245
Views
2
CrossRef citations to date
0
Altmetric
Review

Role of exosomes and its emerging therapeutic applications in the pathophysiology of non-infectious diseases

, & ORCID Icon
Pages 534-548 | Received 24 Mar 2022, Accepted 12 Apr 2022, Published online: 10 May 2022

References

  • Ailawadi, S., et al., 2015. Pathologic function and therapeutic potential of exosomes in cardiovascular disease. Biochimica et biophysica acta (bba) - molecular basis of disease, 1852 (1), 1–11.
  • Alvarez, M.L., et al., 2012. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney international, 82 (9), 1024–1032.
  • Alvarez-Erviti, L., et al., 2011. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature biotechnology, 29 (4), 341–345.
  • Ananbeh, H., Vodicka, P., and Skalnikova, H.K., 2021. Emerging roles of exosomes in Huntington’s disease. International journal of molecular sciences, 22 (8), 4085.
  • Arslan, F., et al., 2013. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem cell research, 10 (3), 301–312.
  • Arthur, A., et al., 2008. Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues. Stem cells, 26 (7), 1787–1795.
  • Atay, S. and Godwin, A.K., 2014. Tumor-derived exosomes: a message delivery system for tumor progression. Communicative and integrative biology, 7 (0), e28231.
  • Azmi, A.S., Bao, B., and Sarkar, F.H., 2013. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer metastasis reviews, 32 (3-4), 623–642.
  • Baglio, S.R., et al., 2015. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem cell research & therapy, 6 (1), 127.
  • Barile, L., et al., 2014. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovascular research, 103 (4), 530–541.
  • Bartel, D.P., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell, 116 (2), 281–297.
  • Bellin, G., et al., 2019. Exosome in cardiovascular diseases: a complex world full of hope. Cells, 8 (2), 166.
  • Berindan-Neagoe, I., et al., 2014. MicroRNAome genome: a treasure for cancer diagnosis and therapy. CA: a cancer journal for clinicians, 64 (5), 311–336.
  • Besse, B., et al., 2016. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology, 5 (4), e1071008.
  • Boese, A.S., et al., 2016. MicroRNA abundance is altered in synaptoneurosomes during prion disease. Molecular and cellular neurosciences, 71, 13–24.
  • Böing, A.N., et al., 2014. Single-step isolation of extracellular vesicles by size-exclusion chromatography. Journal of extracellular vesicles, 3 (1),23430.
  • Boulanger, C.M., et al., 2017. Extracellular vesicles in coronary artery disease. Nature reviews cardiology, 14 (5), 259–272.
  • Buonfiglioli, A., et al., 2019. let-7 MicroRNAs regulate microglial function and suppress glioma gfrowth through toll-like receptor 7. Cell reports, 29 (11), 3460–3471.e7.
  • Camussi, G., et al., 2010. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney international, 78 (9), 838–848.
  • Chakraborty, C., et al., 2017. Therapeutic miRNA and siRNA: moving from bench to clinic as next generation medicine. Molecular therapy - nucleic acids, 8, 132–143.
  • Challagundla, K.B., et al., 2015. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. Journal of the national cancer institute, 107 (7), djv135.
  • Chalmin, F., et al., 2010. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. Journal of Clinical Investigation, 120 (2), 457–471.
  • Chargaff, E. and West, R., 1946. The biological significance of the thromboplastic protein of blood. The Journal of biological chemistry, 166 (1), 189–197.
  • Chen, C., et al., 2010. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab on a chip, 10 (4), 505–511.
  • Cherfils-Vicini, J., et al., 2010. Triggering of TLR7 and TLR8 expressed by human lung cancer cells induces cell survival and chemoresistance. The journal of clinical investigation, 120 (4), 1285–1297.
  • Clayton, A., et al., 2007. Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer research, 67 (15), 7458–7466.
  • Cogswell, J.P., et al., 2008. Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. Journal of alzheimer’s disease, 14 (1), 27–41.
  • Colombo, M., Raposo, G., and Théry, C., 2014. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annual review of cell and developmental biology, 30, 255–289.
  • Coumans, F.A.W., et al., 2017. Methodological guidelines to study extracellular vesicles. Circulation research, 120 (10), 1632–1648.
  • Crotti, A., et al., 2019. BIN1 favors the spreading of Tau via extracellular vesicles. Scientific reports, 9 (1), 9477–9477.
  • Danzer, K.M., et al., 2012. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Molecular neurodegeneration, 7 (1), 1–18.
  • de Jong, O.G., et al., 2012. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. Journal of extracellular vesicles, 1 (1), 18396.
  • Di Leva, G., Garofalo, M., and Croce, C.M., 2014. MicroRNAs in cancer. Annual review of pathology, 9, 287–314.
  • Díez-Planelles, C., et al., 2016. Circulating microRNAs in Huntington’s disease: emerging mediators in metabolic impairment. Pharmacological research, 108, 102–110.
  • Dutta, R., et al., 2017. Myocyte-derived Hsp90 modulates collagen upregulation via biphasic activation of STAT-3 in fibroblasts during cardiac hypertrophy. Molecular and cellular biology, 37 (6), e00611–16.
  • El Andaloussi, S., et al., 2013. Extracellular vesicles: biology and emerging therapeutic opportunities. Nature reviews. Drug discovery, 12 (5), 347–357.
  • Fais, S., 2013. NK cell-released exosomes: natural nanobullets against tumors. Oncoimmunology, 2 (1), e22337.
  • Frydrychowicz, M., et al., 2015. Exosomes-structure, biogenesis and biological role in non-small-cell lung cancer. Scandinavian journal of immunology, 81 (1), 2–10.
  • Fu, H., et al., 2018. Exosomal TRIM3 is a novel marker and therapy target for gastric cancer. Journal of experimental & clinical cancer research, 37 (1), 162.
  • Gardiner, C., et al., 2016. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. Journal of extracellular vesicles, 5 (1), 32945.
  • Garrido, C., et al., 2001. Heat shock proteins: endogenous modulators of apoptotic cell death. Biochemical and biophysical research communications, 286 (3), 433–442.
  • Goloudina, A.R., Demidov, O.N., and Garrido, C., 2012. Inhibition of HSP70: a challenging anti-cancer strategy. Cancer letters, 325 (2), 117–124.
  • Gorabi, A.M., et al., 2019. The therapeutic potential of mesenchymal stem cell–derived exosomes in treatment of neurodegenerative diseases. Molecular neurobiology, 56 (12), 8157–8167.
  • Goutagny, N., et al., 2012. Targeting pattern recognition receptors in cancer immunotherapy. Targeted oncology, 7 (1), 29–54.
  • Grammatikakis, I., Gorospe, M., and Abdelmohsen, K., 2013. Modulation of cancer traits by tumor suppressor microRNAs. International journal of molecular sciences, 14 (1), 1822–1842.
  • Gray, W.D., et al., 2015. Identification of therapeutic covariant microRNA clusters in hypoxia-treated cardiac progenitor cell exosomes using systems biology. Circulation research, 116 (2), 255–263.
  • Gu, X., et al., 2015. Improved vaccine efficacy of tumor exosome compared to tumor lysate loaded dendritic cells in mice. International journal of cancer, 136 (4), E74–E84.
  • Guermonprez, P., et al., 2002. Antigen presentation and T cell stimulation by dendritic cells. Annual review of immunology, 20, 621–667.
  • Gupta, S. and Knowlton, A.A., 2007. HSP60 trafficking in adult cardiac myocytes: role of the exosomal pathway. American journal of physiology heart and circulatory physiology, 292 (6), H3052–H3056.
  • Hanahan, D. and Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell, 144 (5), 646–674.
  • Haney, M.J., et al., 2015. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. Journal of Controlled Release, 207, 18–30.
  • Hannafon, B.N. and Ding, W.-Q., 2015. Cancer stem cells and exosome signaling. Stem cell investigation, 2, 11–11.
  • Harding, C. and Stahl, P., 1983. Transferrin recycling in reticulocytes: pH and iron are important determinants of ligand binding and processing. Biochemical and biophysical research communications, 113 (2), 650–658.
  • Harding, C.V., Heuser, J.E., and Stahl, P.D., 2013. Exosomes: looking back three decades and into the future. Journal of cell biology, 200 (4), 367–371.
  • Hemler, M.E., 2014. Tetraspanin proteins promote multiple cancer stages. Nature reviews cancer, 14 (1), 49–60.
  • Henningsson, F., et al., 2011. Ige-mediated enhancement of CD4+ T cell responses in mice requires antigen presentation by CD11c + cells and not by B cells. PLoS one, 6 (7), e21760.
  • Hergenreider, E., et al., 2012. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nature cell biology, 14 (3), 249–256.
  • Hessvik, N.P. and Llorente, A., 2018. Current knowledge on exosome biogenesis and release. Cellular and molecular life sciences, 75 (2), 193–208.
  • Hong, X., Schouest, B., and Xu, H., 2017. Effects of exosome on the activation of CD4+ T cells in rhesus macaques: a potential application for HIV latency reactivation. Scientific reports, 7 (1), 1–9.
  • Hoshino, A., et al., 2015. Tumour exosome integrins determine organotropic metastasis. Nature, 527 (7578), 329–335.
  • Hosseini, M., et al., 2017. Exosome-encapsulated microRNAs as potential circulating biomarkers in colon cancer. Current pharmaceutical design, 23 (11), 1705–1709.
  • Huntzinger, E. and Izaurralde, E., 2011. Gene silencing by microRNAs: contributions of translational repression and mRNA decay. Nature reviews genetics, 12 (2), 99–110.
  • Hurwitz, S.N., et al., 2016. Proteomic profiling of NCI-60 extracellular vesicles uncovers common protein cargo and cancer type-specific biomarkers. Oncotarget, 7 (52), 86999–87015.
  • Inamura, K., 2017. Diagnostic and therapeutic potential of microRNAs in lung cancer. Cancers, 9 (12), 49.
  • Isola, A.L. and Chen, S., 2017. Extracellular vesicles: important players in immune homeostasis. Annals of translational medicine, 5 (S1), S16–S4108.
  • Javeed, N. and Mukhopadhyay, D., 2017. Exosomes and their role in the micro-/macro-environment: a comprehensive review. Journal of biomedical research, 31, 386–394.
  • Jiang, Y., et al., 2019. Serum secreted miR-137-containing exosomes affects oxidative stress of neurons by regulating OXR1 in Parkinson’s disease. Brain research, 1722, 146331.
  • Junn, E., et al., 2009. Repression of alpha-synuclein expression and toxicity by microRNA-7. Proceedings of the national academy of sciences of the United States of America, 106 (31), 13052–13057.
  • Kamerkar, S., et al., 2017. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 546 (7659), 498–503.
  • Kang, D., et al., 2008. Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry. Journal of proteome research, 7 (8), 3475–3480.
  • Katsu, M., et al., 2019. MicroRNA expression profiles of neuron-derived extracellular vesicles in plasma from patients with amyotrophic lateral sclerosis. Neuroscience letters, 708, 134176.
  • Kimura, H., et al., 2019. CKAP4, a DKK1 receptor, is a biomarker in exosomes derived from pancreatic cancer and a molecular target for therapy. Clinical cancer research, 25 (6), 1936–1947.
  • Kitamura, Y., et al., 2018. Proteomic profiling of exosomal proteins for blood-based biomarkers in Parkinson’s disease. Neuroscience, 392, 121–128.
  • Koniusz, S., et al., 2016. Extracellular vesicles in physiology, pathology, and therapy of the immune and central nervous system, with focus on extracellular vesicles derived from mesenchymal stem cells as therapeutic tools. Frontiers in cellular neuroscience, 10, 109.
  • Kooijmans, S.A.A., et al., 2012. Exosome mimetics: a novel class of drug delivery systems. International journal of nanomedicine, 7, 1525–1541.
  • Kowal, J., et al., 2016. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proceedings of the national academy of sciences of the united states of America, 113 (8), E968–E977.
  • Kowal, J., Tkach, M., and Théry, C., 2014. Biogenesis and secretion of exosomes. Current opinion in cell biology, 29, 116–125.
  • Kunigelis, K.E. and Graner, M.W., 2015. The dichotomy of tumor exosomes (TEX) in cancer immunity: is it all in the ConTEXt? Vaccines, 3 (4), 1019–1051.
  • Kuwabara, Y., et al., 2011. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circulation cardiovascular genetics, 4 (4), 446–454.
  • Lee, K., et al., 2015. Acoustic purification of extracellular microvesicles. ACS nano, 9 (3), 2321–2327.
  • Li, M., Liao, L., and Tian, W., 2020. Extracellular vesicles derived from apoptotic cells: an essential link between death and regeneration. Frontiers in cell and developmental biology, 8, 1063.
  • Lianos, G.D., et al., 2015. The role of heat shock proteins in cancer. Cancer letters, 360 (2), 114–118.
  • Lin, J., et al., 2015. Exosomes: novel biomarkers for clinical diagnosis. The scientific world journal, 2015, 1–8.
  • Liu, J., et al., 2014. Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. International journal of molecular sciences, 15 (1), 758–773.
  • Liu, W., et al., 2019. Role of exosomes in central nervous system diseases. Frontiers in molecular neuroscience, 12, 240.
  • Liu, Y., et al., 2016. STAT3-regulated exosomal miR-21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer letters, 370 (1), 125–135.
  • Lobb, R.J., Lima, L.G., and Möller, A., 2017. Exosomes: key mediators of metastasis and pre-metastatic niche formation. Seminars in cell & developmental biology, 67, 3–10.
  • Loyer, X., et al., 2014. Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circulation research, 114 (3), 434–443.
  • Luan, X., et al., 2017. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta pharmacologica sinica, 38 (6), 754–763.
  • Luzio, J.P., Gray, S.R., and Bright, N.A., 2010. Endosome-lysosome fusion. Biochemical society transactions, 38 (6), 1413–1416.
  • Madison, M.N. and Okeoma, C.M., 2015. Exosomes: implications in HIV-1 pathogenesis. Viruses, 7 (7), 4093–4118.
  • Maeda, H. and Khatami, M., 2018. Analyses of repeated failures in cancer therapy for solid tumors: poor tumor‐selective drug delivery, low therapeutic efficacy and unsustainable costs. Clinical and translational medicine, 7 (1), 11.
  • Malik, Z.A., et al., 2013. Cardiac myocyte exosomes: stability, HSP60, and proteomics. American journal of physiology heart and circulatory physiology, 304 (7), 954–H965.
  • Martinez, M.C. and Andriantsitohaina, R., 2011. Microparticles in angiogenesis: therapeutic potential. Circulation research, 109 (1), 110–119.
  • Marton, A., et al., 2012. Melanoma cell-derived exosomes alter macrophage and dendritic cell functions in vitro. Immunology letters, 148 (1), 34–38.
  • McKelvey, K.J., et al., 2015. Exosomes: mechanisms of uptake. Journal of circulating biomarkers, 4 (1), 7.
  • Melo, S.A., et al., 2014. Cancer exosomes perform cell-independent MicroRNA biogenesis and promote tumorigenesis. Cancer cell, 26 (5), 707–721.
  • Meng, F., et al., 2016. Psmir: a database of potential associations between small molecules and miRNAs. Scientific reports, 6 (1), 1–6.
  • Moore, C., et al., 2017. The emerging role of exosome and microvesicle- (EMV-) based cancer therapeutics and immunotherapy. International journal of cancer, 141 (3), 428–436.
  • Morel, O., et al., 2008. Platelet microparticles and vascular cells interactions: a checkpoint between the haemostatic and thrombotic responses. Platelets, 19 (1), 9–23.
  • Motoyama, K., et al., 2008. Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clinical cancer research, 14 (8), 2334–2340.
  • Mulcahy, L.A., Pink, R.C., and Carter, D.R., 2014. Routes and mechanisms of extracellular vesicle uptake. Journal of extracellular vesicles, 3 (1),24641.
  • Multhoff, G., et al., 2001. A 14-mer Hsp70 peptide stimulates natural killer (NK) cell activity. Cell stress & chaperones, 6 (4), 337–344. > 2.0.CO;2.
  • Niu, L., et al., 2019. Tumor-derived exosomal proteins as diagnostic biomarkers in non-small cell lung cancer. Cancer science, 110 (1), 433–442.
  • Nordin, J.Z., et al., 2015. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine: nanotechnology, biology, and medicine, 11 (4), 879–883.
  • Öhlund, D., Elyada, E., and Tuveson, D., 2014. Fibroblast heterogeneity in the cancer wound. Journal of experimental medicine, 211 (8), 1503–1523.
  • Ohmichi, T., et al., 2019. Quantification of brain-derived extracellular vesicles in plasma as a biomarker to diagnose Parkinson’s and related diseases. Parkinsonism & related disorders, 61, 82–87.
  • Ortega, A., et al., 2020. Exosomes as drug delivery systems: endogenous nanovehicles for treatment of systemic lupus erythematosus. Pharmaceutics 2021, 13 (1), 3.
  • Pace Di Pace, A.L., et al., 2020. Characterization of human NK cell-derived exosomes: role of DNAM1 receptor in exosome-mediated cytotoxicity against umor. Cancers, 12 (3), 661.
  • Pan, B.T. and Johnstone, R.M., 1983. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell, 33 (3), 967–978.
  • Phan, T.K., Ozkocak, D.C., and Poon, I.K.H., 2020. Unleashing the therapeutic potential of apoptotic bodies. Biochemical society transactions, 48 (5), 2079–2088.
  • Piao, Y.J., et al., 2018. Breast cancer cell-derived exosomes and macrophage polarization are associated with lymph node metastasis. Oncotarget, 9 (7), 7398–7410.
  • Pinto, S., et al., 2017. Exosomes from NSC-34 cells transfected with hSOD1-G93A are enriched in miR-124 and drive alterations in microglia phenotype. Frontiers in neuroscience, 11, 273.
  • Piper, R.C. and Katzmann, D.J., 2007. Biogenesis and function of multivesicular bodies. Annual review of cell and developmental biology, 23 (1), 519–547.
  • Pitt, J.M., et al., 2014. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. Journal of immunology, 193 (3), 1006–1011.
  • Pitt, J.M., Kroemer, G., and Zitvogel, L., 2016. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. Journal of clinical investigation, 126 (4), 1139–1143.
  • Poon, I.K.H., et al., 2019. Moving beyond size and phosphatidylserine exposure: evidence for a diversity of apoptotic cell-derived extracellular vesicles in vitro. Journal of extracellular vesicles, 8 (1), 1608786–1608786.
  • Pusic, A.D., et al., 2014. IFNγ-stimulated dendritic cell exosomes as a potential therapeutic for remyelination. Journal of neuroimmunology, 266 (1–2), 12–23.
  • Qi, Y., et al., 2020. Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug delivery, 27 (1), 745–755.
  • Quail, D.F. and Joyce, J.A., 2013. Microenvironmental regulation of tumor progression and metastasis. Nature medicine, 19 (11), 1423–1437.
  • Raposo, G., et al., 1996. B lymphocytes secrete antigen-presenting vesicles. The journal of experimental medicine, 183 (3), 1161–1172.
  • Ratajczak, J., et al., 2006. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia, 20 (9), 1487–1495.
  • Reátegui, E., et al., 2018. Engineered nanointerfaces for microfluidic isolation and molecular profiling of tumor-specific extracellular vesicles. Nature communications, 9 (1), 175.
  • Rekker, K., et al., 2014. Comparison of serum exosome isolation methods for microRNA profiling. Clinical biochemistry, 47 (1–2), 135–138.
  • Santavanond, J.P., et al., 2021. Apoptotic bodies: mechanism of formation, isolation and functional relevance. Subcellular biochemistry, 97, 61–88.
  • Sempere, L.F., et al., 2004. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome biology, 5 (3), R13.
  • Shakespear, N., et al., 2020. Astrocyte-derived exosomal microRNA miR-200a-3p prevents MPP+-induced apoptotic cell death through down-regulation of MKK4. Neurochemical research, 45 (5), 1020–1033.
  • Shao, H., et al., 2018. New technologies for analysis of extracellular vesicles. Chemical reviews, 118 (4), 1917–1950.
  • Skotland, T., Sandvig, K., and Llorente, A., 2017. Lipids in exosomes: current knowledge and the way forward. Progress in lipid research, 66, 30–41.
  • Ståhl, A. l., et al., 2019. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatric nephrology (Berlin, Germany), 34 (1), 11–30.
  • Strachan, D.C., et al., 2013. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8+ T cells. Oncoimmunology, 2 (12), e26968.
  • Sun, D., et al., 2010. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Molecular therapy: the journal of the American society of gene therapy, 18 (9), 1606–1614.
  • Sun, D., et al., 2013. Exosomes are endogenous nanoparticles that can deliver biological information between cells. Advanced drug delivery reviews, 65 (3), 342–347.
  • Sung, B.H., et al., 2015. Directional cell movement through tissues is controlled by exosome secretion. Nature communications, 6 (1), 7164.
  • Sung, J.Y., et al., 2001. Induction of neuronal cell death by Rab5A-dependent endocytosis of alpha-synuclein. The journal of biological chemistry, 276 (29), 27441–27448.
  • Tadokoro, H., et al., 2013. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. The journal of biological chemistry, 288 (48), 34343–34351.
  • Tang, K., et al., 2012. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nature communications, 3 (1), 1–11.
  • Tassew, N.G., et al., 2017. Exosomes mediate mobilization of autocrine Wnt10b to promote axonal regeneration in the injured CNS. Cell reports, 20 (1), 99–111.
  • Taylor, D.D. and Gercel-Taylor, C., 2008. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecologic oncology, 110 (1), 13–21.
  • Tayoun, T., et al., 2019. CTC-derived models: a window into the seeding capacity of circulating tumor cells (CTCs). Cells, 8 (10), 11–45.
  • Thakur, B.K., et al., 2014. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell research, 24 (6), 766–769.
  • Théry, C., et al., 2006. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current protocols in cell biology, 30 (1), 1.
  • Thind, A. and Wilson, C., 2016. Exosomal miRNAs as cancer biomarkers and therapeutic targets. Journal of extracellular vesicles, 5 (1), 31292.
  • Tian, Y., et al., 2014. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials, 35 (7), 2383–2390.
  • Tickner, J.A., et al., 2014. Functions and therapeutic roles of exosomes in cancer. Frontiers in oncology, 4, 127.
  • Tofaris, G.K., 2017. A critical assessment of exosomes in the pathogenesis and stratification of Parkinson’s disease. The journal of parkinson’s disease, 7 (4), 569–576.
  • Trams, E.G., et al., 1981. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochimica et biophysica acta, 645 (1), 63–70.
  • Valadi, H., et al., 2007. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature cell biology, 9 (6), 654–659.
  • Vanlandingham, P.A. and Ceresa, B.P., 2009. Rab7 regulates late endocytic trafficking downstream of multivesicular body biogenesis and cargo sequestration. The journal of biological chemistry, 284 (18), 12110–12124.
  • Vidal, M., Mangeat, P., and Hoekstra, D., 1997. Aggregation reroutes molecules from a recycling to a vesicle-mediated secretion pathway during reticulocyte maturation. Journal of cell science, 110 (16), 1867–1877.
  • Vlassov, A.V., et al., 2012. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochimica et biophysica acta (bba) - general subjects, 1820 (7), 940–948.
  • Vyas, N. and Dhawan, J., 2017. Exosomes: mobile platforms for targeted and synergistic signaling across cell boundaries. Cellular and molecular life sciences, 74 (9), 1567–1576.
  • Wang, K., et al., 2017. Enhanced cardioprotection by human endometrium mesenchymal stem cells driven by exosomal microRNA-21. Stem cells translational medicine, 6 (1), 209–222.
  • Wang, L., et al., 2018. Reduced exosome miR-425 and miR-744 in the plasma represents the progression of fibrosis and heart failure. The Kaohsiung journal of medical sciences, 34 (11), 626–633.
  • Wang, W. X., et al., 2008. The Expression of MicroRNA miR-107 Decreases Early in Alzheimer's disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. The journal of neuroscience: the official journal of the society for neuroscience, 28 (5), 1213–1223.
  • Wang, X., et al., 2014. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. Journal of molecular and cellular cardiology, 74, 139–150.
  • Wang, Z., et al., 2016. Exosomes in tumor microenvironment: novel transporters and biomarkers. Journal of translational medicine, 14 (1), 297.
  • Winkler, C.W., Taylor, K.G., and Peterson, K.E., 2014. Location is everything: let-7b microRNA and TLR7 signaling results in a painful TRP. Science signaling, 7 (327), e14.
  • Wollert, T. and Hurley, J.H., 2010. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature, 464 (7290), 864–869.
  • Wu, S. W., et al., 2019. CTL-derived exosomes enhance the activation of CTLs stimulated by low-affinity peptides. Frontiers in immunology, 10, 1274.
  • Wu, X., Zheng, T., and Zhang, B., 2017. Exosomes in Parkinson’s disease. Neuroscience bulletin, 33 (3), 331–338.
  • Wubbolts, R., et al., 2003. Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation. The journal of biological chemistry, 278 (13), 10963–10972.
  • Wunsch, B.H., et al., 2016. Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm. Nature nanotechnology, 11 (11), 936–940.
  • Xiao, J., et al., 2016. Cardiac progenitor cell-derived exosomes prevent cardiomyocytes apoptosis through exosomal miR-21 by targeting PDCD4. Cell death & disease, 7 (6), e2277–e2277.
  • Xu, X., Lai, Y., and Hua, Z.C., 2019. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Bioscience reports, 39 (1), BSR20180992.
  • Yan, M., et al., 2015. miR-21-3p regulates cardiac hypertrophic response by targeting histone deacetylase-8. Cardiovascular research, 105 (3), 340–352.
  • Yang, T., et al., 2015. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in danio rerio. Pharmaceutical research, 32 (6), 2003–2014.
  • Ye, S.B., et al., 2014. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget, 5 (14), 5439–5452.
  • Yoo, C.E., et al., 2012. A direct extraction method for microRNAs from exosomes captured by immunoaffinity beads. Analytical biochemistry, 431 (2), 96–98.
  • Yu, X., et al., 2012. Mechanism of TNF-α autocrine effects in hypoxic cardiomyocytes: initiated by hypoxia inducible factor 1α, presented by exosomes. Journal of molecular and cellular cardiology, 53 (6), 848–857.
  • Yuyama, K., et al., 2012. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-β by microglia. The journal of biological chemistry, 287 (14), 10977–10989.
  • Zamani, P., et al., 2019. The therapeutic and diagnostic role of exosomes in cardiovascular diseases. Trends in cardiovascular medicine, 29 (6), 313–323.
  • Zhang, Y., et al., 2019. Exosomes: biogenesis, biologic function and clinical potential. Cell & bioscience, 9, 19–18.
  • Zhao, Z., et al., 2016. A microfluidic exosearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab on a chip, 16 (3), 489–496.
  • Zhou, B., et al., 2020. Application of exosomes as liquid biopsy in clinical diagnosis. Signal transduction and targeted therapy, 5 (1), 1–14.
  • Zhou, H., et al., 2019. Exosomes in ischemic heart disease: novel carriers for bioinformation. Biomedicine & pharmacotherapy = biomedecine & pharmacotherapie, 120, 109451.
  • Zhou, W., et al., 2014. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer cell, 25 (4), 501–515.
  • Zhu, S., et al., 2008. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell research, 18 (3), 350–359.
  • Zitvogel, L., et al., 1998. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nature medicine, 4 (5), 594–600.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.