kim, C.W. MHC independent anti-tumor immune responses induced by Hsp70-enriched exosomes generate tumor regression in murine models. Cancer Lett. 2009, 275, 256–265. [Google Scholar] [CrossRef] [PubMed]
- Mukhopadhyay, U.K.; Mak, A.S. p53: Is the guardian of the genome also a suppressor of cell invasion? Cell Cycle 2009, 8, 2481. [Google Scholar] [CrossRef] [PubMed]
- Muller, P.A.; Vousden, K.H. p53 mutations in cancer. Nat. Cell Biol. 2013, 15, 2–8. [Google Scholar] [CrossRef]
- Yu, X.; Riley, T.; Levine, A.J. The regulation of the endosomal compartment by p53 the tumor suppressor gene. FEBS J. 2009, 276, 2201–2212. [Google Scholar] [CrossRef]
- Lespagnol, A.; Duflaut, D.; Beekman, C.; Blanc, L.; Fiucci, G.; Marine, J.C.; Vidal, M.; Amson, R.; Telerman, A. Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 2008, 15, 1723–1733. [Google Scholar] [CrossRef] [Green Version]
- Putz, U.; Howitt, J.; Doan, A.; Goh, C.P.; Low, L.H.; Silke, J.; Tan, S.S. The tumor suppressor PTEN is exported in exosomes and has phosphatase activity in recipient cells. Sci. Signal. 2012, 5, ra70. [Google Scholar] [CrossRef]
- Ristorcelli, E.; Beraud, E.; Mathieu, S.; Lombardo, D.; Verine, A. Essential role of Notch signaling in apoptosis of human pancreatic tumoral cells mediated by exosomal nanoparticles. Int. J. Cancer 2009, 125, 1016–1026. [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, 715–725e714. [Google Scholar] [CrossRef] [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.; Lehtio, 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] [Green Version]
- Montecalvo, A.; Larregina, A.T.; Shufesky, W.J.; Stolz, D.B.; Sullivan, M.L.; Karlsson, J.M.; Baty, C.J.; Gibson, G.A.; Erdos, G.; Wang, Z.; et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 2011, 119, 756–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bronisz, A.; Godlewski, J.; Chiocca, E.A. Extracellular Vesicles and MicroRNAs: Their Role in Tumorigenicity and Therapy for Brain Tumors. Cell Mol. Neurobiol. 2016, 36, 361–376. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kosaka, M.; Kang, M.R.; Yang, G.; Li, L.C. Targeted p21WAF1/CIP1 activation by RNAa inhibits hepatocellular carcinoma cells. Nucleic Acid Ther. 2012, 22, 335–343. [Google Scholar] [CrossRef] [Green Version]
- Che, Y.; Shi, X.; Shi, Y.; Jiang, X.; Ai, Q.; Shi, Y.; Gong, F.; Jiang, W. Exosomes derived from miR-143-overexpressing MSCs inhibit cell migration and invasion in human prostate cancer by downregulating TFF3. Mol. Ther. Nucleic Acids 2019, 18, 232–244. [Google Scholar] [CrossRef] [Green Version]
- Ahadi, A.; Brennan, S.; Kennedy, P.J.; Hutvagner, G.; Tran, N. Long non-coding RNAs harboring miRNA seed regions are enriched in prostate cancer exosomes. Sci. Rep. 2016, 6, 24922. [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]
- 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]
- Deng, J.; Galipeau, J. Reprogramming of B cells into regulatory cells with engineered fusokines. Infect. Disord. Drug Targets 2012, 12, 248–254. [Google Scholar] [CrossRef] [Green Version]
- Dong, H.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.; Tamada, K.; et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800. [Google Scholar] [CrossRef]
- Gordon, S.R.; Maute, R.L.; Dulken, B.W.; Hutter, G.; George, B.M.; McCracken, M.N.; Gupta, R.; Tsai, J.M.; Sinha, R.; Corey, D.; et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017, 545, 495–499. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Huang, L.; Guan, X.; Li, H.; Zhang, Q.Q.; Han, C.; Wang, Y.J.; Wang, C.; Zhang, Y.; Qu, C.; et al. ER-α36, a novel variant of ERα, is involved in the regulation of Tamoxifen-sensitivity of glioblastoma cells. Steroids 2016, 111, 127–133. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, S.; Floros, T.; Theodoraki, M.N.; Hong, C.S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pasquier, J.; Thawadi, H.A.; Ghiabi, P.; Abu-Kaoud, N.; Maleki, M.; Guerrouahen, B.S.; Vidal, F.; Courderc, B.; Ferron, G.; Martinez, A.; et al. Microparticles mediated cross-talk between tumoral and endothelial cells promote the constitution of a pro-metastatic vascular niche through Arf6 up regulation. Cancer Microenviron. 2014, 7, 41–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horie, K.; Kawakami, K.; Fujita, Y.; Sugaya, M.; Kameyama, K.; Mizutani, K.; Deguchi, T.; Ito, M. Exosomes expressing carbonic anhydrase 9 promote angiogenesis. Biochem. Biophys. Res. Commun. 2017, 492, 356–361. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Wang, S.; Zhu, R.; Li, H.; Han, Q.; Zhao, R.C. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J. Hematol. Oncol. 2016, 9, 42. [Google Scholar] [CrossRef] [Green Version]
- De Veirman, K.; Wang, J.; Xu, S.; Leleu, X.; Himpe, E.; Maes, K.; De Bruyne, E.; Van Valckenborgh, E.; Vanderkerken, K.; Menu, E.; et al. Induction of miR-146a by multiple myeloma cells in mesenchymal stromal cells stimulates their pro-tumoral activity. Cancer Lett. 2016, 377, 17–24. [Google Scholar] [CrossRef]
- Dhondt, B.; Rousseau, Q.; De Wever, O.; Hendrix, A. Function of extracellular vesicle-associated miRNAs in metastasis. Cell Tissue Res. 2016, 365, 621–641. [Google Scholar] [CrossRef]
- 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, e0141429. [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] [PubMed] [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] [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]
- Huarte, M.; Guttman, M.; Feldser, D.; Garber, M.; Koziol, M.J.; Kenzelmann-Broz, D.; Khalil, A.M.; Zuk, O.; Amit, I.; Rabani, M.; et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 2010, 142, 409–419. [Google Scholar] [CrossRef] [Green Version]
- Hewson, C.; Morris, K.V. Form and Function of Exosome-Associated Long Non-coding RNAs in Cancer. Curr. Top. Microbiol. Immunol. 2016, 394, 41–56. [Google Scholar]
- Kino, T.; Hurt, D.E.; Ichijo, T.; Nader, N.; Chrousos, G.P. Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci. Signal. 2010, 3, ra8. [Google Scholar] [CrossRef] [Green Version]
- Sun, X.; Du, P.; Yuan, W.; Du, Z.; Yu, M.; Yu, X.; Hu, T. Long non-coding RNA HOTAIR regulates cyclin J via inhibition of microRNA-205 expression in bladder cancer. Cell Death Dis. 2015, 6, e1907. [Google Scholar] [CrossRef] [Green Version]
- Sun, R.; Qin, C.; Jiang, B.; Fang, S.; Pan, X.; Peng, L.; Liu, Z.; Li, W.; Li, Y.; Li, G. Down-Regulation of MALAT1 inhibits cervical cancer cell invasion and metastasis by inhibition of epithelial-mesenchymal transition. Mol. Biosyst. 2016, 12, 952–962. [Google Scholar] [CrossRef]
- Yang, L.; Bai, H.S.; Deng, Y.; Fan, L. High MALAT1 expression predicts a poor prognosis of cervical cancer and promotes cancer cell growth and invasion. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3187–3193. [Google Scholar]
- Emmrich, S.; Streltsov, A.; Schmidt, F.; Thangapandi, V.R.; Reinhardt, D.; Klusmann, J.H. LincRNAs MONC and MIR100HG act as oncogenes in acute megakaryoblastic leukemia. Mol. Cancer 2014, 13, 171. [Google Scholar] [CrossRef] [Green Version]
- Nath, B.; Szabo, G. Hypoxia and hypoxia inducible factors: Diverse roles in liver diseases. Hepatology 2011, 55, 622–633. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.X.; Sun, Y.; Gao, L.; Meng, Q.; Yang, B.Y. Long non-coding RNA DANCR facilitates glioma malignancy by sponging miR-33a-5p. Neoplasma 2018, 65, 790–798. [Google Scholar] [CrossRef] [PubMed]
- Bolat, F.; Kayaselcuk, F.; Nursal, T.Z.; Yagmurdur, M.C.; Bal, N.; Demirhan, B. Microvessel density, VEGF expression, and tumor-associated macrophages in breast tumors: Correlations with prognostic parameters. J. Exp. Clin. Cancer Res. 2006, 25, 365–372. [Google Scholar]
- Liu, Y.; Luo, F.; Wang, B.; Li, H.; Xu, Y.; Liu, X.; Shi, L.; Lu, X.; Xu, W.; Lu, L.; et al. STAT3-Regulated exosomal miR-21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 2015, 370, 125–135. [Google Scholar] [CrossRef] [PubMed]
- Leong, H.; Mathur, P.S.; Greene, G.L. Green tea catechins inhibit angiogenesis through suppression of STAT3 activation. Breast Cancer Res. Treat. 2009, 117, 505–515. [Google Scholar] [CrossRef]
- Dong, Y.; Lu, B.; Zhang, X.; Zhang, J.; Lai, L.; Li, D.; Wu, Y.; Song, Y.; Luo, J.; Pang, X.; et al. Cucurbitacin E, a tetracyclic triterpenes compound from Chinese medicine, inhibits tumor angiogenesis through VEGFR2-mediated Jak2-STAT3 signaling pathway. Carcinogenesis 2010, 31, 2097–2104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, G.; Wu, X.; Jiang, Z.; Kasman, I.; Yao, J.; Guan, Y.; Oeh, J.; Modrusan, Z.; Bais, C.; Sampath, D.; et al. Tumour-Secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012, 31, 3513–3523. [Google Scholar] [CrossRef] [PubMed]
- 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, 3748–3757. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Paget, S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989, 8, 98–101. [Google Scholar] [CrossRef] [Green Version]
- Akhtar, M.; Haider, A.; Rashid, S.; Al-Nabet, A. Paget’s “Seed and Soil” Theory of Cancer Metastasis: An Idea Whose Time has Come. Adv. Anatom. Pathol. 2019, 26, 69–74. [Google Scholar] [CrossRef] [PubMed]
- Chaffer, C.L.; Morris, M.J. The feeding response to melanin-concentrating hormone is attenuated by antagonism of the NPY Y(1)-receptor in the rat. Endocrinology 2002, 143, 191–197. [Google Scholar] [CrossRef] [PubMed]
- Ocana, O.H.; Corcoles, R.; Fabra, A.; Moreno-Bueno, G.; Acloque, H.; Vega, S.; Barrallo-Gimeno, A.; Cano, A.; Nieto, M.A. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 2012, 22, 709–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Cao, X. Immunosuppressive cells in tumor immune escape and metastasis. J. Mol. Med. 2016, 94, 509–522. [Google Scholar] [CrossRef]
- Bergfeld, S.A.; Blavier, L.; DeClerck, Y.A. Bone marrow-derived mesenchymal stromal cells promote survival and drug resistance in tumor cells. Mol. Cancer Ther. 2014, 13, 962–975. [Google Scholar] [CrossRef] [Green Version]
- Camorani, S.; Hill, B.S.; Fontanella, R.; Greco, A.; Gramanzini, M.; Auletta, L.; Gargiulo, S.; Albanese, S.; Lucarelli, E.; Cerchia, L.; et al. Inhibition of Bone Marrow-Derived Mesenchymal Stem Cells Homing Towards Triple-Negative Breast Cancer Microenvironment Using an Anti-PDGFRβ Aptamer. Theranostics 2017, 7, 3595–3607. [Google Scholar] [CrossRef]
- Luo, J.; Ok Lee, S.; Liang, L.; Huang, C.K.; Li, L.; Wen, S.; Chang, C. Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene 2014, 33, 2768–2778. [Google Scholar] [CrossRef] [Green Version]
- Cui, T.X.; Kryczek, I.; Zhao, L.; Zhao, E.; Kuick, R.; Roh, M.H.; Vatan, L.; Szeliga, W.; Mao, Y.; Thomas, D.G.; et al. Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity 2013, 39, 611–621. [Google Scholar] [CrossRef] [Green Version]
- Weyemi, U.; Redon, C.E.; Sethi, T.K.; Burrell, A.S.; Jailwala, P.; Kasoji, M.; Abrams, N.; Merchant, A.; Bonner, W.M. Twist1 and Slug mediate H2AX-regulated epithelial-mesenchymal transition in breast cells. Cell Cycle 2016, 15, 2398–2404. [Google Scholar] [CrossRef] [Green Version]
- Brabletz, T.; Hlubek, F.; Spaderna, S.; Schmalhofer, O.; Hiendlmeyer, E.; Jung, A.; Kirchner, T. Invasion and metastasis in colorectal cancer: Epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and β-catenin. Cells Tissues Organs 2005, 179, 56–65. [Google Scholar] [CrossRef]
- Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
- Okuda, H.; Kobayashi, A.; Xia, B.; Watabe, M.; Pai, S.K.; Hirota, S.; Xing, F.; Liu, W.; Pandey, P.R.; Fukuda, K.; et al. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells. Cancer Res. 2012, 72, 537–547. [Google Scholar] [CrossRef] [Green Version]
- Lock, R.; Kenific, C.M.; Leidal, A.M.; Salas, E.; Debnath, J. Autophagy-Dependent production of secreted factors facilitates oncogenic RAS-driven invasion. Cancer Discov. 2014, 4, 466–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, Y.F.; Shi, Y.H.; Shen, Y.H.; Ding, Z.B.; Ke, A.W.; Zhou, J.; Qiu, S.J.; Fan, J. Promoting colonization in metastatic HCC cells by modulation of autophagy. PLoS ONE 2013, 8, e74407. [Google Scholar] [CrossRef] [PubMed]
- Zhu, H.; Wang, D.; Zhang, L.; Xie, X.; Wu, Y.; Liu, Y.; Shao, G.; Su, Z. Upregulation of autophagy by hypoxia-inducible factor-1α promotes EMT and metastatic ability of CD133+ pancreatic cancer stem-like cells during intermittent hypoxia. Oncol. Rep. 2014, 32, 935–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellodi, C.; Lidonnici, M.R.; Hamilton, A.; Helgason, G.V.; Soliera, A.R.; Ronchetti, M.; Galavotti, S.; Young, K.W.; Selmi, T.; Yacobi, R.; et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J. Clin. Investig. 2009, 119, 1109–1123. [Google Scholar] [CrossRef]
- Zhu, H.; Wang, D.; Liu, Y.; Su, Z.; Zhang, L.; Chen, F.; Zhou, Y.; Wu, Y.; Yu, M.; Zhang, Z.; et al. Role of the Hypoxia-inducible factor-1 α induced autophagy in the conversion of non-stem pancreatic cancer cells into CD133+ pancreatic cancer stem-like cells. Cancer Cell Int. 2013, 13, 119. [Google Scholar] [CrossRef] [Green Version]
- Husemann, Y.; Klein, C.A. The analysis of metastasis in transgenic mouse models. Transgenic Res. 2009, 18, 1–5. [Google Scholar] [CrossRef]
- Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Rev. Cancer 2009, 9, 239–252. [Google Scholar] [CrossRef]
- Pollard, J.W. Trophic macrophages in development and disease. Nat. Rev. Immunol. 2009, 9, 259–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Yao, Y.; Gong, C.; Yu, F.; Su, S.; Liu, B.; Deng, H.; Wang, F.; Lin, L.; Yao, H.; et al. CCL18 from tumor-associated macrophages promotes breast cancer metastasis via PITPNM3. Cancer Cell 2011, 19, 541–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, M.; Chen, J.; Su, F.; Yu, B.; Lin, L.; Liu, Y.; Huang, J.D.; Song, E. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 2011, 10, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mazzone, M.; Dettori, D.; de Oliveira, R.L.; Loges, S.; Schmidt, T.; Jonckx, B.; Tian, Y.M.; Lanahan, A.A.; Pollard, P.; de Almodovar, C.R.; et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 2009, 136, 839–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef] [PubMed]
- Tsai, J.H.; Yang, J. Epithelial-Mesenchymal plasticity in carcinoma metastasis. Genes Dev. 2013, 27, 2192–2206. [Google Scholar] [CrossRef] [Green Version]
- Park, S.M.; Gaur, A.B.; Lengyel, E.; Peter, M.E. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22, 894–907. [Google Scholar] [CrossRef] [Green Version]
- Le, M.T.; Hamar, P.; Guo, C.; Basar, E.; Perdigao-Henriques, R.; Balaj, L.; Lieberman, J. miR-200-containing extracellular vesicles promote breast cancer cell metastasis. J. Clin. Investig. 2014, 124, 5109–5128. [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]
- 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]
- Zomer, A.; Maynard, C.; Verweij, F.J.; Kamermans, A.; Schafer, 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] [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. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Modica, M.; Regondi, V.; Sandri, M.; Iorio, M.V.; Zanetti, A.; Tagliabue, E.; Casalini, P.; Triulzi, T. Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett. 2016, 384, 94–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lotvall, 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] [Green Version]
- Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.; 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]
- Wu, X.; Somlo, G.; Yu, Y.; Palomares, M.R.; Li, A.X.; Zhou, W.; Chow, A.; Yen, Y.; Rossi, J.J.; Gao, H.; et al. De novo sequencing of circulating miRNAs identifies novel markers predicting clinical outcome of locally advanced breast cancer. J. Transl. Med. 2012, 10, 42. [Google Scholar] [CrossRef] [Green Version]
- Thery, C.; Duban, L.; Segura, E.; Veron, P.; Lantz, O.; Amigorena, S. Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat. Immunol. 2002, 3, 1156–1162. [Google Scholar] [CrossRef]
- Clayton, A.; Mason, M.D. Exosomes in tumour immunity. Curr. Oncol. 2009, 16, 46–49. [Google Scholar] [CrossRef]
- Greening, D.W.; Gopal, S.K.; Xu, R.; Simpson, R.J.; Chen, W. Exosomes and their roles in immune regulation and cancer. Semin. Cell Dev. Biol. 2015, 40, 72–81. [Google Scholar] [CrossRef]
- Yu, S.; Liu, C.; Su, K.; Wang, J.; Liu, Y.; Zhang, L.; Li, C.; Cong, Y.; Kimberly, R.; Grizzle, W.E.; et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J. Immunol. 2007, 178, 6867–6875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, G.; Zhou, L.; Qian, Y.; Fu, M.; Chen, 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] [PubMed] [Green Version]
- Andreola, G.; Rivoltini, L.; Castelli, C.; Huber, V.; Perego, P.; Deho, P.; Squarcina, P.; Accornero, P.; Lozupone, F.; Lugini, L.; et al. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J. Exp. Med. 2002, 195, 1303–1316. [Google Scholar] [CrossRef]
- Clayton, A.; Mitchell, J.P.; Court, J.; Linnane, S.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes down-modulate NKG2D expression. J. Immunol. 2008, 180, 7249–7258. [Google Scholar] [CrossRef] [Green Version]
- Clayton, A.; Mitchell, J.P.; Court, J.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer Res. 2007, 67, 7458–7466. [Google Scholar] [CrossRef] [Green Version]
- Yin, Y.; Cai, X.; Chen, X.; Liang, H.; Zhang, Y.; Li, J.; Wang, Z.; Zhang, W.; Yokoyama, S.; Wang, C.; 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]
- Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef]
- Poggio, M.; Hu, T.; Pai, C.C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-Tumor Immunity and Memory. Cell 2019, 177, 414–427.e413. [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]
- Theodoraki, M.N.; Yerneni, S.S.; Hoffmann, T.K.; Gooding, W.E.; Whiteside, T.L. Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin. Cancer Res. 2018, 24, 896–905. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Han, X. Anti-PD-1/PD-L1 therapy of human cancer: Past, present, and future. J. Clin. Investig. 2015, 125, 3384–3391. [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, E2110–E2116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Andre, F.; Schartz, N.E.; Chaput, N.; Flament, C.; Raposo, G.; Amigorena, S.; Angevin, E.; Zitvogel, L. Tumor-Derived exosomes: A new source of tumor rejection antigens. Vaccine 2002, 20, A28–A31. [Google Scholar] [CrossRef]
- Skokos, D.; Botros, H.G.; Demeure, C.; Morin, J.; Peronet, R.; Birkenmeier, G.; Boudaly, S.; Mecheri, S. Mast cell-derived exosomes induce phenotypic and functional maturation of dendritic cells and elicit specific immune responses in vivo. J. Immunol. 2003, 170, 3037–3045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, I.; Nabet, B.Y. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol. Cancer 2019, 18, 32. [Google Scholar] [CrossRef] [PubMed]
- Azmi, A.S.; Bao, B.; Sarkar, F.H. Exosomes in cancer development, metastasis, and drug resistance: A comprehensive review. Cancer Metastasis Rev. 2013, 32, 623–642. [Google Scholar] [CrossRef] [Green Version]
- Valcz, G.; Buzas, E.I.; Sebestyen, A.; Krenacs, T.; Szallasi, Z.; Igaz, P.; Molnar, B. Extracellular Vesicle-Based Communication May Contribute to the Co-Evolution of Cancer Stem Cells and Cancer-Associated Fibroblasts in Anti-Cancer Therapy. Cancers 2020, 12, 2324. [Google Scholar] [CrossRef]
- Steinbichler, T.B.; Dudas, J.; Skvortsov, S.; Ganswindt, U.; Riechelmann, H.; Skvortsova, I.I. Therapy resistance mediated by exosomes. Mol. Cancer 2019, 18, 58. [Google Scholar] [CrossRef]
- Moitra, K.; Lou, H.; Dean, M. Multidrug efflux pumps and cancer stem cells: Insights into multidrug resistance and therapeutic development. Clin. Pharmacol. Ther. 2011, 89, 491–502. [Google Scholar] [CrossRef]
- Januchowski, R.; Sterzynska, K.; Zaorska, K.; Sosinska, P.; Klejewski, A.; Brazert, M.; Nowicki, M.; Zabel, M. Analysis of MDR genes expression and cross-resistance in eight drug resistant ovarian cancer cell lines. J. Ovarian Res. 2016, 9, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bebawy, M.; Combes, V.; Lee, E.; Jaiswal, R.; Gong, J.; Bonhoure, A.; Grau, G.E. Membrane microparticles mediate transfer of P-glycoprotein to drug sensitive cancer cells. Leukemia 2009, 23, 1643–1649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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, e50999. [Google Scholar] [CrossRef] [PubMed]
- Sousa, D.; Lima, R.T.; Vasconcelos, M.H. Intercellular Transfer of Cancer Drug Resistance Traits by Extracellular Vesicles. Trends Mol. Med. 2015, 21, 595–608. [Google Scholar] [CrossRef]
- Gong, J.; Luk, F.; Jaiswal, R.; George, A.M.; Grau, G.E.; Bebawy, M. Microparticle drug sequestration provides a parallel pathway in the acquisition of cancer drug resistance. Eur. J. Pharmacol. 2013, 721, 116–125. [Google Scholar] [CrossRef]
- Federici, C.; Petrucci, F.; Caimi, S.; Cesolini, A.; Logozzi, M.; Borghi, M.; D’Ilio, S.; Lugini, L.; Violante, N.; Azzarito, T.; et al. Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS ONE 2014, 9, e88193. [Google Scholar] [CrossRef] [Green Version]
- Aung, T.; Chapuy, B.; Vogel, D.; Wenzel, D.; Oppermann, M.; Lahmann, M.; Weinhage, T.; Menck, K.; Hupfeld, T.; Koch, R.; et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 2011, 108, 15336–15341. [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. 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]
- Akhter, M.Z.; Sharawat, S.K.; Kumar, V.; Kochat, V.; Equbal, Z.; Ramakrishnan, M.; Kumar, U.; Mathur, S.; Kumar, L.; Mukhopadhyay, A. Aggressive serous epithelial ovarian cancer is potentially propagated by EpCAM(+)CD45(+) phenotype. Oncogene 2018, 37, 2089–2103. [Google Scholar] [CrossRef]
- Ji, R.; Zhang, B.; Zhang, X.; Xue, J.; Yuan, X.; Yan, Y.; Wang, M.; Zhu, W.; Qian, H.; Xu, W. Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle 2015, 14, 2473–2483. [Google Scholar] [CrossRef] [Green Version]
- Boing, A.N.; Stap, J.; Hau, C.M.; Afink, G.B.; Ris-Stalpers, C.; Reits, E.A.; Sturk, A.; van Noorden, C.J.; Nieuwland, R. Active caspase-3 is removed from cells by release of caspase-3-enriched vesicles. Biochim. Biophys. Acta 2013, 1833, 1844–1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Zhang, X.; Wei, P.; Zhang, J.; Niu, Y.; Kang, N.; Zhang, Y.; Zhang, W.; Xing, N. Livin, Survivin and Caspase 3 as early recurrence markers in non-muscle-invasive bladder cancer. World J. Urol. 2014, 32, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
- Manier, S.; Sacco, A.; Leleu, X.; Ghobrial, I.M.; Roccaro, A.M. Bone marrow microenvironment in multiple myeloma progression. J. Biomed. Biotechnol. 2012, 2012, 157496. [Google Scholar] [CrossRef]
- Xu, F.H.; Sharma, S.; Gardner, A.; Tu, Y.; Raitano, A.; Sawyers, C.; Lichtenstein, A. Interleukin-6-induced inhibition of multiple myeloma cell apoptosis: Support for the hypothesis that protection is mediated via inhibition of the JNK/SAPK pathway. Blood 1998, 92, 241–251. [Google Scholar] [CrossRef] [PubMed]
- Rodrigues, C.F.D.; Serrano, E.; Patricio, M.I.; Val, M.M.; Albuquerque, P.; Fonseca, J.; Gomes, C.M.F.; Abrunhosa, A.J.; Paiva, A.; Carvalho, L.; et al. Stroma-Derived IL-6, G-CSF and Activin-A mediated dedifferentiation of lung carcinoma cells into cancer stem cells. Sci. Rep. 2018, 8, 11573. [Google Scholar] [CrossRef] [PubMed]
- Hazawa, M.; Tomiyama, K.; Saotome-Nakamura, A.; Obara, C.; Yasuda, T.; Gotoh, T.; Tanaka, I.; Yakumaru, H.; Ishihara, H.; Tajima, K. Radiation increases the cellular uptake of exosomes through CD29/CD81 complex formation. Biochem. Biophys. Res. Commun. 2014, 446, 1165–1171. [Google Scholar] [CrossRef] [PubMed]
- Dutta, S.; Warshall, C.; Bandyopadhyay, C.; Dutta, D.; Chandran, B. Interactions between exosomes from breast cancer cells and primary mammary epithelial cells leads to generation of reactive oxygen species which induce DNA damage response, stabilization of p53 and autophagy in epithelial cells. PLoS ONE 2014, 9, e97580. [Google Scholar] [CrossRef] [Green Version]
- Mutschelknaus, L.; Peters, C.; Winkler, K.; Yentrapalli, R.; Heider, T.; Atkinson, M.J.; Moertl, S. Exosomes Derived from Squamous Head and Neck Cancer Promote Cell Survival after Ionizing Radiation. PLoS ONE 2016, 11, e0152213. [Google Scholar] [CrossRef]
- Arscott, W.T.; Tandle, A.T.; Zhao, S.; Shabason, J.E.; Gordon, I.K.; Schlaff, C.D.; Zhang, G.; Tofilon, P.J.; Camphausen, K.A. Ionizing radiation and glioblastoma exosomes: Implications in tumor biology and cell migration. Transl. Oncol. 2013, 6, 638–648. [Google Scholar] [CrossRef] [Green Version]
- Peiris-Pages, M.; Sotgia, F.; Lisanti, M.P. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget 2015, 6, 10728–10745. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Tang, Y.; Tan, Y.; Wei, Q.; Yu, W. Cancer-Associated fibroblasts in radiotherapy: Challenges and new opportunities. Cell Commun. Signal. 2019, 17, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McMillin, D.W.; Negri, J.M.; Mitsiades, C.S. The role of tumour-stromal interactions in modifying drug response: Challenges and opportunities. Nat. Rev. Drug Discov. 2013, 12, 217–228. [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] [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]
- Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10(+)GPR77(+) Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018, 172, 841–856.e816. [Google Scholar] [CrossRef]
- Mitra, A.; Mishra, L.; Li, S. EMT, CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget 2015, 6, 10697–10711. [Google Scholar] [CrossRef] [Green Version]
- Steinbichler, T.B.; Dudas, J.; Skvortsov, S.; Ganswindt, U.; Riechelmann, H.; Skvortsova, I.I. Therapy resistance mediated by cancer stem cells. Semin. Cancer Biol. 2018, 53, 156–167. [Google Scholar] [CrossRef]
- Johansson, J.; Landgren, M.; Fernell, E.; Lewander, T.; Venizelos, N. Decreased binding capacity (Bmax) of muscarinic acetylcholine receptors in fibroblasts from boys with attention-deficit/hyperactivity disorder (ADHD). Atten. Defic. Hyperact. Disord. 2013, 5, 267–271. [Google Scholar] [CrossRef] [Green Version]
- Grinde, M.T.; Vik, J.; Camilio, K.A.; Martinez-Zubiaurre, I.; Hellevik, T. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci. Rep. 2017, 7, 46714. [Google Scholar] [CrossRef] [Green Version]
- Wei, Y.; Lai, X.; Yu, S.; Chen, S.; Ma, Y.; Zhang, Y.; Li, H.; Zhu, X.; Yao, L.; Zhang, J. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res. Treat. 2014, 147, 423–431. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bouvy, C.; Wannez, A.; Laloy, J.; Chatelain, C.; Dogne, J.M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leukemia Res. 2017, 62, 70–76. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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] [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]
- Abels, E.R.; Maas, S.L.N.; Nieland, L.; Wei, Z.; Cheah, P.S.; Tai, E.; Kolsteeg, C.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.e3107. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Momen-Heravi, F.; Bala, S. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogram monocytes via NF-κB pathway. Oncotarget 2018, 9, 34838–34854. [Google Scholar] [CrossRef] [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]
- Yan, W.; Wu, X.; Zhou, W.; Fong, M.Y.; Cao, M.; Liu, J.; Liu, X.; Chen, C.H.; Fadare, O.; Pizzo, D.P.; et al. Cancer-Cell-Secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat. Cell Biol. 2018, 20, 597–609. [Google Scholar] [CrossRef] [Green Version]
- 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, C.; 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]
- Ohshima, K.; Inoue, K.; Fujiwara, A.; Hatakeyama, K.; Kanto, K.; Watanabe, Y.; Muramatsu, K.; Fukuda, Y.; Ogura, S.-I.; Yamaguchi, K. Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS ONE 2010, 5, e13247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, S.; Peng, L.; Yang, J.; Sang, H.; Jin, D.; Li, X.; Chen, M.; Zhang, W.; Dang, Y.; Zhang, G. Exosomal transfer of miR-15b-3p enhances tumorigenesis and malignant transformation through the DYNLT1/Caspase-3/Caspase-9 signaling pathway in gastric cancer. J. Exp. Clin. Cancer Res. 2020, 39, 32. [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] [Green Version]
- Fruhbeis, C.; Frohlich, D.; Kramer-Albers, E.M. Emerging roles of exosomes in neuron-glia communication. Front. Physiol. 2012, 3, 119. [Google Scholar] [CrossRef] [Green Version]
- Howitt, J.; Hill, A.F. Exosomes in the Pathology of Neurodegenerative Diseases. J. Biol. Chem. 2016, 291, 26589–26597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Anca, M.; Fenoglio, C.; Serpente, M.; Arosio, B.; Cesari, M.; Scarpini, E.A.; Galimberti, D. Exosome Determinants of Physiological Aging and Age-Related Neurodegenerative Diseases. Front. Aging Neurosci. 2019, 11, 232. [Google Scholar] [CrossRef] [Green Version]
- Ridder, K.; Keller, S.; Dams, M.; Rupp, A.K.; Schlaudraff, J.; Del Turco, D.; Starmann, J.; Macas, J.; Karpova, D.; Devraj, K.; et al. Extracellular vesicle-mediated transfer of genetic information between the hematopoietic system and the brain in response to inflammation. PLoS Biol. 2014, 12, e1001874. [Google Scholar] [CrossRef]
- Perez, M.; Avila, J.; Hernandez, F. Propagation of Tau via Extracellular Vesicles. Front. Neurosci. 2019, 13, 698. [Google Scholar] [CrossRef] [PubMed]
- Guix, F.X.; Corbett, G.T.; Cha, D.J.; Mustapic, M.; Liu, W.; Mengel, D.; Chen, Z.; Aikawa, E.; Young-Pearse, T.; Kapogiannis, D.; et al. Detection of Aggregation-Competent Tau in Neuron-Derived Extracellular Vesicles. Int. J. Mol. Sci. 2018, 19, 663. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Balaji, V.; Kaniyappan, S.; Kruger, L.; Irsen, S.; Tepper, K.; Chandupatla, R.; Maetzler, W.; Schneider, A.; Mandelkow, E.; et al. The release and trans-synaptic transmission of Tau via exosomes. Mol. Neurodegener. 2017, 12, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Polanco, J.C.; Li, C.; Durisic, N.; Sullivan, R.; Gotz, J. Exosomes taken up by neurons hijack the endosomal pathway to spread to interconnected neurons. Acta Neuropathologica Commun. 2018, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Querfurth, H.W.; LaFerla, F.M. Alzheimer’s disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saman, S.; Kim, W.; Raya, M.; Visnick, Y.; Miro, S.; Saman, S.; Jackson, B.; McKee, A.C.; Alvarez, V.E.; Lee, N.C. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J. Biol. Chem. 2012, 287, 3842–3849. [Google Scholar] [CrossRef] [Green Version]
- Rajendran, L.; Honsho, M.; Zahn, T.R.; Keller, P.; Geiger, K.D.; Verkade, P.; Simons, K. Alzheimer’s disease β-amyloid peptides are released in association with exosomes. Proc. Natl. Acad. Sci. USA 2006, 103, 11172–11177. [Google Scholar] [CrossRef] [Green Version]
- Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-Modulated exosome secretion promotes clearance of amyloid-beta by microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef] [Green Version]
- Alvarez-Erviti, L.; Seow, Y.; Schapira, A.H.; Gardiner, C.; Sargent, I.L.; Wood, M.J.; Cooper, J.M. Lysosomal dysfunction increases exosome-mediated α-synuclein release and transmission. Neurobiol. Dis. 2011, 42, 360–367. [Google Scholar] [CrossRef] [Green Version]
- Sundelof, J.; Giedraitis, V.; Irizarry, M.C.; Sundstrom, J.; Ingelsson, E.; Ronnemaa, E.; Arnlov, J.; Gunnarsson, M.D.; Hyman, B.T.; Basun, H.; et al. Plasma β amyloid and the risk of Alzheimer disease and dementia in elderly men: A prospective, population-based cohort study. Arch. Neurol. 2008, 65, 256–263. [Google Scholar] [CrossRef] [Green Version]
- Ghidoni, R.; Paterlini, A.; Albertini, V.; Stoppani, E.; Binetti, G.; Fuxe, K.; Benussi, L.; Agnati, L.F. A window into the heterogeneity of human cerebrospinal fluid Aβ peptides. J. Biomed. Biotechnol. 2011, 2011, 697036. [Google Scholar] [CrossRef] [Green Version]
- Fuller, S.; Steele, M.; Munch, G. Activated astroglia during chronic inflammation in Alzheimer’s disease—Do they neglect their neurosupportive roles? Mutat. Res. 2009, 690, 40–49. [Google Scholar] [CrossRef]
- Kobayashi, K.; Hernandez, L.D.; Galan, J.E.; Janeway, C.A., Jr.; Medzhitov, R.; Flavell, R.A. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell 2002, 110, 191–202. [Google Scholar] [CrossRef] [Green Version]
- Bieberich, E.; MacKinnon, S.; Silva, J.; Noggle, S.; Condie, B.G. Regulation of cell death in mitotic neural progenitor cells by asymmetric distribution of prostate apoptosis response 4 (PAR-4) and simultaneous elevation of endogenous ceramide. J. Cell Biol. 2003, 162, 469–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bieberich, E. Integration of glycosphingolipid metabolism and cell-fate decisions in cancer and stem cells: Review and hypothesis. Glycoconj. J. 2004, 21, 315–327. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Dinkins, M.; He, Q.; Zhu, G.; Poirier, C.; Campbell, A.; Mayer-Proschel, M.; Bieberich, E. Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4) potential mechanism of apoptosis induction in Alzheimer disease (AD). J. Biol. Chem. 2012, 287, 21384–21395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bakhti, M.; Winter, C.; Simons, M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 2010, 286, 787–796. [Google Scholar] [CrossRef] [Green Version]
- Frohlich, D.; Kuo, W.P.; Fruhbeis, C.; Sun, J.J.; Zehendner, C.M.; Luhmann, H.J.; Pinto, S.; Toedling, J.; Trotter, J.; Kramer-Albers, E.M. Multifaceted effects of oligodendroglial exosomes on neurons: Impact on neuronal firing rate, signal transduction and gene regulation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Hakulinen, J.; Sankkila, L.; Sugiyama, N.; Lehti, K.; Keski-Oja, J. Secretion of active membrane type 1 matrix metalloproteinase (MMP-14) into extracellular space in microvesicular exosomes. J. Cell Biochem. 2008, 105, 1211–1218. [Google Scholar] [CrossRef]
- Sarkar, S.; Yong, V.W. Inflammatory cytokine modulation of matrix metalloproteinase expression and invasiveness of glioma cells in a 3-dimensional collagen matrix. J. Neurooncol. 2009, 91, 157–164. [Google Scholar] [CrossRef]
- Jy, W.; Minagar, A.; Jimenez, J.J.; Sheremata, W.A.; Mauro, L.M.; Horstman, L.L.; Bidot, C.; Ahn, Y.S. Endothelial microparticles (EMP) bind and activate monocytes: Elevated EMP-monocyte conjugates in multiple sclerosis. Front. Biosci. 2004, 9, 3137–3144. [Google Scholar] [CrossRef] [Green Version]
- Quandt, J.; Dorovini-Zis, K. The β chemokines CCL4 and CCL5 enhance adhesion of specific CD4+ T cell subsets to human brain endothelial cells. J. Neuropathol. Exp. Neurol. 2004, 63, 350–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galazka, G.; Mycko, M.P.; Selmaj, I.; Raine, C.S.; Selmaj, K.W. Multiple sclerosis: Serum-Derived exosomes express myelin proteins. Mult. Scler. 2017, 24, 449–458. [Google Scholar] [CrossRef] [PubMed]
- Pieragostino, D.; Cicalini, I.; Lanuti, P.; Ercolino, E.; di Ioia, M.; Zucchelli, M.; Zappacosta, R.; Miscia, S.; Marchisio, M.; Sacchetta, P.; et al. Enhanced release of acid sphingomyelinase-enriched exosomes generates a lipidomics signature in CSF of Multiple Sclerosis patients. Sci. Rep. 2018, 8, 3071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirsch, E.; Ruberg, M.; Portier, M.M.; Dardenne, M.; Agid, Y. Characterization of two antigens in parkinsonian Lewy bodies. Brain Res. 1988, 441, 139–144. [Google Scholar] [CrossRef]
- Harraz, M.M.; Eacker, S.M.; Wang, X.; Dawson, T.M.; Dawson, V.L. MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proc. Natl. Acad. Sci. USA 2012, 109, 18962–18967. [Google Scholar] [CrossRef] [Green Version]
- Fortin, D.L.; Nemani, V.M.; Voglmaier, S.M.; Anthony, M.D.; Ryan, T.A.; Edwards, R.H. Neural activity controls the synaptic accumulation of α-synuclein. J. Neurosci. 2005, 25, 10913–10921. [Google Scholar] [CrossRef] [Green Version]
- Goedert, M. The significance of tau and α-synuclein inclusions in neurodegenerative diseases. Curr. Opin. Genet. Dev. 2001, 11, 343–351. [Google Scholar] [CrossRef]
- Prusiner, S.B.; Woerman, A.L.; Mordes, D.A.; Watts, J.C.; Rampersaud, R.; Berry, D.B.; Patel, S.; Oehler, A.; Lowe, J.K.; Kravitz, S.N.; et al. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc. Natl. Acad. Sci. USA 2015, 112, E5308–E5317. [Google Scholar] [CrossRef] [Green Version]
- Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42. [Google Scholar] [CrossRef] [Green Version]
- Grozdanov, V.; Danzer, K.M. Release and uptake of pathologic α-synuclein. Cell Tissue Res. 2018, 373, 175–182. [Google Scholar] [CrossRef] [PubMed]
- Ishizawa, T.; Mattila, P.; Davies, P.; Wang, D.; Dickson, D.W. Colocalization of tau and α-synuclein epitopes in Lewy bodies. J. Neuropathol. Exp. Neurol. 2003, 62, 389–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clinton, L.K.; Blurton-Jones, M.; Myczek, K.; Trojanowski, J.Q.; LaFerla, F.M. Synergistic Interactions between Aβ, τ, and α-synuclein: Acceleration of neuropathology and cognitive decline. J. Neurosci. 2010, 30, 7281–7289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, D.; Li, Y.P.; Li, Y.X.; Zhu, X.H.; Du, X.G.; Zhou, M.; Li, W.B.; Deng, H.Y. Effect of Regulatory Network of Exosomes and microRNAs on Neurodegenerative Diseases. Chin. Med. J. 2018, 131, 2216–2225. [Google Scholar] [CrossRef] [PubMed]
- Kalani, A.; Tyagi, A.; Tyagi, N. Exosomes: Mediators of neurodegeneration, neuroprotection and therapeutics. Mol. Neurobiol. 2013, 49, 590–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leidinger, P.; Backes, C.; Deutscher, S.; Schmitt, K.; Mueller, S.C.; Frese, K.; Haas, J.; Ruprecht, K.; Paul, F.; Stahler, C.; et al. A blood based 12-miRNA signature of Alzheimer disease patients. Genome Biol. 2013, 14, R78. [Google Scholar] [CrossRef] [Green Version]
- Zuccato, C.; Ciammola, A.; Rigamonti, D.; Leavitt, B.R.; Goffredo, D.; Conti, L.; MacDonald, M.E.; Friedlander, R.M.; Silani, V.; Hayden, M.R.; et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science 2001, 293, 493–498. [Google Scholar] [CrossRef] [Green Version]
- Hoss, A.G.; Labadorf, A.; Beach, T.G.; Latourelle, J.C.; Myers, R.H. microRNA Profiles in Parkinson’s Disease Prefrontal Cortex. Front. Aging Neurosci. 2016, 8, 36. [Google Scholar] [CrossRef] [Green Version]
- Reed, E.R.; Latourelle, J.C.; Bockholt, J.H.; Bregu, J.; Smock, J.; Paulsen, J.S.; Myers, R.H. MicroRNAs in CSF as prodromal biomarkers for Huntington disease in the PREDICT-HD study. Neurology 2017, 90, e264–e272. [Google Scholar] [CrossRef]
- Si, Y.; Cui, X.; Crossman, D.K.; Hao, J.; Kazamel, M.; Kwon, Y.; King, P.H. Muscle microRNA signatures as biomarkers of disease progression in amyotrophic lateral sclerosis. Neurobiol. Dis. 2018, 114, 85–94. [Google Scholar] [CrossRef]
- Chen, J.J.; Zhao, B.; Zhao, J.; Li, S. Potential Roles of Exosomal MicroRNAs as Diagnostic Biomarkers and Therapeutic Application in Alzheimer’s Disease. Neural Plast. 2017, 2017, 7027380. [Google Scholar] [CrossRef]
- Cardo, L.F.; Coto, E.; de Mena, L.; Ribacoba, R.; Moris, G.; Menendez, M.; Alvarez, V. Profile of microRNAs in the plasma of Parkinson’s disease patients and healthy controls. J. Neurol. 2013, 260, 1420–1422. [Google Scholar] [CrossRef] [PubMed]
- Ebrahimkhani, S.; Vafaee, F.; Young, P.E.; Hur, S.S.J.; Hawke, S.; Devenney, E.; Beadnall, H.; Barnett, M.H.; Suter, C.M.; Buckland, M.E. Exosomal microRNA signatures in multiple sclerosis reflect disease status. Sci. Rep. 2017, 7, 14293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kimura, K.; Hohjoh, H.; Fukuoka, M.; Sato, W.; Oki, S.; Tomi, C.; Yamaguchi, H.; Kondo, T.; Takahashi, R.; Yamamura, T. Circulating exosomes suppress the induction of regulatory T cells via let-7i in multiple sclerosis. Nat. Commun. 2018, 9, 17. [Google Scholar] [CrossRef] [PubMed]
- Properzi, F.; Ferroni, E.; Poleggi, A.; Vinci, R. The regulation of exosome function in the CNS: Implications for neurodegeneration. Swiss Med. Wkly. 2015, 145, w14204. [Google Scholar] [CrossRef]
- Lehmann, S.M.; Kruger, C.; Park, B.; Derkow, K.; Rosenberger, K.; Baumgart, J.; Trimbuch, T.; Eom, G.; Hinz, M.; Kaul, D.; et al. An unconventional role for miRNA: Let-7 activates Toll-like receptor 7 and causes neurodegeneration. Nat. Neurosci. 2012, 15, 827–835. [Google Scholar] [CrossRef]
- Winkler, C.W.; Taylor, K.G.; Peterson, K.E. Location is everything: Let-7b microRNA and TLR7 signaling results in a painful TRP. Sci. Signal. 2014, 7, pe14. [Google Scholar] [CrossRef]
- Azimi, M.; Ghabaee, M.; Naser Moghadasi, A.; Izad, M. Altered Expression of miR-326 in T Cell-derived Exosomes of Patients with Relapsing-Remitting Multiple Sclerosis. Iran J. Allergy Asthma Immunol. 2019, 18, 108–113. [Google Scholar] [CrossRef] [Green Version]
- Nicolas, M.; Hassan, B.A. Amyloid precursor protein and neural development. Development 2014, 141, 2543–2548. [Google Scholar] [CrossRef] [Green Version]
- Vilardo, E.; Barbato, C.; Ciotti, M.; Cogoni, C.; Ruberti, F. MicroRNA-101 regulates amyloid precursor protein expression in hippocampal neurons. J. Biol. Chem. 2010, 285, 18344–18351. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.G.; Wang, J.L.; Li, L.; Wang, P.C. MicroRNA-384 regulates both amyloid precursor protein and β-secretase expression and is a potential biomarker for Alzheimer’s disease. Int. J. Mol. Med. 2014, 34, 160–166. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Chen, C.F.; Wang, A.H.; Lin, Q.F. MiR-16 regulates cell death in Alzheimer’s disease by targeting amyloid precursor protein. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 4020–4027. [Google Scholar] [PubMed]
- Dickson, J.R.; Kruse, C.; Montagna, D.R.; Finsen, B.; Wolfe, M.S. Alternative polyadenylation and miR-34 family members regulate tau expression. J. Neurochem. 2013, 127, 739–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santa-Maria, I.; Alaniz, M.E.; Renwick, N.; Cela, C.; Fulga, T.A.; Van Vactor, D.; Tuschl, T.; Clark, L.N.; Shelanski, M.L.; McCabe, B.D.; et al. Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Investig. 2015, 125, 681–686. [Google Scholar] [CrossRef] [PubMed]
- Du, J.J.; Chen, S.D. Current Nondopaminergic Therapeutic Options for Motor Symptoms of Parkinson’s Disease. Chin. Med. J. 2017, 130, 1856–1866. [Google Scholar] [CrossRef] [PubMed]
- McMillan, K.J.; Murray, T.K.; Bengoa-Vergniory, N.; Cordero-Llana, O.; Cooper, J.; Buckley, A.; Wade-Martins, R.; Uney, J.B.; O’Neill, M.J.; Wong, L.F.; et al. Loss of MicroRNA-7 Regulation Leads to α-Synuclein Accumulation and Dopaminergic Neuronal Loss In Vivo. Mol. Ther. 2017, 25, 2404–2414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Guescini, M.; Genedani, S.; Stocchi, V.; Agnati, L.F. Astrocytes and Glioblastoma cells release exosomes carrying mtDNA. J. Neural Transm. 2010, 117, 1. [Google Scholar] [CrossRef]
- Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769. [Google Scholar] [CrossRef] [Green Version]
- Huang, X.; Yuan, T.; Tschannen, M.; Sun, Z.; Jacob, H.; Du, M.; Liang, M.; Dittmar, R.L.; Liu, Y.; Liang, M. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genom. 2013, 14, 319. [Google Scholar] [CrossRef] [Green Version]
- Aliotta, J.M.; Pereira, M.; Johnson, K.W.; de Paz, N.; Dooner, M.S.; Puente, N.; Ayala, C.; Brilliant, K.; Berz, D.; Lee, D. Microvesicle entry into marrow cells mediates tissue-specific changes in mRNA by direct delivery of mRNA and induction of transcription. Exp. Hematol. 2010, 38, 233–245. [Google Scholar] [CrossRef] [Green Version]
- Camussi, G.; Deregibus, M.C.; Bruno, S.; Cantaluppi, V.; Biancone, L. Exosomes/Microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010, 78, 838–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, S.-S.; Ishibashi, O.; Ishikawa, G.; Ishikawa, T.; Katayama, A.; Mishima, T.; Takizawa, T.; Shigihara, T.; Goto, T.; Izumi, A. Human villous trophoblasts express and secrete placenta-specific microRNAs into maternal circulation via exosomes. Biol. Reprod. 2009, 81, 717–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome theranostics: Biology and translational medicine. Theranostics 2018, 8, 237. [Google Scholar] [CrossRef] [PubMed]
- De Jong, O.G.; Verhaar, M.C.; Chen, Y.; Vader, P.; Gremmels, H.; Posthuma, G.; Schiffelers, R.M.; Gucek, M.; van Balkom, B.W. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. J. Extracell. Vesicles 2012, 1, 18396. [Google Scholar] [CrossRef] [PubMed]
- Laulagnier, K.; Vincent-Schneider, H.; Hamdi, S.; Subra, C.; Lankar, D.; Record, M. Characterization of exosome subpopulations from RBL-2H3 cells using fluorescent lipids. Blood Cells Mol. Dis. 2005, 35, 116–121. [Google Scholar] [CrossRef]
- Oksvold, M.P.; Kullmann, A.; Forfang, L.; Kierulf, B.; Li, M.; Brech, A.; Vlassov, A.V.; Smeland, E.B.; Neurauter, A.; Pedersen, K.W. Expression of B-cell surface antigens in subpopulations of exosomes released from B-cell lymphoma cells. Clin. Ther. 2014, 36, 847–862.e841. [Google Scholar] [CrossRef] [Green Version]
- Smith, Z.J.; Lee, C.; Rojalin, T.; Carney, R.P.; Hazari, S.; Knudson, A.; Lam, K.; Saari, H.; Ibañez, E.L.; Viitala, T. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J. Extracell. Vesicles 2015, 4, 28533. [Google Scholar] [CrossRef] [Green Version]
- Mathivanan, S.; Ji, H.; Simpson, R.J. Exosomes: Extracellular organelles important in intercellular communication. J. Proteom. 2010, 73, 1907–1920. [Google Scholar] [CrossRef]
- Van Der Vlist, E.J.; Nolte, E.N.; Stoorvogel, W.; Arkesteijn, G.J.; Wauben, M.H. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat. Protoc. 2012, 7, 1311–1326. [Google Scholar] [CrossRef]
- Pospichalova, V.; Svoboda, J.; Dave, Z.; Kotrbova, A.; Kaiser, K.; Klemova, D.; Ilkovics, L.; Hampl, A.; Crha, I.; Jandakova, E. Simplified protocol for flow cytometry analysis of fluorescently labeled exosomes and microvesicles using dedicated flow cytometer. J. Extracell. Vesicles 2015, 4, 25530. [Google Scholar] [CrossRef]
- Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Mathivanan, S.; Ji, H.; Simpson, R.J. Two distinct populations of exosomes are released from LIM1863 colon carcinoma cell-derived organoids. Mol. Cell. Proteom. 2013, 12, 587–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferguson, S.W.; Nguyen, J. Exosomes as therapeutics: The implications of molecular composition and exosomal heterogeneity. J. Control. Release 2016, 228, 179–190. [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.; Würdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balaj, L.; Lessard, R.; Dai, L.; Cho, Y.-J.; Pomeroy, S.L.; Breakefield, X.O.; Skog, J. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2011, 2, 1–9. [Google Scholar] [CrossRef]
- Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, A.; Okada, R.; Nagao, K.; Kawamata, Y.; Hanyu, A.; Yoshimoto, S.; Takasugi, M.; Watanabe, S.; Kanemaki, M.T.; Obuse, C. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun. 2017, 8, 15287. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Png, K.J.; Halberg, N.; Yoshida, M.; Tavazoie, S.F. A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Nature 2012, 481, 190–194. [Google Scholar] [CrossRef]
- Rabinowits, G.; Gerçel-Taylor, C.; Day, J.M.; Taylor, D.D.; Kloecker, G.H. Exosomal microRNA: A diagnostic marker for lung cancer. Clin. Lung Cancer 2009, 10, 42–46. [Google Scholar] [CrossRef]
- Skog, J.; Würdinger, T.; Van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T.; 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]
- Koppers-Lalic, D.; Hackenberg, M.; Bijnsdorp, I.V.; van Eijndhoven, M.A.; Sadek, P.; Sie, D.; Zini, N.; Middeldorp, J.M.; Ylstra, B.; de Menezes, R.X. Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Rep. 2014, 8, 1649–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tran, N. Cancer exosomes as miRNA factories. Trends Cancer 2016, 2, 329–331. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- 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]
- Jenjaroenpun, P.; Kremenska, Y.; Nair, V.M.; Kremenskoy, M.; Joseph, B.; Kurochkin, I.V. Characterization of RNA in exosomes secreted by human breast cancer cell lines using next-generation sequencing. PeerJ 2013, 1, e201. [Google Scholar] [CrossRef] [Green Version]
- Baglio, S.R.; Rooijers, K.; Koppers-Lalic, D.; Verweij, F.J.; Lanzón, M.P.; Zini, N.; Naaijkens, B.; Perut, F.; Niessen, H.W.; Baldini, N. Human bone marrow-and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res. Ther. 2015, 6, 127. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Zeringer, E.; Barta, T.; Schageman, J.; Cheng, A.; Vlassov, A.V. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130502. [Google Scholar] [CrossRef]
- Kumar, P.; Anaya, J.; Mudunuri, S.B.; Dutta, A. Meta-Analysis of tRNA derived RNA fragments reveals that they are evolutionarily conserved and associate with AGO proteins to recognize specific RNA targets. BMC Biol. 2014, 12, 78. [Google Scholar] [CrossRef]
- Gopinath, S.C.; Wadhwa, R.; Kumar, P.K. Expression of noncoding vault RNA in human malignant cells and its importance in mitoxantrone resistance. Mol. Cancer Res. 2010, 8, 1536–1546. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.-S.; Choi, D.-Y.; Yun, S.J.; Choi, S.-M.; Kang, J.W.; Jung, J.W.; Hwang, D.; Kim, K.P.; Kim, D.-W. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J. Proteome Res. 2012, 11, 839–849. [Google Scholar] [CrossRef]
- Simpson, R.J.; Jensen, S.S.; Lim, J.W. Proteomic profiling of exosomes: Current perspectives. Proteomics 2008, 8, 4083–4099. [Google Scholar] [CrossRef] [PubMed]
- Salomon, C.; Ryan, J.; Sobrevia, L.; Kobayashi, M.; Ashman, K.; Mitchell, M.; Rice, G.E. Exosomal signaling during hypoxia mediates microvascular endothelial cell migration and vasculogenesis. PLoS ONE 2013, 8, e68451. [Google Scholar] [CrossRef] [PubMed]
- Hegmans, J.P.; Bard, M.P.; Hemmes, A.; Luider, T.M.; Kleijmeer, M.J.; Prins, J.-B.; Zitvogel, L.; Burgers, S.A.; Hoogsteden, H.C.; Lambrecht, B.N. Proteomic analysis of exosomes secreted by human mesothelioma cells. Am. J. Pathol. 2004, 164, 1807–1815. [Google Scholar] [CrossRef] [Green Version]
- Simons, M.; Raposo, G. Exosomes–Vesicular carriers for intercellular communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
- Bakela, K.; Kountourakis, N.; Aivaliotis, M.; Athanassakis, I. Soluble MHC-II proteins promote suppressive activity in CD 4+ T cells. Immunology 2015, 144, 158–169. [Google Scholar] [CrossRef] [Green Version]
- Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; Degeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E. Syndecan–Syntenin–ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef]
- Theos, A.C.; Truschel, S.T.; Tenza, D.; Hurbain, I.; Harper, D.C.; Berson, J.F.; Thomas, P.C.; Raposo, G.; Marks, M.S. A lumenal domain-dependent pathway for sorting to intralumenal vesicles of multivesicular endosomes involved in organelle morphogenesis. Dev. Cell 2006, 10, 343–354. [Google Scholar] [CrossRef] [Green Version]
- Reggiori, F.; Pelham, H.R. Sorting of proteins into multivesicular bodies: Ubiquitin-dependent and-independent targeting. EMBO J. 2001, 20, 5176–5186. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Khan, S.; Jiang, H.; Antonyak, M.A.; Chen, X.; Spiegelman, N.A.; Shrimp, J.H.; Cerione, R.A.; Lin, H. Identifying the functional contribution of the defatty-acylase activity of SIRT6. Nat. Chem. Biol. 2016, 12, 614–620. [Google Scholar] [CrossRef] [Green Version]
- Hsu, D.-H.; Paz, P.; Villaflor, G.; Rivas, A.; Mehta-Damani, A.; Angevin, E.; Zitvogel, L.; Le Pecq, J.-B. Exosomes as a tumor vaccine: Enhancing potency through direct loading of antigenic peptides. J. Immunother. 2003, 26, 440–450. [Google Scholar] [CrossRef] [PubMed]
- Morse, M.A.; Garst, J.; Osada, T.; Khan, S.; Hobeika, A.; Clay, T.M.; Valente, N.; Shreeniwas, R.; Sutton, M.A.; Delcayre, A. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J. Transl. Med. 2005, 3, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Formiga, F.R.; Pelacho, B.; Garbayo, E.; Imbuluzqueta, I.; Díaz-Herráez, P.; Abizanda, G.; Gavira, J.J.; Simón-Yarza, T.; Albiasu, E.; Tamayo, E. Controlled delivery of fibroblast growth factor-1 and neuregulin-1 from biodegradable microparticles promotes cardiac repair in a rat myocardial infarction model through activation of endogenous regeneration. J. Control. Release 2014, 173, 132–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palmen, M.; Daemen, M.J.; De Windt, L.J.; Willems, J.; Dassen, W.R.; Heeneman, S.; Zimmermann, R.; Van Bilsen, M.; Doevendans, P.A. Fibroblast growth factor-1 improves cardiac functional recovery and enhances cell survival after ischemia and reperfusion: A fibroblast growth factor receptor, protein kinase C, and tyrosine kinase-dependent mechanism. J. Am. Coll. Cardiol. 2004, 44, 1113–1123. [Google Scholar] [CrossRef] [Green Version]
- Okazaki, T.; Ebihara, S.; Asada, M.; Yamanda, S.; Saijo, Y.; Shiraishi, Y.; Ebihara, T.; Niu, K.; Mei, H.; Arai, H. Macrophage colony-stimulating factor improves cardiac function after ischemic injury by inducing vascular endothelial growth factor production and survival of cardiomyocytes. Am. J. Pathol. 2007, 171, 1093–1103. [Google Scholar] [CrossRef] [Green Version]
- Hill, M.F.; Patel, A.V.; Murphy, A.; Smith, H.M.; Galindo, C.L.; Pentassuglia, L.; Peng, X.; Lenneman, C.G.; Odiete, O.; Friedman, D.B. Intravenous glial growth factor 2 (GGF2) isoform of neuregulin-1β improves left ventricular function, gene and protein expression in rats after myocardial infarction. PLoS ONE 2013, 8, e55741. [Google Scholar] [CrossRef]
- Zgheib, C.; Zouein, F.A.; Kurdi, M.; Booz, G.W. Chronic treatment of mice with leukemia inhibitory factor does not cause adverse cardiac remodeling but improves heart function. Eur. Cytokine Netw. 2012, 23, 191. [Google Scholar] [CrossRef] [Green Version]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Wubbolts, R.; Leckie, R.S.; Veenhuizen, P.T.; Schwarzmann, G.; Möbius, W.; Hoernschemeyer, J.; Slot, J.-W.; Geuze, H.J.; Stoorvogel, W. Proteomic and biochemical analyses of human B cell-derived exosomes Potential implications for their function and multivesicular body formation. J. Biol. Chem. 2003, 278, 10963–10972. [Google Scholar] [CrossRef] [Green Version]
- Krämer-Albers, E.M.; Bretz, N.; Tenzer, S.; Winterstein, C.; Möbius, W.; Berger, H.; Nave, K.A.; Schild, H.; Trotter, J. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteom. Clin. Appl. 2007, 1, 1446–1461. [Google Scholar] [CrossRef]
- Laulagnier, K.; Motta, C.; Hamdi, S.; Roy, S.; Fauvelle, F.; Pageaux, J.-F.; Kobayashi, T.; Salles, J.-P.; Perret, B.; Bonnerot, C. Mast cell-and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem. J. 2004, 380, 161–171. [Google Scholar] [CrossRef] [PubMed]
- Subra, C.; Grand, D.; Laulagnier, K.; Stella, A.; Lambeau, G.; Paillasse, M.; De Medina, P.; Monsarrat, B.; Perret, B.; Silvente-Poirot, S. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J. Lipid Res. 2010, 51, 2105–2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mashouri, L.; Yousefi, H.; Aref, A.R.; Mohammad Ahadi, A.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef] [PubMed]
- Beloribi-Djefaflia, S.; Siret, C.; Lombardo, D. Exosomal lipids induce human pancreatic tumoral MiaPaCa-2 cells resistance through the CXCR4-SDF-1α signaling axis. Oncoscience 2015, 2, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Urbanelli, L.; Magini, A.; Buratta, S.; Brozzi, A.; Sagini, K.; Polchi, A.; Tancini, B.; Emiliani, C. Signaling pathways in exosomes biogenesis, secretion and fate. Genes 2013, 4, 152–170. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Cheruvanky, A.; Hu, X.; Matsumoto, T.; Hiramatsu, N.; Cho, M.E.; Berger, A.; Leelahavanichkul, A.; Doi, K.; Chawla, L.S. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney Int. 2008, 74, 613–621. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Pisitkun, T.; Aponte, A.; Yuen, P.S.; Hoffert, J.D.; Yasuda, H.; Hu, X.; Chawla, L.; Shen, R.-F.; Knepper, M.A. Exosomal Fetuin-A identified by proteomics: A novel urinary biomarker for detecting acute kidney injury. Kidney Int. 2006, 70, 1847–1857. [Google Scholar] [CrossRef] [Green Version]
- Welker, M.-W.; Reichert, D.; Susser, S.; Sarrazin, C.; Martinez, Y.; Herrmann, E.; Zeuzem, S.; Piiper, A.; Kronenberger, B. Soluble serum CD81 is elevated in patients with chronic hepatitis C and correlates with alanine aminotransferase serum activity. PLoS ONE 2012, 7, e30796. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zhang, Y.; Qiu, F.; Qiu, Z. Proteomic identification of exosomal LRG1: A potential urinary biomarker for detecting NSCLC. Electrophoresis 2011, 32, 1976–1983. [Google Scholar] [CrossRef]
- 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]
- Russo, L. Urine Biomarkers. WO2013028788A1, 28 February 2013. [Google Scholar]
- Dong, Z.; Sun, X.; Xu, J.; Han, X.; Xing, Z.; Wang, D.; Ge, J.; Meng, L.; Xu, X. Serum Membrane Type 1-Matrix Metalloproteinase (MT1-MMP) mRNA Protected by Exosomes as a Potential Biomarker for Gastric Cancer. Medical Sci. Monit. 2019, 25, 7770. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Dong, X.; Chen, Y.; Wang, X. Serum exosomal hnRNPH1 mRNA as a novel marker for hepatocellular carcinoma. Clin. Chem. Lab. Med. 2018, 56, 479–484. [Google Scholar] [CrossRef] [PubMed]
- Mao, L.; Li, X.; Gong, S.; Yuan, H.; Jiang, Y.; Huang, W.; Sun, X.; Dang, X. Serum exosomes contain ECRG 4 mRNA that suppresses tumor growth via inhibition of genes involved in inflammation, cell proliferation, and angiogenesis. Cancer Gene Ther. 2018, 25, 248–259. [Google Scholar] [CrossRef] [PubMed]
- Bracci, L.; Lozupone, F.; Parolini, I. The role of exosomes in colorectal cancer disease progression and response to therapy. Cytokine Growth Factor Rev. 2020, 51, 84–91. [Google Scholar] [CrossRef] [PubMed]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Sugimachi, K.; Matsumura, T.; Hirata, H.; Uchi, R.; Ueda, M.; Ueo, H.; Shinden, Y.; Iguchi, T.; Eguchi, H.; Shirabe, K. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 2015, 112, 532–538. [Google Scholar] [CrossRef]
- Takeshita, N.; Hoshino, I.; Mori, M.; Akutsu, Y.; Hanari, N.; Yoneyama, Y.; Ikeda, N.; Isozaki, Y.; Maruyama, T.; Akanuma, N. Serum microRNA expression profile: miR-1246 as a novel diagnostic and prognostic biomarker for oesophageal squamous cell carcinoma. Br. J. Cancer 2013, 108, 644–652. [Google Scholar] [CrossRef]
- Fu, F.; Jiang, W.; Zhou, L.; Chen, Z. Circulating exosomal miR-17-5p and miR-92a-3p predict pathologic stage and grade of colorectal cancer. Trans. Oncol. 2018, 11, 221–232. [Google Scholar] [CrossRef]
- De Miguel Pérez, D.; Martínez, A.R.; Palomo, A.O.; Ureña, M.D.; Puche, J.L.G.; Remacho, A.R.; Hernandez, J.E.; Acosta, J.A.L.; Sánchez, F.G.O.; Serrano, M.J. Extracellular vesicle-miRNAs as liquid biopsy biomarkers for disease identification and prognosis in metastatic colorectal cancer patients. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Zhao, Y.; Song, X.; Song, X.; Niu, L.; Xie, L. Tumor-Derived exosomal miRNA-320d as a biomarker for metastatic colorectal cancer. J. Clin. Lab. Anal. 2019, 33, e23004. [Google Scholar] [CrossRef] [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. 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]
- Brase, J.C.; Johannes, M.; Schlomm, T.; Fälth, M.; Haese, A.; Steuber, T.; Beissbarth, T.; Kuner, R.; Sültmann, H. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int. J. Cancer 2011, 128, 608–616. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Duan, W.; Yan, S.; Xie, Y.; Wang, C. Circulating long non-coding RNA colon cancer-associated transcript 2 protected by exosome as a potential biomarker for colorectal cancer. Biomed. Pharmacother. 2019, 113, 108758. [Google Scholar] [CrossRef] [PubMed]
- Yu, B.; Du, Q.; Li, H.; Liu, H.-Y.; Ye, X.; Zhu, B.; Zhai, Q.; Li, X.-X. Diagnostic potential of serum exosomal colorectal neoplasia differentially expressed long non-coding RNA (CRNDE-p) and microRNA-217 expression in colorectal carcinoma. Oncotarget 2017, 8, 83745. [Google Scholar] [CrossRef] [PubMed]
- Gao, T.; Liu, X.; He, B.; Nie, Z.; Zhu, C.; Zhang, P.; Wang, S. Exosomal lncRNA 91H is associated with poor development in colorectal cancer by modifying HNRNPK expression. Cancer Cell Int. 2018, 18, 11. [Google Scholar] [CrossRef] [PubMed]
- Oehme, F.; Krahl, S.; Gyorffy, B.; Muessle, B.; Rao, V.; Greif, H.; Ziegler, N.; Lin, K.; Thepkaysone, M.-L.; Polster, H. Low level of exosomal long non-coding RNA HOTTIP is a prognostic biomarker in colorectal cancer. RNA Biol. 2019, 16, 1339–1345. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhou, Y.; Lu, J.; Sun, Y.; Xiao, H.; Liu, M.; Tian, L. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med. Oncol. 2014, 31, 148. [Google Scholar] [CrossRef]
- Yamashita, T.; Kamada, H.; Kanasaki, S.; Maeda, Y.; Nagano, K.; Abe, Y.; Inoue, M.; Yoshioka, Y.; Tsutsumi, Y.; Katayama, S. Epidermal growth factor receptor localized to exosome membranes as a possible biomarker for lung cancer diagnosis. Die Pharm. Int. J. Pharm. Sci. 2013, 68, 969–973. [Google Scholar]
- Jakobsen, K.R.; Paulsen, B.S.; Bæk, R.; Varming, K.; Sorensen, B.S.; Jørgensen, M.M. Exosomal proteins as potential diagnostic markers in advanced non-small cell lung carcinoma. J. Extracell. Vesicles 2015, 4, 26659. [Google Scholar] [CrossRef]
- Graner, M.W.; Alzate, O.; Dechkovskaia, A.M.; Keene, J.D.; Sampson, J.H.; Mitchell, D.A.; Bigner, D.D. Proteomic and immunologic analyses of brain tumor exosomes. FASEB J. 2009, 23, 1541–1557. [Google Scholar] [CrossRef] [Green Version]
- Shao, H.; Chung, J.; Balaj, L.; Charest, A.; Bigner, D.D.; Carter, B.S.; Hochberg, F.H.; Breakefield, X.O.; Weissleder, R.; Lee, H. Protein typing of circulating microvesicles allows real-time monitoring of glioblastoma therapy. Nat. Med. 2012, 18, 1835. [Google Scholar] [CrossRef] [PubMed]
- Nilsson, J.; Skog, J.; Nordstrand, A.; Baranov, V.; Mincheva-Nilsson, L.; Breakefield, X.; Widmark, A. Prostate cancer-derived urine exosomes: A novel approach to biomarkers for prostate cancer. Br. J. Cancer 2009, 100, 1603–1607. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.-L.; Lai, Y.-F.; Tang, P.; Chien, K.-Y.; Yu, J.-S.; Tsai, C.-H.; Chen, H.-W.; Wu, C.-C.; Chung, T.; Hsu, C.-W. Comparative and targeted proteomic analyses of urinary microparticles from bladder cancer and hernia patients. J. Proteome Res. 2012, 11, 5611–5629. [Google Scholar] [CrossRef] [PubMed]
- Runz, S.; Keller, S.; Rupp, C.; Stoeck, A.; Issa, Y.; Koensgen, D.; Mustea, A.; Sehouli, J.; Kristiansen, G.; Altevogt, P. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol. Oncol. 2007, 107, 563–571. [Google Scholar] [CrossRef] [PubMed]
- Logozzi, M.; De Milito, A.; Lugini, L.; Borghi, M.; Calabro, L.; Spada, M.; Perdicchio, M.; Marino, M.L.; Federici, C.; Iessi, E. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS ONE 2009, 4, e5219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Logozzi, M.; Mizzoni, D.; Capasso, C.; Del Prete, S.; Di Raimo, R.; Falchi, M.; Angelini, D.F.; Sciarra, A.; Maggi, M.; Supuran, C.T. Plasmatic exosomes from prostate cancer patients show increased carbonic anhydrase IX expression and activity and low pH. J. Enzym. Inhib. Med. Chem. 2020, 35, 280–288. [Google Scholar] [CrossRef] [Green Version]
- Logozzi, M.; Capasso, C.; Di Raimo, R.; Del Prete, S.; Mizzoni, D.; Falchi, M.; Supuran, C.T.; Fais, S. Prostate cancer cells and exosomes in acidic condition show increased carbonic anhydrase IX expression and activity. J. Enzym. Inhib. Med. Chem. 2019, 34, 272–278. [Google Scholar] [CrossRef] [Green Version]
- Palacios-Ferrer, J.L.; García-Ortega, M.B.; Gallardo-Gómez, M.; García, M.Á.; Díaz, C.; Boulaiz, H.; Valdivia, J.; Jurado, J.M.; Almazan-Fernandez, F.M.; Arias-Santiago, S. Metabolomic profile of cancer stem cell-derived exosomes from patients with malignant melanoma. Mol. Oncol. 2020. [Google Scholar] [CrossRef]
- Delpech, J.C.; Herron, S.; Botros, M.B.; Ikezu, T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci. 2019, 42, 361–372. [Google Scholar] [CrossRef]
- Tomlinson, P.R.; Zheng, Y.; Fischer, R.; Heidasch, R.; Gardiner, C.; Evetts, S.; Hu, M.; Wade-Martins, R.; Turner, M.R.; Morris, J. Identification of distinct circulating exosomes in Parkinson’s disease. Ann. Clin. Transl. Neurol. 2015, 2, 353–361. [Google Scholar] [CrossRef]
- Fraser, K.B.; Moehle, M.S.; Alcalay, R.N.; West, A.B. Urinary LRRK2 phosphorylation predicts parkinsonian phenotypes in G2019S LRRK2 carriers. Neurology 2016, 86, 994–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olsson, B.; Lautner, R.; Andreasson, U.; Öhrfelt, A.; Portelius, E.; Bjerke, M.; Hölttä, M.; Rosén, C.; Olsson, C.; Strobel, G. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol. 2016, 15, 673–684. [Google Scholar] [CrossRef]
- Mapstone, M.; Cheema, A.K.; Fiandaca, M.S.; Zhong, X.; Mhyre, T.R.; MacArthur, L.H.; Hall, W.J.; Fisher, S.G.; Peterson, D.R.; Haley, J.M. Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 2014, 20, 415–418. [Google Scholar] [CrossRef] [PubMed]
- Roth, P.; Weller, M. Challenges to targeting epidermal growth factor receptor in glioblastoma: Escape mechanisms and combinatorial treatment strategies. Neuro Oncol. 2014, 16, 14–19. [Google Scholar] [CrossRef] [PubMed]
- Sharif, S.; Ghahremani, M.H.; Soleimani, M. Delivery of Exogenous miR-124 to Glioblastoma Multiform Cells by Wharton’s Jelly Mesenchymal Stem Cells Decreases Cell Proliferation and Migration, and Confers Chemosensitivity. Stem Cell Rev. Rep. 2018, 14, 236–246. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Wu, Z.H.; Lou, P.Y.; Chai, C.; Han, S.Y.; Ning, J.F.; Li, M. Human bone marrow-derived mesenchymal stem cell-secreted exosomes overexpressing microRNA-34a ameliorate glioblastoma development via down-regulating MYCN. Cell. Oncol. 2019, 42, 783–799. [Google Scholar] [CrossRef]
- Liu, Q.; Yu, Z.; Yuan, S.; Xie, W.; Li, C.; Hu, Z.; Xiang, Y.; Wu, N.; Wu, L.; Bai, L.; et al. Circulating exosomal microRNAs as prognostic biomarkers for non-small-cell lung cancer. Oncotarget 2017, 8, 13048–13058. [Google Scholar] [CrossRef] [Green Version]
- Kanaoka, R.; Iinuma, H.; Dejima, H.; Sakai, T.; Uehara, H.; Matsutani, N.; Kawamura, M. Usefulness of Plasma Exosomal MicroRNA-451a as a Noninvasive Biomarker for Early Prediction of Recurrence and Prognosis of Non-Small Cell Lung Cancer. Oncology 2018, 94, 311–323. [Google Scholar] [CrossRef] [PubMed]
- Dejima, H.; Iinuma, H.; Kanaoka, R.; Matsutani, N.; Kawamura, M. Exosomal microRNA in plasma as a non-invasive biomarker for the recurrence of non-small cell lung cancer. Oncol. Lett. 2017, 13, 1256–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, X.X.; Yu, R.; Wu, X.; Wu, S.Y.; Pi, C.; Chen, Z.H.; Zhang, X.C.; Gao, C.Y.; Shao, Y.W.; Liu, L.; et al. Correlation of plasma exosomal microRNAs with the efficacy of immunotherapy in EGFR/ALK wild-type advanced non-small cell lung cancer. J. Immunother. Cancer 2020, 8, e000376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eichelser, C.; Stuckrath, I.; Muller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 2014, 5, 9650–9663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hannafon, B.N.; Trigoso, Y.D.; Calloway, C.L.; Zhao, Y.D.; Lum, D.H.; Welm, A.L.; Zhao, Z.J.; Blick, K.E.; Dooley, W.C.; Ding, W.Q. Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Res. 2016, 18, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoshikawa, M.; Iinuma, H.; Umemoto, Y.; Yanagisawa, T.; Matsumoto, A.; Jinno, H. Exosome-Encapsulated microRNA-223-3p as a minimally invasive biomarker for the early detection of invasive breast cancer. Oncol. Lett. 2018, 15, 9584–9592. [Google Scholar] [CrossRef] [Green Version]
- Gong, C.; Tian, J.; Wang, Z.; Gao, Y.; Wu, X.; Ding, X.; Qiang, L.; Li, G.; Han, Z.; Yuan, Y.; et al. Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified microRNA 159 for triple-negative breast cancer therapy. J. Nanobiotechnol. 2019, 17, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, S.; Yin, Y.; Jin, G.; Li, D.; Li, M.; Hu, Y.; Feng, Y.; Liu, Y.; Bian, Z.; Wang, X. Exosome-mediated delivery of miR-204-5p inhibits tumor growth and chemoresistance. Cancer Med. 2020, 9, 5989–5998. [Google Scholar] [CrossRef] [PubMed]
- Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 2020, 18, 10. [Google Scholar] [CrossRef] [PubMed]
- Dai, J.; Escara-Wilke, J.; Keller, J.M.; Jung, Y.; Taichman, R.S.; Pienta, K.J.; Keller, E.T. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J. Exp. Med. 2019, 216, 2883–2899. [Google Scholar] [CrossRef]
- Takahara, K.; Ii, M.; Inamoto, T.; Nakagawa, T.; Ibuki, N.; Yoshikawa, Y.; Tsujino, T.; Uchimoto, T.; Saito, K.; Takai, T. microRNA-145 mediates the inhibitory effect of adipose tissue-derived stromal cells on prostate cancer. Stem. Cells Dev. 2016, 25, 1290–1298. [Google Scholar] [CrossRef]
- Lässer, C. Exosomes in diagnostic and therapeutic applications: Biomarker, vaccine and RNA interference delivery vehicle. Exp. Opin. Biol. Ther. 2015, 15, 103–117. [Google Scholar] [CrossRef]
- Greco, K.A.; Franzen, C.A.; Foreman, K.E.; Flanigan, R.C.; Kuo, P.C.; Gupta, G.N. PLK-1 silencing in bladder cancer by siRNA delivered with exosomes. Urology 2016, 91, 241.e1–241.e7. [Google Scholar] [CrossRef]
- Smyth, T.J.; Redzic, J.S.; Graner, M.W.; Anchordoquy, T.J. Examination of the specificity of tumor cell derived exosomes with tumor cells in vitro. Biochimica et Biophysica Acta Biomembr. 2014, 1838, 2954–2965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Zhuang, X.; Zhang, S.; Deng, Z.-B.; Grizzle, W.; Miller, D.; Zhang, H.-G. Exosomes are endogenous nanoparticles that can deliver biological information between cells. Adv. Drug Deliv. Rev. 2013, 65, 342–347. [Google Scholar] [CrossRef] [PubMed]
- Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.-S.; Roh, T.-Y.; Park, J.; Nilsson, J.; Lotvall, J.; Kim, Y.-K.; Gho, Y.S. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 2013, 7, 7698–7710. [Google Scholar] [CrossRef]
- Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef]
- Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef]
- Lee, J.; Kim, J.; Jeong, M.; Lee, H.; Goh, U.; Kim, H.; Kim, B.; Park, J.-H. Liposome-Based engineering of cells to package hydrophobic compounds in membrane vesicles for tumor penetration. Nano Lett. 2015, 15, 2938–2944. [Google Scholar] [CrossRef]
- Bovy, N.; Blomme, B.; Frères, P.; Dederen, S.; Nivelles, O.; Lion, M.; Carnet, O.; Martial, J.A.; Noël, A.; Thiry, M. Endothelial exosomes contribute to the antitumor response during breast cancer neoadjuvant chemotherapy via microRNA transfer. Oncotarget 2015, 6, 10253. [Google Scholar] [CrossRef] [Green Version]
- Didiot, M.-C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R. Exosome-Mediated delivery of hydrophobically modified siRNA for huntingtin mRNA silencing. Mol. Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [Green Version]
- Wahlgren, J.; Karlson, T.D.L.; Brisslert, M.; Vaziri, S.F.; Telemo, E.; Sunnerhagen, P.; Valadi, H. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012, 40, e130. [Google Scholar] [CrossRef] [Green Version]
- Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef] [PubMed]
- Ohno, S.-I.; Takanashi, M.; Sudo, K.; Ueda, S.; Ishikawa, A.; Matsuyama, N.; Fujita, K.; Mizutani, T.; Ohgi, T.; Ochiya, T. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 2013, 21, 185–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.K.; Finniss, S.; Cazacu, S.; Bucris, E.; Ziv-Av, A.; Xiang, C.; Bobbitt, K.; Rempel, S.A.; Hasselbach, L.; Mikkelsen, T. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget 2013, 4, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katakowski, M.; Buller, B.; Zheng, X.; Lu, Y.; Rogers, T.; Osobamiro, O.; Shu, W.; Jiang, F.; Chopp, M. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013, 335, 201–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Brien, K.; Lowry, M.C.; Corcoran, C.; Martinez, V.G.; Daly, M.; Rani, S.; Gallagher, W.M.; Radomski, M.W.; MacLeod, R.A.; O’Driscoll, L. miR-134 in extracellular vesicles reduces triple-negative breast cancer aggression and increases drug sensitivity. Oncotarget 2015, 6, 32774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’brien, K.; Khan, S.; Gilligan, K.; Zafar, H.; Lalor, P.; Glynn, C.; O’Flatharta, C.; Ingoldsby, H.; Dockery, P.; De Bhulbh, A. Employing mesenchymal stem cells to support tumor-targeted delivery of extracellular vesicle (EV)-encapsulated microRNA-379. Oncogene 2018, 37, 2137–2149. [Google Scholar] [CrossRef] [Green Version]
- Shtam, T.A.; Kovalev, R.A.; Varfolomeeva, E.Y.; Makarov, E.M.; Kil, Y.V.; Filatov, M.V. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun. Signal. 2013, 11, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Shimbo, K.; Miyaki, S.; Ishitobi, H.; Kato, Y.; Kubo, T.; Shimose, S.; Ochi, M. Exosome-Formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem. Biophys. Res. Commun. 2014, 445, 381–387. [Google Scholar] [CrossRef]
- Lou, G.; Song, X.; Yang, F.; Wu, S.; Wang, J.; Chen, Z.; Liu, Y. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J. Hematol. Oncol. 2015, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; San Chan, Y.; Wei, L.; Chin, S.M.; Azad, A. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 1–15. [Google Scholar] [CrossRef]
- Meyer, C.; Losacco, J.; Stickney, Z.; Li, L.; Marriott, G.; Lu, B. Pseudotyping exosomes for enhanced protein delivery in mammalian cells. Int. J. Nanomed. 2017, 12, 3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sterzenbach, U.; Putz, U.; Low, L.-H.; Silke, J.; Tan, S.-S.; Howitt, J. Engineered exosomes as vehicles for biologically active proteins. Mol. Ther. 2017, 25, 1269–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, Y.; Schorey, J.S. Targeting soluble proteins to exosomes using a ubiquitin tag. Biotechnol. Bioeng. 2016, 113, 1315–1324. [Google Scholar] [CrossRef]
CT Identification | Aim of Study | Source of Exosomes | Associated Markers | Promoted by |
---|---|---|---|---|
NCT04182893 | Identification of benign and malignant pulmonary nodules | Blood and alveolar lavage fluid | Exosomal RNA | Shanghai Chest Hospital, Shanghai, China |
NCT04499794 | Study of exosome EML4-ALK fusion in NSCLC clinical diagnosis | Plasma | EML4-ALK fusion | Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China |
NCT03032913 | Onco-exosome quantification in diagnosis of pancreatic cancer | Blood | Onco-exosomes | CHU de Bordeaux, Bordeaux, France |
NCT04529915 | Early diagnosis of lung cancer using blood plasma-derived exosomes | Blood | Exosomal proteins | Korea University Guro Hospital, Seoul, Republic of Korea |
NCT04394572 | Identification of new diagnostic protein markers for colorectal cancer | Blood | Exosomal proteins | CHU Reims, Reims, France |
NCT04155359 | Diagnosis of bladder cancer in hematuria patients | Urine | sncRNAs | Integrated Medical Professionals, Farmingdale, New York, United States |
NCT03974204 | Analysis of exosomes in cerebrospinal fluid for breast cancer patients | Cerebrospinal fluid and blood | Exosomal proteins | Centre Hospitalier Régional Universitaire de Lille, Lille, Hauts-de-France, France |
NCT03830619 | Exosomal long noncoding RNAs as potential biomarkers for lung cancer diagnosis | Plasma | Exosomal lncRNAs | Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China |
NCT03562715 | Role of exosomal miRNAs 136, 494 and 495 in pre-eclampsia diagnosis | Blood | miRNAs 136, 494 and 495 | Cairo University, Cairo, Egypt |
NCT03415984 | Estimation of age-related macular degeneration (ARMD) prevalence in Parkinson’s patients | Not defined | Pro-inflammatory components | Fondation Ophtalmologique Adolphe de Rothschild, Paris, France |
NCT04523389 | Analysis of extracellular vesicle contents as biomarkers in colorectal cancer patients | Blood | miRNAs | CHU Dijon Bourgogne, Dijon, France |
NCT03108677 | Evaluation of circulating exosomal RNA as biomarker for lung metastases of primary high-grade osteosarcoma | Blood | Exosomal RNA | Ruijin Hospital, Shanghai, China |
NCT04258735 | Genomic analysis of metastatic breast cancer patients | Blood | ctDNA and RNA | Sungkyunkwan University School of Medicine, Seoul, Republic of Korea |
NCT04053855 | Evaluation of urinary exosomes presence from clear cell renal cell carcinoma | Urine | CD9,CD63,CD81,CA9 and VGEFR2 | CHU Saint-Etienne, Saint-Étienne, France |
NCT04315753 | assessment of exosomes’ role in improving lung cancer management and early detection | Blood | Exosomal antigens | Istituto Clinico Humanitas, Rozzano, Milano, Italy |
NCT04459182 | evaluation of miRNA in exosomes in obese and OSA patients with endothelial dysfunction | Not defined | miRNA | University Hospital, Angers, France |
NCT04556916 | Early detection of prostate cancer | Blood | Exosomes | University Hospital, Montpellier, France |
NCT02464930 | Evaluation of microRNA expression in blood and cytology for detecting Barrett’s esophagus and associated neoplasia | Bile and serum | miRNAs 192-5p, 215-5p and 194-5p | Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States |
NCT03800121 | Study of exosomes in monitoring patients with sarcoma | Blood | Exosomal RNA and proteins | Centre Georges François Leclerc, Dijon, France |
NCT04154332 | Defining the functional role of exosomes in the development of preeclampsia leading to cardiovascular remodeling | Urine and Blood | Exosome abnormalities | University of Alabama,, Birmingham, Alabama, United States |
NCT03102268 | Characterization of cholangiocarcinoma-derived exosomal ncRNAs | Plasma | Non-coding RNAs | Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China |
NCT03581435 | Study of circulating exosome proteomics in gallbladder carcinoma patients | Blood | Exosomal proteins | Xinhua Hospital, Shanghai, China |
NCT03738319 | Analysis of non-coding RNAs in epithelia ovarian cancer | Blood | miRNA and lncRNA | Peking Union Medical College Hospital, Beijing, China |
NCT03911999 | Investigating relationship between urinary exosomes and aggressiveness of prostate cancer | Urine | Exosomal miRNA | Prince of Wales Hospital, Hong Kong, Hong Kong |
NCT04120272 | Search for biomarkers for early detection and prevention of delirium | Urine and blood | miRNA | College of Nursing, Yonsei University, Seoul, Republic of Korea |
NCT03419000 | Evaluation of microRNAs as biomarkers of respiratory dysfunction in refractory epilepsy | Blood | miRNAs | Hospices Civils de Lyon, Bron, France |
NCT04534647 | Assessment of correlation between serological and urinary biomarkers and systemic lupus erythematosus | Blood and urine | Exosomes | Liga Panamericana de Asociaciones de Reumatologia (PANLAR), Rosario, Argentina |
NCT02147418 | Exosome testing as screening modality for human papillomavirus-positive oropharyngeal squamous cell carcinoma | Saliva | Exosomal proteins | New Mexico Cancer Care Alliance, Albuquerque, New Mexico, United States |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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
Aheget, H.; Mazini, L.; Martin, F.; Belqat, B.; Marchal, J.A.; Benabdellah, K. Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers 2021, 13, 84. https://doi.org/10.3390/cancers13010084
Aheget H, Mazini L, Martin F, Belqat B, Marchal JA, Benabdellah K. Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers. 2021; 13(1):84. https://doi.org/10.3390/cancers13010084
Chicago/Turabian StyleAheget, Houssam, Loubna Mazini, Francisco Martin, Boutaïna Belqat, Juan Antonio Marchal, and Karim Benabdellah. 2021. "Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases" Cancers 13, no. 1: 84. https://doi.org/10.3390/cancers13010084
APA StyleAheget, H., Mazini, L., Martin, F., Belqat, B., Marchal, J. A., & Benabdellah, K. (2021). Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers, 13(1), 84. https://doi.org/10.3390/cancers13010084