Current Knowledge and Future Perspectives on Mesenchymal Stem Cell-Derived Exosomes as a New Therapeutic Agent

Hyeon Su Joo, Ju Hun Suh, Hyeon Ji Lee, Eun Song Bang, Jung Min Lee, Hyeon Su Joo, Ju Hun Suh, Hyeon Ji Lee, Eun Song Bang, Jung Min Lee

Abstract

Mesenchymal stem cells (MSCs) are on the cusp of regenerative medicine due to their differentiation capacity, favorable culture conditions, ability to be manipulated in vitro, and strong immunomodulatory activity. Recent studies indicate that the pleiotropic effects of MSCs, especially their immunomodulatory potential, can be largely attributed to paracrine factors. Exosomes, vesicles that are 30-150 nanometers in diameter that function in cell-cell communication, are one of the key paracrine effectors. MSC-derived exosomes are enriched with therapeutic miRNAs, mRNAs, cytokines, lipids, and growth factors. Emerging evidences support the compelling possibility of using MSC-derived exosomes as a new form of therapy for treating several different kinds of disease such as heart, kidney, immune diseases, neural injuries, and neurodegenerative disease. This review provides a summary of current knowledge and discusses engineering of MSC-derived exosomes for their use in translational medicine.

Keywords: exosome; mesenchymal stem cell; pre-conditioning; translational medicine.

Conflict of interest statement

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results

Figures

Figure 1
Figure 1
Exosome biogenesis and its application. (A) Exosome biogenesis. Three pathways for exosome biogenesis. (❶) ESCRT-dependent pathway and related proteins, (❷) ESCRT-independent pathway and related proteins, (❸) Direct budding of plasma membrane and related proteins. (B) Exosome components. MFGE8: milk fat globule-EGF factor 8 protein; ICAM-1: intercellular adhesion molecule 1; LAMP1,2: lysosomal-associated membrane protein 1,2; MHC I, II: major histocompatibility complex I, II. MAPK: mitogen-activated protein kinase; ERK: extracellular signal-regulated kinase; GAPDH: glyceraldehyde 3-phosphate dehydrogenase; PGK1: phosphoglycerate kinase 1. (C) Applications of exosomes. (①) Drug delivery: Therapeutic agents such as chemicals, peptides, and siRNAs can be delivered into patients. (②) Diagnosis: Exosomes derived from patients can be used for disease diagnosis. (③) Therapy: Exosomes derived from MSCs can be used to treat several diseases.
Figure 2
Figure 2
Pre-conditioning approaches for MSC to enhance the secretion and therapeutic efficacy of exosomes. LPS: lipopolysaccharides; NO: nitric oxide; DFO: deferoxamine; IFN: interferon; TNF: tumor necrosis factor; IL: interleukin; TGF: transforming growth factor; IDO: indoleamine-pyrrole 2,3-dioxygenase; HIF: hypoxia-inducible factors; miRNAs: micro RNAs; UV: ultraviolet.

References

    1. Maumus M., Jorgensen C., Noel D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie. 2013;95:2229–2234. doi: 10.1016/j.biochi.2013.04.017.
    1. Kfoury Y., Scadden D.T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16:239–253. doi: 10.1016/j.stem.2015.02.019.
    1. Su J., Chen X., Huang Y., Li W., Li J., Cao K., Cao G., Zhang L., Li F., Roberts A.I., et al. Phylogenetic distinction of iNOS and IDO function in mesenchymal stem cell-mediated immunosuppression in mammalian species. Cell Death Differ. 2014;21:388–396. doi: 10.1038/cdd.2013.149.
    1. Aggarwal S., Pittenger M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–1822. doi: 10.1182/blood-2004-04-1559.
    1. Chabannes D., Hill M., Merieau E., Rossignol J., Brion R., Soulillou J.P., Anegon I., Cuturi M.C. A role for heme oxygenase-1 in the immunosuppressive effect of adult rat and human mesenchymal stem cells. Blood. 2007;110:3691–3694. doi: 10.1182/blood-2007-02-075481.
    1. Cao W., Yang Y., Wang Z., Liu A., Fang L., Wu F., Hong J., Shi Y., Leung S., Dong C., et al. Leukemia inhibitory factor inhibits T helper 17 cell differentiation and confers treatment effects of neural progenitor cell therapy in autoimmune disease. Immunity. 2011;35:273–284. doi: 10.1016/j.immuni.2011.06.011.
    1. Augello A., Tasso R., Negrini S.M., Amateis A., Indiveri F., Cancedda R., Pennesi G. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur. J. Immunol. 2005;35:1482–1490. doi: 10.1002/eji.200425405.
    1. Sioud M., Mobergslien A., Boudabous A., Floisand Y. Evidence for the involvement of galectin-3 in mesenchymal stem cell suppression of allogeneic T-cell proliferation. Scand. J. Immunol. 2010;71:267–274. doi: 10.1111/j.1365-3083.2010.02378.x.
    1. Hsu W.T., Lin C.H., Chiang B.L., Jui H.Y., Wu K.K., Lee C.M. Prostaglandin E2 potentiates mesenchymal stem cell-induced IL-10+IFN-gamma+CD4+ regulatory T cells to control transplant arteriosclerosis. J. Immunol. 2013;190:2372–2380. doi: 10.4049/jimmunol.1202996.
    1. Groh M.E., Maitra B., Szekely E., Koc O.N. Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp. Hematol. 2005;33:928–934. doi: 10.1016/j.exphem.2005.05.002.
    1. Zhou Y., Yamamoto Y., Xiao Z., Ochiya T. The Immunomodulatory Functions of Mesenchymal Stromal/Stem Cells Mediated via Paracrine Activity. J. Clin. Med. 2019;8:1205. doi: 10.3390/jcm8071025.
    1. Zhang B., Yin Y., Lai R.C., Tan S.S., Choo A.B., Lim S.K. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014;23:1233–1244. doi: 10.1089/scd.2013.0479.
    1. Wolf P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967;13:269–288. doi: 10.1111/j.1365-2141.1967.tb08741.x.
    1. Gurunathan S., Kang M.H., Jeyaraj M., Qasim M., Kim J.H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells. 2019;8:307. doi: 10.3390/cells8040307.
    1. Maas S.L.N., Breakefield X.O., Weaver A.M. Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017;27:172–188. doi: 10.1016/j.tcb.2016.11.003.
    1. Iraci N., Leonardi T., Gessler F., Vega B., Pluchino S. Focus on Extracellular Vesicles: Physiological Role and Signalling Properties of Extracellular Membrane Vesicles. Int. J. Mol. Sci. 2016;17:171. doi: 10.3390/ijms17020171.
    1. Casado S., Lobo M., Paino C.L. Dynamics of plasma membrane surface related to the release of extracellular vesicles by mesenchymal stem cells in culture. Sci. Rep. 2017;7 doi: 10.1038/s41598-017-07265-x.
    1. Abels E.R., Breakefield X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016;36:301–312. doi: 10.1007/s10571-016-0366-z.
    1. Kahroba H., Hejazi M.S., Samadi N. Exosomes: From carcinogenesis and metastasis to diagnosis and treatment of gastric cancer. Cell. Mol. Life Sci. 2019;76:1747–1758. doi: 10.1007/s00018-019-03035-2.
    1. Li M., Rong Y., Chuang Y.S., Peng D., Emr S.D. Ubiquitin-dependent lysosomal membrane protein sorting and degradation. Mol. Cell. 2015;57:467–478. doi: 10.1016/j.molcel.2014.12.012.
    1. Piper R.C., Luzio J.P. Ubiquitin-dependent sorting of integral membrane proteins for degradation in lysosomes. Curr. Opin. Cell Biol. 2007;19:459–465. doi: 10.1016/j.ceb.2007.07.002.
    1. Baietti M.F., Zhang Z., Mortier E., Melchior A., Degeest G., Geeraerts A., Ivarsson Y., Depoortere F., Coomans C., Vermeiren E., et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012;14:677–685. doi: 10.1038/ncb2502.
    1. Livshits M.A., Khomyakova E., Evtushenko E.G., Lazarev V.N., Kulemin N.A., Semina S.E., Generozov E.V., Govorun V.M. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 2015;5 doi: 10.1038/srep17319.
    1. Street J.M., Koritzinsky E.H., Glispie D.M., Yuen P.S.T. Urine Exosome Isolation and Characterization. Methods Mol. Biol. 2017;1641:413–423.
    1. Yamashita T., Takahashi Y., Nishikawa M., Takakura Y. Effect of exosome isolation methods on physicochemical properties of exosomes and clearance of exosomes from the blood circulation. Eur. J. Pharm. Biopharm. 2016;98:1–8. doi: 10.1016/j.ejpb.2015.10.017.
    1. Li P., Kaslan M., Lee S.H., Yao J., Gao Z. Progress in Exosome Isolation Techniques. Theranostics. 2017;7:789–804. doi: 10.7150/thno.18133.
    1. Yu L.L., Zhu J., Liu J.X., Jiang F., Ni W.K., Qu L.S., Ni R.Z., Lu C.H., Xiao M.B. A Comparison of Traditional and Novel Methods for the Separation of Exosomes from Human Samples. BioMed Res. Int. 2018;2018 doi: 10.1155/2018/3634563.
    1. Xu Y., Shen L., Li F., Yang J., Wan X., Ouyang M. MicroRNA-16-5p-containing exosomes derived from bone marrow-derived mesenchymal stem cells inhibit proliferation, migration, and invasion, while promoting apoptosis of colorectal cancer cells by downregulating ITGA2. J. Cell. Physiol. 2019;234:21380–21394. doi: 10.1002/jcp.28747.
    1. Boing A.N., van der Pol E., Grootemaat A.E., Coumans F.A., Sturk A., Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles. 2014;3 doi: 10.3402/jev.v3.23430.
    1. Doyle L.M., Wang M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells. 2019;8:727. doi: 10.3390/cells8070727.
    1. De Gassart A., Geminard C., Fevrier B., Raposo G., Vidal M. Lipid raft-associated protein sorting in exosomes. Blood. 2003;102:4336–4344. doi: 10.1182/blood-2003-03-0871.
    1. Zakharova L., Svetlova M., Fomina A.F. T cell exosomes induce cholesterol accumulation in human monocytes via phosphatidylserine receptor. J. Cell. Physiol. 2007;212:174–181. doi: 10.1002/jcp.21013.
    1. Xiang X., Poliakov A., Liu C., Liu Y., Deng Z.B., Wang J., Cheng Z., Shah S.V., Wang G.J., Zhang L., et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer. 2009;124:2621–2633. doi: 10.1002/ijc.24249.
    1. Keerthikumar S., Chisanga D., Ariyaratne D., Al Saffar H., Anand S., Zhao K., Samuel M., Pathan M., Jois M., Chilamkurti N., et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016;428:688–692. doi: 10.1016/j.jmb.2015.09.019.
    1. Kim D.K., Lee J., Simpson R.J., Lotvall J., Gho Y.S. EVpedia: A community web resource for prokaryotic and eukaryotic extracellular vesicles research. Semin. Cell Dev. Biol. 2015;40:4–7. doi: 10.1016/j.semcdb.2015.02.005.
    1. Andreu Z., Yanez-Mo M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014;5:442. doi: 10.3389/fimmu.2014.00442.
    1. Conde-Vancells J., Rodriguez-Suarez E., Embade N., Gil D., Matthiesen R., Valle M., Elortza F., Lu S.C., Mato J.M., Falcon-Perez J.M. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J. Proteome Res. 2008;7:5157–5166. doi: 10.1021/pr8004887.
    1. Subra C., Grand D., Laulagnier K., Stella A., Lambeau G., Paillasse M., De Medina P., Monsarrat B., Perret B., Silvente-Poirot S., et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J. Lipid Res. 2010;51:2105–2120. doi: 10.1194/jlr.M003657.
    1. Lin J., Li J., Huang B., Liu J., Chen X., Chen X.M., Xu Y.M., Huang L.F., Wang X.Z. Exosomes: Novel biomarkers for clinical diagnosis. Sci. World J. 2015;2015:657086. doi: 10.1155/2015/657086.
    1. 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. doi: 10.1016/j.addr.2012.07.002.
    1. Choi J.Y., Kim S., Kwak H.B., Park D.H., Park J.H., Ryu J.S., Park C.S., Kang J.H. Extracellular Vesicles as a Source of Urological Biomarkers: Lessons Learned from Advances and Challenges in Clinical Applications to Major Diseases. Int. Neurourol. J. 2017;21:83–96. doi: 10.5213/inj.1734961.458.
    1. Kalani A., Tyagi A., Tyagi N. Exosomes: Mediators of neurodegeneration, neuroprotection and therapeutics. Mol. Neurobiol. 2014;49:590–600. doi: 10.1007/s12035-013-8544-1.
    1. Rabinowits G., Gercel-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. doi: 10.3816/CLC.2009.n.006.
    1. Escola J.M., Kleijmeer M.J., Stoorvogel W., Griffith J.M., Yoshie O., Geuze H.J. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J. Biol. Chem. 1998;273:20121–20127. doi: 10.1074/jbc.273.32.20121.
    1. Thery C., Regnault A., Garin J., Wolfers J., Zitvogel L., Ricciardi-Castagnoli P., Raposo G., Amigorena S. Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J. Cell Biol. 1999;147:599–610. doi: 10.1083/jcb.147.3.599.
    1. Li X., Tsibouklis J., Weng T., Zhang B., Yin G., Feng G., Cui Y., Savina I.N., Mikhalovska L.I., Sandeman S.R., et al. Nano carriers for drug transport across the blood-brain barrier. J. Drug Target. 2017;25:17–28. doi: 10.1080/1061186X.2016.1184272.
    1. Strimbu K., Tavel J.A. What are biomarkers? Curr. Opin. HIV AIDS. 2010;5:463–466. doi: 10.1097/COH.0b013e32833ed177.
    1. Stremersch S., De Smedt S.C., Raemdonck K. Therapeutic and diagnostic applications of extracellular vesicles. J. Control. Release. 2016;244:167–183. doi: 10.1016/j.jconrel.2016.07.054.
    1. Vlassov A.V., Magdaleno S., Setterquist R., Conrad R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta. 2012;1820:940–948. doi: 10.1016/j.bbagen.2012.03.017.
    1. Saeedi S., Israel S., Nagy C., Turecki G. The emerging role of exosomes in mental disorders. Transl. Psychiatry. 2019;9 doi: 10.1038/s41398-019-0459-9.
    1. Chen C.C., Liu L., Ma F., Wong C.W., Guo X.E., Chacko J.V., Farhoodi H.P., Zhang S.X., Zimak J., Ségaliny A.J., et al. Elucidation of exosome migration across the blood–brain barrier model in vitro. Cell. Mol. Bioeng. 2016;9:509–529. doi: 10.1007/s12195-016-0458-3.
    1. Gomzikova M.O., Rizvanov A.A.J.B. Current trends in regenerative medicine: From cell to cell-free therapy. Bionanoscience. 2017;7:240–245. doi: 10.1007/s12668-016-0348-0.
    1. Teng X., Chen L., Chen W., Yang J., Yang Z., Shen Z. Mesenchymal Stem Cell-Derived Exosomes Improve the Microenvironment of Infarcted Myocardium Contributing to Angiogenesis and Anti-Inflammation. Cell. Physiol. Biochem. 2015;37:2415–2424. doi: 10.1159/000438594.
    1. Feng Y., Huang W., Wani M., Yu X., Ashraf M. Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PLoS ONE. 2014;9:e88685. doi: 10.1371/journal.pone.0088685.
    1. Zhou Y., Xu H., Xu W., Wang B., Wu H., Tao Y., Zhang B., Wang M., Mao F., Yan Y., et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res. Ther. 2013;4:34. doi: 10.1186/scrt194.
    1. Willis G.R., Fernandez-Gonzalez A., Anastas J., Vitali S.H., Liu X., Ericsson M., Kwong A., Mitsialis S.A., Kourembanas S. Mesenchymal Stromal Cell Exosomes Ameliorate Experimental Bronchopulmonary Dysplasia and Restore Lung Function through Macrophage Immunomodulation. Am. J. Respir. Crit. Care Med. 2018;197:104–116. doi: 10.1164/rccm.201705-0925OC.
    1. Cho B.S., Kim J.O., Ha D.H., Yi Y.W. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res. Ther. 2018;9:187. doi: 10.1186/s13287-018-0939-5.
    1. Fang S., Xu C., Zhang Y., Xue C., Yang C., Bi H., Qian X., Wu M., Ji K., Zhao Y., et al. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-beta/SMAD2 Pathway During Wound Healing. Stem Cells Transl. Med. 2016;5:1425–1439. doi: 10.5966/sctm.2015-0367.
    1. Zhang B., Wang M., Gong A., Zhang X., Wu X., Zhu Y., Shi H., Wu L., Zhu W., Qian H., et al. HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells. 2015;33:2158–2168. doi: 10.1002/stem.1771.
    1. Nakamura Y., Miyaki S., Ishitobi H., Matsuyama S., Nakasa T., Kamei N., Akimoto T., Higashi Y., Ochi M. Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration. FEBS Lett. 2015;589:1257–1265. doi: 10.1016/j.febslet.2015.03.031.
    1. Riazifar M., Mohammadi M.R., Pone E.J., Yeri A., Lasser C., Segaliny A.I., McIntyre L.L., Shelke G.V., Hutchins E., Hamamoto A., et al. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS Nano. 2019;13:6670–6688. doi: 10.1021/acsnano.9b01004.
    1. Xiong Y., Mahmood A., Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen. Res. 2017;12:19–22. doi: 10.4103/1673-5374.198966.
    1. Zhang Y., Chopp M., Meng Y., Katakowski M., Xin H., Mahmood A., Xiong Y. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J. Neurosurg. 2015;122:856–867. doi: 10.3171/2014.11.JNS14770.
    1. Ni H., Yang S., Siaw-Debrah F., Hu J., Wu K., He Z., Yang J., Pan S., Lin X., Ye H., et al. Exosomes Derived from Bone Mesenchymal Stem Cells Ameliorate Early Inflammatory Responses Following Traumatic Brain Injury. Front. Neurosci. 2019;13:14. doi: 10.3389/fnins.2019.00014.
    1. Huang J.H., Yin X.M., Xu Y., Xu C.C., Lin X., Ye F.B., Cao Y., Lin F.Y. Systemic Administration of Exosomes Released from Mesenchymal Stromal Cells Attenuates Apoptosis, Inflammation, and Promotes Angiogenesis after Spinal Cord Injury in Rats. J. Neurotrauma. 2017;34:3388–3396. doi: 10.1089/neu.2017.5063.
    1. Lankford K.L., Arroyo E.J., Nazimek K., Bryniarski K., Askenase P.W., Kocsis J.D. Intravenously delivered mesenchymal stem cell-derived exosomes target M2-type macrophages in the injured spinal cord. PLoS ONE. 2018;13:e0190358. doi: 10.1371/journal.pone.0190358.
    1. Wang L., Pei S., Han L., Guo B., Li Y., Duan R., Yao Y., Xue B., Chen X., Jia Y. Mesenchymal Stem Cell-Derived Exosomes Reduce A1 Astrocytes via Downregulation of Phosphorylated NFkappaB P65 Subunit in Spinal Cord Injury. Cell. Physiol. Biochem. 2018;50:1535–1559. doi: 10.1159/000494652.
    1. Ruppert K.A., Nguyen T.T., Prabhakara K.S., Toledano Furman N.E., Srivastava A.K., Harting M.T., Cox C.S., Jr., Olson S.D. Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles Modify Microglial Response and Improve Clinical Outcomes in Experimental Spinal Cord Injury. Sci. Rep. 2018;8:480. doi: 10.1038/s41598-017-18867-w.
    1. Zhang Z.G., Chopp M. Exosomes in stroke pathogenesis and therapy. J. Clin. Investig. 2016;126:1190–1197. doi: 10.1172/JCI81133.
    1. Reza-Zaldivar E.E., Hernandez-Sapiens M.A., Gutierrez-Mercado Y.K., Sandoval-Avila S., Gomez-Pinedo U., Marquez-Aguirre A.L., Vazquez-Mendez E., Padilla-Camberos E., Canales-Aguirre A.A. Mesenchymal stem cell-derived exosomes promote neurogenesis and cognitive function recovery in a mouse model of Alzheimer’s disease. Neural Regen. Res. 2019;14:1626–1634.
    1. Cui G.H., Wu J., Mou F.F., Xie W.H., Wang F.B., Wang Q.L., Fang J., Xu Y.W., Dong Y.R., Liu J.R., et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J. 2018;32:654–668. doi: 10.1096/fj.201700600R.
    1. Besse B., Charrier M., Lapierre V., Dansin E., Lantz O., Planchard D., Le Chevalier T., Livartoski A., Barlesi F., Laplanche A., et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5:e1071008. doi: 10.1080/2162402X.2015.1071008.
    1. Noronha N.C., Mizukami A., Caliari-Oliveira C., Cominal J.G., Rocha J.L.M., Covas D.T., Swiech K., Malmegrim K.C.R. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res. Ther. 2019;10:131. doi: 10.1186/s13287-019-1224-y.
    1. Park J.S., Suryaprakash S., Lao Y.H., Leong K.W. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods. 2015;84:3–16. doi: 10.1016/j.ymeth.2015.03.002.
    1. Phan J., Kumar P., Hao D., Gao K., Farmer D., Wang A. Engineering mesenchymal stem cells to improve their exosome efficacy and yield for cell-free therapy. J. Extracell. Vesicles. 2018;7:1522236. doi: 10.1080/20013078.2018.1522236.
    1. Mendt M., Kamerkar S., Sugimoto H., McAndrews K.M., Wu C.C., Gagea M., Yang S., Blanko E.V.R., Peng Q., Ma X., et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight. 2018;3:99263. doi: 10.1172/jci.insight.99263.
    1. Del Piccolo N., Placone J., He L., Agudelo S.C., Hristova K. Production of plasma membrane vesicles with chloride salts and their utility as a cell membrane mimetic for biophysical characterization of membrane protein interactions. Anal. Chem. 2012;84:8650–8655. doi: 10.1021/ac301776j.
    1. Wu H., Oliver A.E., Ngassam V.N., Yee C.K., Parikh A.N., Yeh Y. Preparation, characterization, and surface immobilization of native vesicles obtained by mechanical extrusion of mammalian cells. Integr. Biol. 2012;4:685–692. doi: 10.1039/c2ib20022h.
    1. Pick H., Schmid E.L., Tairi A.P., Ilegems E., Hovius R., Vogel H. Investigating cellular signaling reactions in single attoliter vesicles. J. Am. Chem. Soc. 2005;127:2908–2912. doi: 10.1021/ja044605x.
    1. Gomzikova M.O., Zhuravleva M.N., Vorobev V.V., Salafutdinov I.I., Laikov A.V., Kletukhina S.K., Martynova E.V., Tazetdinova L.G., Ntekim A.I., Khaiboullina S.F., et al. Angiogenic Activity of Cytochalasin B-Induced Membrane Vesicles of Human Mesenchymal Stem Cells. Cells. 2019;9:95. doi: 10.3390/cells9010095.
    1. Datta A., Kim H., McGee L., Johnson A.E., Talwar S., Marugan J., Southall N., Hu X., Lal M., Mondal D., et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: A drug repurposing strategy for advanced cancer. Sci. Rep. 2018;8:8161. doi: 10.1038/s41598-018-26411-7.
    1. McAndrews K.M., Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Mol. Cancer. 2019;18:52. doi: 10.1186/s12943-019-0963-9.
    1. Laulagnier K., Grand D., Dujardin A., Hamdi S., Vincent-Schneider H., Lankar D., Salles J.P., Bonnerot C., Perret B., Record M. PLD2 is enriched on exosomes and its activity is correlated to the release of exosomes. FEBS Lett. 2004;572:11–14. doi: 10.1016/j.febslet.2004.06.082.
    1. Cheng Q., Li X., Wang Y., Dong M., Zhan F.H., Liu J. The ceramide pathway is involved in the survival, apoptosis and exosome functions of human multiple myeloma cells in vitro. Acta Pharmacol. Sin. 2018;39:561–568. doi: 10.1038/aps.2017.118.
    1. King H.W., Michael M.Z., Gleadle J.M. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer. 2012;12 doi: 10.1186/1471-2407-12-421.
    1. Parolini I., Federici C., Raggi C., Lugini L., Palleschi S., De Milito A., Coscia C., Iessi E., Logozzi M., Molinari A., et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009;284:34211–34222. doi: 10.1074/jbc.M109.041152.
    1. Kanemoto S., Nitani R., Murakami T., Kaneko M., Asada R., Matsuhisa K., Saito A., Imaizumi K. Multivesicular body formation enhancement and exosome release during endoplasmic reticulum stress. Biochem. Biophys. Res. Commun. 2016;480:166–172. doi: 10.1016/j.bbrc.2016.10.019.
    1. Savina A., Furlan M., Vidal M., Colombo M.I. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J. Biol. Chem. 2003;278:20083–20090. doi: 10.1074/jbc.M301642200.
    1. Lehmann B.D., Paine M.S., Brooks A.M., McCubrey J.A., Renegar R.H., Wang R., Terrian D.M. Senescence-associated exosome release from human prostate cancer cells. Cancer Res. 2008;68:7864–7871. doi: 10.1158/0008-5472.CAN-07-6538.
    1. Le M., Fernandez-Palomo C., McNeill F.E., Seymour C.B., Rainbow A.J., Mothersill C.E. Exosomes are released by bystander cells exposed to radiation-induced biophoton signals: Reconciling the mechanisms mediating the bystander effect. PLoS ONE. 2017;12:e0173685. doi: 10.1371/journal.pone.0173685.
    1. Aubertin K., Silva A.K., Luciani N., Espinosa A., Djemat A., Charue D., Gallet F., Blanc-Brude O., Wilhelm C. Massive release of extracellular vesicles from cancer cells after photodynamic treatment or chemotherapy. Sci. Rep. 2016;6:35376. doi: 10.1038/srep35376.
    1. Vulpis E., Cecere F., Molfetta R., Soriani A., Fionda C., Peruzzi G., Caracciolo G., Palchetti S., Masuelli L., Simonelli L., et al. Genotoxic stress modulates the release of exosomes from multiple myeloma cells capable of activating NK cell cytokine production: Role of HSP70/TLR2/NF-kB axis. Oncoimmunology. 2017;6:e1279372. doi: 10.1080/2162402X.2017.1279372.
    1. Montermini L., Meehan B., Garnier D., Lee W.J., Lee T.H., Guha A., Al-Nedawi K., Rak J. Inhibition of oncogenic epidermal growth factor receptor kinase triggers release of exosome-like extracellular vesicles and impacts their phosphoprotein and DNA content. J. Biol. Chem. 2015;290:24534–24546. doi: 10.1074/jbc.M115.679217.
    1. Hessvik N.P., Overbye A., Brech A., Torgersen M.L., Jakobsen I.S., Sandvig K., Llorente A. PIKfyve inhibition increases exosome release and induces secretory autophagy. Cell. Mol. Life Sci. 2016;73:4717–4737. doi: 10.1007/s00018-016-2309-8.
    1. Madeo M., Colbert P.L., Vermeer D.W., Lucido C.T., Cain J.T., Vichaya E.G., Grossberg A.J., Muirhead D., Rickel A.P., Hong Z., et al. Cancer exosomes induce tumor innervation. Nat. Commun. 2018;9:4284. doi: 10.1038/s41467-018-06640-0.
    1. Lee T.H., Chennakrishnaiah S., Meehan B., Montermini L., Garnier D., D’Asti E., Hou W., Magnus N., Gayden T., Jabado N., et al. Barriers to horizontal cell transformation by extracellular vesicles containing oncogenic H-ras. Oncotarget. 2016;7:51991–52002. doi: 10.18632/oncotarget.10627.
    1. 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. doi: 10.1016/j.bbrc.2014.07.109.
    1. Zhang C., Xiao X., Chen M., Aldharee H., Chen Y., Long W. Liver kinase B1 restoration promotes exosome secretion and motility of lung cancer cells. Oncol. Rep. 2018;39:376–382.
    1. Lee H.Y., Chen C.K., Ho C.M., Lee S.S., Chang C.Y., Chen K.J., Jou Y.S. EIF3C-enhanced exosome secretion promotes angiogenesis and tumorigenesis of human hepatocellular carcinoma. Oncotarget. 2018;9:13193–13205. doi: 10.18632/oncotarget.24149.
    1. Domenis R., Cifu A., Quaglia S., Pistis C., Moretti M., Vicario A., Parodi P.C., Fabris M., Niazi K.R., Soon-Shiong P., et al. Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes. Sci. Rep. 2018;8:13325. doi: 10.1038/s41598-018-31707-9.
    1. Harting M.T., Srivastava A.K., Zhaorigetu S., Bair H., Prabhakara K.S., Toledano Furman N.E., Vykoukal J.V., Ruppert K.A., Cox C.S., Jr., Olson S.D. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem Cells. 2018;36:79–90. doi: 10.1002/stem.2730.
    1. Varkouhi A.K., Jerkic M., Ormesher L., Gagnon S., Goyal S., Rabani R., Masterson C., Spring C., Chen P.Z., Gu F.X., et al. Extracellular Vesicles from Interferon-gamma-primed Human Umbilical Cord Mesenchymal Stromal Cells Reduce Escherichia coli-induced Acute Lung Injury in Rats. Anesthesiology. 2019;130:778–790. doi: 10.1097/ALN.0000000000002655.
    1. Liang Y.C., Wu Y.P., Li X.D., Chen S.H., Ye X.J., Xue X.Y., Xu N. TNF-alpha-induced exosomal miR-146a mediates mesenchymal stem cell-dependent suppression of urethral stricture. J. Cell. Physiol. 2019;234:23243–23255. doi: 10.1002/jcp.28891.
    1. Lu Z., Chen Y., Dunstan C., Roohani-Esfahani S., Zreiqat H. Priming Adipose Stem Cells with Tumor Necrosis Factor-Alpha Preconditioning Potentiates Their Exosome Efficacy for Bone Regeneration. Tissue Eng. Part A. 2017;23:1212–1220. doi: 10.1089/ten.tea.2016.0548.
    1. Song Y., Dou H., Li X., Zhao X., Li Y., Liu D., Ji J., Liu F., Ding L., Ni Y., et al. Exosomal miR-146a Contributes to the Enhanced Therapeutic Efficacy of Interleukin-1beta-Primed Mesenchymal Stem Cells Against Sepsis. Stem Cells. 2017;35:1208–1221. doi: 10.1002/stem.2564.
    1. Zhang Q., Fu L., Liang Y., Guo Z., Wang L., Ma C., Wang H. Exosomes originating from MSCs stimulated with TGF-beta and IFN-gamma promote Treg differentiation. J. Cell. Physiol. 2018;233:6832–6840. doi: 10.1002/jcp.26436.
    1. Zhang H.C., Liu X.B., Huang S., Bi X.Y., Wang H.X., Xie L.X., Wang Y.Q., Cao X.F., Lv J., Xiao F.J., et al. Microvesicles derived from human umbilical cord mesenchymal stem cells stimulated by hypoxia promote angiogenesis both in vitro and in vivo. Stem Cells Dev. 2012;21:3289–3297. doi: 10.1089/scd.2012.0095.
    1. 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. doi: 10.1371/journal.pone.0068451.
    1. Han Y.D., Bai Y., Yan X.L., Ren J., Zeng Q., Li X.D., Pei X.T., Han Y. Co-transplantation of exosomes derived from hypoxia-preconditioned adipose mesenchymal stem cells promotes neovascularization and graft survival in fat grafting. Biochem. Biophys. Res. Commun. 2018;497:305–312. doi: 10.1016/j.bbrc.2018.02.076.
    1. Han Y., Ren J., Bai Y., Pei X., Han Y. Exosomes from hypoxia-treated human adipose-derived mesenchymal stem cells enhance angiogenesis through VEGF/VEGF-R. Int. J. Biochem. Cell Biol. 2019;109:59–68. doi: 10.1016/j.biocel.2019.01.017.
    1. Almeria C., Weiss R., Roy M., Tripisciano C., Kasper C., Weber V., Egger D. Hypoxia Conditioned Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Increased Vascular Tube Formation in vitro. Front. Bioeng. Biotechnol. 2019 doi: 10.3389/fbioe.2019.00292.
    1. Xue C., Shen Y., Li X., Li B., Zhao S., Gu J., Chen Y., Ma B., Wei J., Han Q., et al. Exosomes Derived from Hypoxia-Treated Human Adipose Mesenchymal Stem Cells Enhance Angiogenesis Through the PKA Signaling Pathway. Stem Cells Dev. 2018;27:456–465. doi: 10.1089/scd.2017.0296.
    1. Zhu L.P., Tian T., Wang J.Y., He J.N., Chen T., Pan M., Xu L., Zhang H.X., Qiu X.T., Li C.C., et al. Hypoxia-elicited mesenchymal stem cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention of cell death in myocardial infarction. Theranostics. 2018;8:6163–6177. doi: 10.7150/thno.28021.
    1. Park H., Park H., Mun D., Kang J., Kim H., Kim M., Cui S., Lee S.H., Joung B. Extracellular Vesicles Derived from Hypoxic Human Mesenchymal Stem Cells Attenuate GSK3beta Expression via miRNA-26a in an Ischemia-Reperfusion Injury Model. Yonsei Med. J. 2018;59:736–745. doi: 10.3349/ymj.2018.59.6.736.
    1. Lee S.C., Jeong H.J., Lee S.K., Kim S.J. Hypoxic Conditioned Medium from Human Adipose-Derived Stem Cells Promotes Mouse Liver Regeneration Through JAK/STAT3 Signaling. Stem Cells Transl. Med. 2016;5:816–825. doi: 10.5966/sctm.2015-0191.
    1. Lin S., Zhu B., Huang G., Zeng Q., Wang C. Microvesicles derived from human bone marrow mesenchymal stem cells promote U2OS cell growth under hypoxia: The role of PI3K/AKT and HIF-1alpha. Hum. Cell. 2019;32:64–74. doi: 10.1007/s13577-018-0224-z.
    1. Li L., Jin S., Zhang Y. Ischemic preconditioning potentiates the protective effect of mesenchymal stem cells on endotoxin-induced acute lung injury in mice through secretion of exosome. Int. J. Clin. Exp. Med. 2015;8:3825–3832.
    1. Showalter M.R., Wancewicz B., Fiehn O., Archard J.A., Clayton S., Wagner J., Deng P., Halmai J., Fink K.D., Bauer G., et al. Primed mesenchymal stem cells package exosomes with metabolites associated with immunomodulation. Biochem. Biophys. Res. Commun. 2019;512:729–735. doi: 10.1016/j.bbrc.2019.03.119.
    1. Ren W., Hou J., Yang C., Wang H., Wu S., Wu Y., Zhao X., Lu C. Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J. Exp. Clin. Cancer Res. 2019;38:62. doi: 10.1186/s13046-019-1027-0.
    1. Lee S.C., Jeong H.J., Lee S.K., Kim S.J. Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome. Stem Cell Res. Ther. 2015;6:75. doi: 10.1186/s13287-015-0072-7.
    1. Ti D., Hao H., Tong C., Liu J., Dong L., Zheng J., Zhao Y., Liu H., Fu X., Han W. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J. Transl. Med. 2015;13 doi: 10.1186/s12967-015-0642-6.
    1. Kink J.A., Forsberg M.H., Reshetylo S., Besharat S., Childs C.J., Pederson J.D., Gendron-Fitzpatrick A., Graham M., Bates P.D., Schmuck E.G., et al. Macrophages Educated with Exosomes from Primed Mesenchymal Stem Cells Treat Acute Radiation Syndrome by Promoting Hematopoietic Recovery. Biol. Blood Marrow Transplant. 2019;25:2124–2133. doi: 10.1016/j.bbmt.2019.07.026.
    1. Xu R., Zhang F., Chai R., Zhou W., Hu M., Liu B., Chen X., Liu M., Xu Q., Liu N., et al. Exosomes derived from pro-inflammatory bone marrow-derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization. J. Cell. Mol. Med. 2019;23:7617–7631. doi: 10.1111/jcmm.14635.
    1. Kim Y.E., Sung S.I., Chang Y.S., Ahn S.Y., Sung D.K., Park W.S. Thrombin Preconditioning Enhances Therapeutic Efficacy of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells in Severe Neonatal Hypoxic Ischemic Encephalopathy. Int. J. Mol. Sci. 2019;20:2477. doi: 10.3390/ijms20102477.
    1. Alzahrani F.A. Melatonin improves therapeutic potential of mesenchymal stem cells-derived exosomes against renal ischemia-reperfusion injury in rats. Am. J. Transl. Res. 2019;11:2887–2907.
    1. Wang Y., Ma W.Q., Zhu Y., Han X.Q., Liu N. Exosomes Derived from Mesenchymal Stromal Cells Pretreated with Advanced Glycation End Product-Bovine Serum Albumin Inhibit Calcification of Vascular Smooth Muscle Cells. Front. Endocrinol. (Lausanne) 2018;9:524. doi: 10.3389/fendo.2018.00524.
    1. Ding J., Wang X., Chen B., Zhang J., Xu J. Exosomes Derived from Human Bone Marrow Mesenchymal Stem Cells Stimulated by Deferoxamine Accelerate Cutaneous Wound Healing by Promoting Angiogenesis. BioMed Res. Int. 2019;2019:9742765. doi: 10.1155/2019/9742765.
    1. Ruan X.F., Li Y.J., Ju C.W., Shen Y., Lei W., Chen C., Li Y., Yu H., Liu Y.T., Kim I.M., et al. Exosomes from Suxiao Jiuxin pill-treated cardiac mesenchymal stem cells decrease H3K27 demethylase UTX expression in mouse cardiomyocytes in vitro. Acta Pharmacol. Sin. 2018;39:579–586. doi: 10.1038/aps.2018.18.
    1. Du W., Zhang K., Zhang S., Wang R., Nie Y., Tao H., Han Z., Liang L., Wang D., Liu J., et al. Enhanced proangiogenic potential of mesenchymal stem cell-derived exosomes stimulated by a nitric oxide releasing polymer. Biomaterials. 2017;133:70–81. doi: 10.1016/j.biomaterials.2017.04.030.
    1. Jalnapurkar S., Moirangthem R.D., Singh S., Limaye L., Kale V. Microvesicles Secreted by Nitric Oxide-Primed Mesenchymal Stromal Cells Boost the Engraftment Potential of Hematopoietic Stem Cells. Stem Cells. 2019;37:128–138. doi: 10.1002/stem.2912.
    1. Bai Y., Han Y.D., Yan X.L., Ren J., Zeng Q., Li X.D., Pei X.T., Han Y. Adipose mesenchymal stem cell-derived exosomes stimulated by hydrogen peroxide enhanced skin flap recovery in ischemia-reperfusion injury. Biochem. Biophys. Res. Commun. 2018;500:310–317. doi: 10.1016/j.bbrc.2018.04.065.
    1. Li H., Zuo S., He Z., Yang Y., Pasha Z., Wang Y., Xu M. Paracrine factors released by GATA-4 overexpressed mesenchymal stem cells increase angiogenesis and cell survival. Am. J. Physiol. Heart Circ. Physiol. 2010;299:H1772–H1781. doi: 10.1152/ajpheart.00557.2010.
    1. He J.G., Li H.R., Han J.X., Li B.B., Yan D., Li H.Y., Wang P., Luo Y. GATA-4-expressing mouse bone marrow mesenchymal stem cells improve cardiac function after myocardial infarction via secreted exosomes. Sci. Rep. 2018;8:9047. doi: 10.1038/s41598-018-27435-9.
    1. Yu B., Kim H.W., Gong M., Wang J., Millard R.W., Wang Y., Ashraf M., Xu M. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int. J. Cardiol. 2015;182:349–360. doi: 10.1016/j.ijcard.2014.12.043.
    1. Kang K., Ma R., Cai W., Huang W., Paul C., Liang J., Wang Y., Zhao T., Kim H.W., Xu M., et al. Exosomes Secreted from CXCR4 Overexpressing Mesenchymal Stem Cells Promote Cardioprotection via Akt Signaling Pathway following Myocardial Infarction. Stem Cells Int. 2015;2015:659890. doi: 10.1155/2015/659890.
    1. Ma J., Zhao Y., Sun L., Sun X., Zhao X., Sun X., Qian H., Xu W., Zhu W. Exosomes Derived from Akt-Modified Human Umbilical Cord Mesenchymal Stem Cells Improve Cardiac Regeneration and Promote Angiogenesis via Activating Platelet-Derived Growth Factor D. Stem Cells Transl. Med. 2017;6:51–59. doi: 10.5966/sctm.2016-0038.
    1. Ling W., Zhang J., Yuan Z., Ren G., Zhang L., Chen X., Rabson A.B., Roberts A.I., Wang Y., Shi Y. Mesenchymal stem cells use IDO to regulate immunity in tumor microenvironment. Cancer Res. 2014;74:1576–1587. doi: 10.1158/0008-5472.CAN-13-1656.
    1. He J.G., Xie Q.L., Li B.B., Zhou L., Yan D. Exosomes Derived from IDO1-Overexpressing Rat Bone Marrow Mesenchymal Stem Cells Promote Immunotolerance of Cardiac Allografts. Cell Transplant. 2018 doi: 10.1177/0963689718805375.
    1. Gong X.H., Liu H., Wang S.J., Liang S.W., Wang G.G. Exosomes derived from SDF1-overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. J. Cell. Physiol. 2019;234:13878–13893. doi: 10.1002/jcp.28070.
    1. Liu J., Jiang M., Deng S., Lu J., Huang H., Zhang Y., Gong P., Shen X., Ruan H., Jin M., et al. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. Mol. Ther. Nucleic Acids. 2018;11:103–115. doi: 10.1016/j.omtn.2018.01.010.
    1. Chen Y., Zhao Y., Chen W., Xie L., Zhao Z.A., Yang J., Chen Y., Lei W., Shen Z. MicroRNA-133 overexpression promotes the therapeutic efficacy of mesenchymal stem cells on acute myocardial infarction. Stem Cell Res. Ther. 2017;8:268. doi: 10.1186/s13287-017-0722-z.
    1. Pan J., Alimujiang M., Chen Q., Shi H., Luo X. Exosomes derived from miR-146a-modified adipose-derived stem cells attenuate acute myocardial infarction-induced myocardial damage via downregulation of early growth response factor 1. J. Cell. Biochem. 2019;120:4433–4443. doi: 10.1002/jcb.27731.
    1. Luo Q., Guo D., Liu G., Chen G., Hang M., Jin M. Exosomes from MiR-126-Overexpressing Adscs Are Therapeutic in Relieving Acute Myocardial Ischaemic Injury. Cell. Physiol. Biochem. 2017;44:2105–2116. doi: 10.1159/000485949.
    1. Xin H., Li Y., Liu Z., Wang X., Shang X., Cui Y., Zhang Z.G., Chopp M. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells. 2013;31:2737–2746. doi: 10.1002/stem.1409.
    1. Xin H., Wang F., Li Y., Lu Q.E., Cheung W.L., Zhang Y., Zhang Z.G., Chopp M. Secondary Release of Exosomes from Astrocytes Contributes to the Increase in Neural Plasticity and Improvement of Functional Recovery After Stroke in Rats Treated with Exosomes Harvested from MicroRNA 133b-Overexpressing Multipotent Mesenchymal Stromal Cells. Cell Transplant. 2017;26:243–257.
    1. Shen H., Yao X., Li H., Li X., Zhang T., Sun Q., Ji C., Chen G. Role of Exosomes Derived from miR-133b Modified MSCs in an Experimental Rat Model of Intracerebral Hemorrhage. J. Mol. Neurosci. 2018;64:421–430. doi: 10.1007/s12031-018-1041-2.
    1. Zhao L., Jiang X., Shi J., Gao S., Zhu Y., Gu T., Shi E. Exosomes derived from bone marrow mesenchymal stem cells overexpressing microRNA-25 protect spinal cords against transient ischemia. J. Thorac. Cardiovasc. Surg. 2019;157:508–517. doi: 10.1016/j.jtcvs.2018.07.095.
    1. Yuan Z., Kolluri K.K., Gowers K.H., Janes S.M. TRAIL delivery by MSC-derived extracellular vesicles is an effective anticancer therapy. J. Extracell. Vesicles. 2017;6:1265291. doi: 10.1080/20013078.2017.1265291.
    1. 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;8:122. doi: 10.1186/s13045-015-0220-7.
    1. Li H., Yang C., Shi Y., Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. J. Nanobiotechnol. 2018;16:103. doi: 10.1186/s12951-018-0429-z.
    1. Yu L., Gui S., Liu Y., Qiu X., Zhang G., Zhang X., Pan J., Fan J., Qi S., Qiu B. Exosomes derived from microRNA-199a-overexpressing mesenchymal stem cells inhibit glioma progression by down-regulating AGAP2. Aging (Albany N. Y.) 2019;11:5300–5318. doi: 10.18632/aging.102092.
    1. Lang F.M., Hossain A., Gumin J., Momin E.N., Shimizu Y., Ledbetter D., Shahar T., Yamashita S., Parker Kerrigan B., Fueyo J., et al. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro-Oncol. 2018;20:380–390. doi: 10.1093/neuonc/nox152.
    1. 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. doi: 10.1016/j.omtn.2019.08.010.
    1. Xie C., Du L.Y., Guo F., Li X., Cheng B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol. Cell. Biochem. 2019;458:11–26. doi: 10.1007/s11010-019-03526-7.
    1. Gonzalez-King H., Garcia N.A., Ontoria-Oviedo I., Ciria M., Montero J.A., Sepulveda P. Hypoxia Inducible Factor-1alpha Potentiates Jagged 1-Mediated Angiogenesis by Mesenchymal Stem Cell-Derived Exosomes. Stem Cells. 2017;35:1747–1759. doi: 10.1002/stem.2618.
    1. Tao S.C., Yuan T., Zhang Y.L., Yin W.J., Guo S.C., Zhang C.Q. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 2017;7:180–195. doi: 10.7150/thno.17133.
    1. Mao G., Zhang Z., Hu S., Zhang Z., Chang Z., Huang Z., Liao W., Kang Y. Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem. Cell Res. Ther. 2018;9:247. doi: 10.1186/s13287-018-1004-0.
    1. Chen S., Tang Y., Liu Y., Zhang P., Lv L., Zhang X., Jia L., Zhou Y. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Prolif. 2019;52:e12669. doi: 10.1111/cpr.12669.
    1. Qu Y., Zhang Q., Cai X., Li F., Ma Z., Xu M., Lu L. Exosomes derived from miR-181-5p-modified adipose-derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. J. Cell. Mol. Med. 2017;21:2491–2502. doi: 10.1111/jcmm.13170.
    1. Chen L., Lu F.B., Chen D.Z., Wu J.L., Hu E.D., Xu L.M., Zheng M.H., Li H., Huang Y., Jin X.Y., et al. BMSCs-derived miR-223-containing exosomes contribute to liver protection in experimental autoimmune hepatitis. Mol. Immunol. 2018;93:38–46. doi: 10.1016/j.molimm.2017.11.008.
    1. Zhang L., Song Y., Chen L., Li D., Feng H., Lu Z., Fan T., Chen Z., Livingston M.J., Geng Q. MiR-20a-containing exosomes from umbilical cord mesenchymal stem cells alleviates liver ischemia/reperfusion injury. J. Cell. Physiol. 2020;235:3698–3710. doi: 10.1002/jcp.29264.
    1. Xu X.P., Huang L.L., Hu S.L., Han J.B., He H.L., Xu J.Y., Xie J.F., Liu A.R., Liu S.Q., Liu L., et al. Genetic Modification of Mesenchymal Stem Cells Overexpressing Angiotensin II Type 2 Receptor Increases Cell Migration to Injured Lung in LPS-Induced Acute Lung Injury Mice. Stem Cells Transl. Med. 2018;7:721–730. doi: 10.1002/sctm.17-0279.
    1. Wang D., Gao B., Yue J., Liu F., Liu Y., Fu W., Si Y. Exosomes from mesenchymal stem cells expressing miR-125b inhibit neointimal hyperplasia via myosin IE. J. Cell. Mol. Med. 2019;23:1528–1540. doi: 10.1111/jcmm.14060.
    1. Roy S., Hochberg F.H., Jones P.S. Extracellular vesicles: The growth as diagnostics and therapeutics; a survey. J. Extracell. Vesicles. 2018;7:1438720. doi: 10.1080/20013078.2018.1438720.
    1. Filho D.M., de Carvalho Ribeiro P., Oliveira L.F., Dos Santos A., Parreira R.C., Pinto M.C.X., Resende R.R. Enhancing the Therapeutic Potential of Mesenchymal Stem Cells with the CRISPR-Cas System. Stem Cell Rev. Rep. 2019;15:463–473. doi: 10.1007/s12015-019-09897-0.
    1. Borger V., Bremer M., Ferrer-Tur R., Gockeln L., Stambouli O., Becic A., Giebel B. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles and Their Potential as Novel Immunomodulatory Therapeutic Agents. Int. J. Mol. Sci. 2017;18:1450. doi: 10.3390/ijms18071450.
    1. Kim S., Kim T.M. Generation of mesenchymal stem-like cells for producing extracellular vesicles. World J. Stem Cells. 2019;11:270–280. doi: 10.4252/wjsc.v11.i5.270.

Source: PubMed

3
Abonnieren