Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapy for Alzheimer’s Disease: Progress and Opportunity
Abstract
:1. Introduction
2. Origins, Classification and Nomenclature of EVs
3. Recommendations in Characterization of EVs
4. Application of MSC-Derived EVs in AD Treatment
5. Therapeutic Mechanisms of MSC-Derived EVs Actions in AD
5.1. Aβ Degradation
5.2. Neuroprotection and Neuroregneration
5.3. Immunomodulation
6. Clinical Trials of MSC-Derived EVs in AD
7. Strategies for EV-Based Therapies
8. Conclusions and Prospects
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Tanaka, M.; Török, N.; Vécsei, L. Novel Pharmaceutical Approaches in Dementia. NeuroPsychopharmacotherapy 2021, 1–18. [Google Scholar] [CrossRef]
- Hodson, R. Alzheimer’s disease. Nature 2018, 559, S1. [Google Scholar] [CrossRef] [Green Version]
- Guo, T.; Zhang, D.; Zeng, Y.; Huang, T.Y.; Xu, H.; Zhao, Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol. Neurodegener. 2020, 15, 1–37. [Google Scholar] [CrossRef] [PubMed]
- Stancu, I.-C.; Vasconcelos, B.; Terwel, D.; Dewachter, I. Models of β-amyloid induced Tau-pathology: The long and “folded” road to understand the mechanism. Mol. Neurodegener. 2014, 9, 51. [Google Scholar] [CrossRef] [Green Version]
- Menting, K.W.; Claassen, J.A.H.R. β-secretase inhibitor; a promising novel therapeutic drug in Alzheimer’s disease. Front. Aging Neurosci. 2014, 6, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barve, K.H.; Kumar, M.S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease. Curr. Neuropharmacol. 2020, 18, 1106–1125. [Google Scholar] [CrossRef]
- Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Rubin, R. Recently Approved Alzheimer Drug Raises Questions That Might Never Be Answered. JAMA 2021, 326, 469–472. [Google Scholar] [CrossRef] [PubMed]
- Lalli, G.; Schott, J.M.; Hardy, J.; De Strooper, B. Aducanumab: A new phase in therapeutic development for Alzheimer’s disease? EMBO Mol. Med. 2021, 13, e14781. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.-K.; Chao, S.-P.; Hu, C.-J. Clinical trials of new drugs for Alzheimer disease. J. Biomed. Sci. 2020, 27, 18. [Google Scholar] [CrossRef]
- Herrup, K. The case for rejecting the amyloid cascade hypothesis. Nat. Neurosci. 2015, 18, 794–799. [Google Scholar] [CrossRef] [PubMed]
- Van Dyck, C.H. Anti-Amyloid-β Monoclonal Antibodies for Alzheimer’s Disease: Pitfalls and Promise. Biol. Psychiatry 2017, 83, 311–319. [Google Scholar] [CrossRef] [Green Version]
- Pittenger, M.F.; Discher, D.E.; Péault, B.M.; Phinney, D.G.; Hare, J.M.; Caplan, A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen. Med. 2019, 4, 22. [Google Scholar] [CrossRef] [Green Version]
- Shi, M.; Zhang, Z.; Xu, R.; Lin, H.; Fu, J.; Zou, Z.; Zhang, A.; Shi, J.; Chen, L.; Lv, S.; et al. Human Mesenchymal Stem Cell Transfusion Is Safe and Improves Liver Function in Acute-on-Chronic Liver Failure Patients. Stem Cells Transl. Med. 2012, 1, 725–731. [Google Scholar] [CrossRef]
- Lin, B.-L.; Chen, J.-F.; Qiu, W.-H.; Wang, K.-W.; Xie, D.-Y.; Chen, X.-Y.; Liu, Q.-L.; Peng, L.; Li, J.-G.; Mei, Y.-Y.; et al. Allogeneic bone marrow-derived mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic liver failure: A randomized controlled trial. Hepatology 2017, 66, 209–219. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.H.; Cho, J.H.; Lee, Y.H.; Lee, J.H.; Kim, S.S.; Kim, M.Y.; Lee, M.G.; Kang, W.Y.; Lee, K.S.; Ahn, Y.K.; et al. Improvement in Left Ventricular Function with Intracoronary Mesenchymal Stem Cell Therapy in a Patient with Anterior Wall ST-Segment Elevation Myocardial Infarction. Cardiovasc. Drugs Ther. 2018, 32, 329–338. [Google Scholar] [CrossRef] [Green Version]
- Karantalis, V.; DiFede, D.L.; Gerstenblith, G.; Pham, S.; Symes, J.; Zambrano, J.P.; Fishman, J.; Pattany, P.; McNiece, I.; Conte, J.; et al. Autologous Mesenchymal Stem Cells Produce Concordant Improvements in Regional Function, Tissue Perfusion, and Fibrotic Burden When Administered to Patients Undergoing Coronary Artery Bypass Grafting. Circ. Res. 2014, 114, 1302–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.S.; Hong, J.M.; Moon, G.J.; Lee, P.H.; Ahn, Y.H.; Bang, O.Y. A Long-Term Follow-Up Study of Intravenous Autologous Mesenchymal Stem Cell Transplantation in Patients With Ischemic Stroke. Stem Cells 2010, 28, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
- Vaquero, J.; Zurita, M.; Rico, M.A.; Aguayo, C.; Bonilla, C.; Marin, E.; Tapiador, N.; Sevilla, M.; Vazquez, D.; Carballido, J.; et al. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: Safety and efficacy of the 100/3 guideline. Cytotherapy 2018, 20, 806–819. [Google Scholar] [CrossRef] [PubMed]
- Chu, K.-A.; Wang, S.-Y.; Yeh, C.-C.; Fu, T.-W.; Fu, Y.-S.; Ko, T.-L.; Chiu, M.-M.; Chen, T.-H.; Tsai, P.-J. Reversal of bleomycin-induced rat pulmonary fibrosis by a xenograft of human umbilical mesenchymal stem cells from Wharton’s jelly. Theranostics 2019, 9, 6646–6664. [Google Scholar] [CrossRef] [PubMed]
- Sivandzade, F.; Cucullo, L. Regenerative Stem Cell Therapy for Neurodegenerative Diseases: An Overview. Int. J. Mol. Sci. 2021, 22, 2153. [Google Scholar] [CrossRef] [PubMed]
- Teixeira, F.G.; Carvalho, M.M.; Neves-Carvalho, A.; Panchalingam, K.M.; Behie, L.A.; Pinto, L.; Sousa, N.; Salgado, A.J. Secretome of Mesenchymal Progenitors from the Umbilical Cord Acts as Modulator of Neural/Glial Proliferation and Differentiation. Stem Cell Rev. Rep. 2014, 11, 288–297. [Google Scholar] [CrossRef] [PubMed]
- Zilka, N.; Zilkova, M.; Kazmerova, Z.; Sarissky, M.; Cigankova, V.; Novak, M. Mesenchymal stem cells rescue the Alzheimer’s disease cell model from cell death induced by misfolded truncated tau. Neuroscience 2011, 193, 330–337. [Google Scholar] [CrossRef]
- Oh, S.H.; Kim, H.N.; Park, H.-J.; Shin, J.Y.; Lee, P.H. Mesenchymal Stem Cells Increase Hippocampal Neurogenesis and Neuronal Differentiation by Enhancing the Wnt Signaling Pathway in an Alzheimer’s Disease Model. Cell Transplant. 2015, 24, 1097–1109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic Administration of Exosomes Released from Mesenchymal Stromal Cells Promote Functional Recovery and Neurovascular Plasticity After Stroke in Rats. Br. J. Pharmacol. 2013, 33, 1711–1715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Merimi, M.; El-Majzoub, R.; Lagneaux, L.; Agha, D.M.; Bouhtit, F.; Meuleman, N.; Fahmi, H.; Lewalle, P.; Fayyad-Kazan, M.; Najar, M. The Therapeutic Potential of Mesenchymal Stromal Cells for Regenerative Medicine: Current Knowledge and Future Understandings. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
- Guo, M.; Yin, Z.; Chen, F.; Lei, P. Mesenchymal stem cell-derived exosome: A promising alternative in the therapy of Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 109. [Google Scholar] [CrossRef]
- Teng, F.; Fussenegger, M. Shedding Light on Extracellular Vesicle Biogenesis and Bioengineering. Adv. Sci. 2020, 8, 2003505. [Google Scholar] [CrossRef] [PubMed]
- Dang, X.T.T.; Kavishka, J.M.; Zhang, D.X.; Pirisinu, M.; Le, M.T.N. Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020, 9, 2191. [Google Scholar] [CrossRef]
- Sidhom, K.; Obi, P.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef]
- Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. BioMed Res. Int. 2018, 2018, 8545347. [Google Scholar] [CrossRef]
- Liangsupree, T.; Multia, E.; Riekkola, M.-L. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. 2020, 1636, 461773. [Google Scholar] [CrossRef] [PubMed]
- Tan, S.J.O.; Floriano, J.F.; Nicastro, L.; Emanueli, C.; Catapano, F. Novel Applications of Mesenchymal Stem Cell-Derived Exosomes for Myocardial Infarction Therapeutics. Biomolecules 2020, 10, 707. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Futter, C.; Pearse, A.; Hewlett, L.J.; Hopkins, C.R. Multivesicular endosomes containing internalized EGF-EGF receptor complexes mature and then fuse directly with lysosomes. J. Cell Biol. 1996, 132, 1011–1023. [Google Scholar] [CrossRef]
- Palmulli, R.; Van Niel, G. To be or not to be. secreted as exosomes, a balance finely tuned by the mechanisms of biogenesis. Essays Biochem. 2018, 62, 177–191. [Google Scholar] [CrossRef] [PubMed]
- Clancy, J.W.; Schmidtmann, M.; D’Souza-Schorey, C. The ins and outs of microvesicles. FASEB BioAdvances 2021, 3, 399–406. [Google Scholar] [CrossRef]
- Taylor, J.; Bebawy, M. Proteins Regulating Microvesicle Biogenesis and Multidrug Resistance in Cancer. Proteomics 2018, 19, e1800165. [Google Scholar] [CrossRef] [Green Version]
- Song, Y.; Kim, Y.; Ha, S.; Sheller-Miller, S.; Yoo, J.; Choi, C.; Park, C.H. The emerging role of exosomes as novel therapeutics: Biology, technologies, clinical applications, and the next. Am. J. Reprod. Immunol. 2020, 85, e13329. [Google Scholar] [CrossRef]
- Nabhan, J.F.; Hu, R.; Oh, R.S.; Cohen, S.N.; Lu, Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc. Natl. Acad. Sci. USA 2012, 109, 4146–4151. [Google Scholar] [CrossRef] [Green Version]
- Wei, D.; Zhan, W.; Gao, Y.; Huang, L.; Gong, R.; Wang, W.; Zhang, R.; Wu, Y.; Gao, S.; Kang, T. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2020, 31, 157–177. [Google Scholar] [CrossRef] [PubMed]
- Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neuro-Oncology 2013, 113, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abou-Ghali, M.; Stiban, J. Regulation of ceramide channel formation and disassembly: Insights on the initiation of apoptosis. Saudi J. Biol. Sci. 2015, 22, 760–772. [Google Scholar] [CrossRef] [Green Version]
- Ma, Q.; Liang, M.; Wu, Y.; Luo, F.; Ma, Z.; Dong, S.; Xu, J.; Dou, C. Osteoclast-derived apoptotic bodies couple bone resorption and formation in bone remodeling. Bone Res. 2021, 9, 5. [Google Scholar] [CrossRef]
- Balakrishnan, A.; Roy, S.; Fleming, T.; Leong, H.S.; Schuurmans, C. The Emerging Role of Extracellular Vesicles in the Glioma Microenvironment: Biogenesis and Clinical Relevance. Cancers 2020, 12, 1964. [Google Scholar] [CrossRef]
- Bazzan, E.; Tinè, M.; Casara, A.; Biondini, D.; Semenzato, U.; Cocconcelli, E.; Balestro, E.; Damin, M.; Radu, C.; Turato, G.; et al. Critical Review of the Evolution of Extracellular Vesicles’ Knowledge: From 1946 to Today. Int. J. Mol. Sci. 2021, 22, 6417. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Xing, D.; Zhu, Y.; Dong, S.; Zhao, B. The State of Exosomes Research: A Global Visualized Analysis. BioMed Res. Int. 2019, 2019, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Palviainen, M.; Saraswat, M.; Varga, Z.; Kitka, D.; Neuvonen, M.; Puhka, M.; Joenväärä, S.; Renkonen, R.; Nieuwland, R.; Takatalo, M.; et al. Extracellular vesicles from human plasma and serum are carriers of extravesicular cargo—Implications for biomarker discovery. PLoS ONE 2020, 15, e0236439. [Google Scholar] [CrossRef]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
- Witwer, K.W.; Théry, C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J. Extracell. Vesicles 2019, 8, 1648167. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 1–18. [Google Scholar] [CrossRef]
- Lässer, C.; Eldh, M.; Lötvall, J. Isolation and Characterization of RNA-Containing Exosomes. J. Vis. Exp. 2012, 59, e3037. [Google Scholar] [CrossRef]
- Gutierrez-Vazquez, C.; Villarroya-Beltri, C.; Mittelbrunn, M.; Sánchez-Madrid, F. Transfer of extracellular vesicles during immune cell-cell interactions. Immunol. Rev. 2012, 251, 125–142. [Google Scholar] [CrossRef] [Green Version]
- Qiu, G.; Zheng, G.; Ge, M.; Wang, J.; Huang, R.; Shu, Q.; Xu, J. Functional proteins of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res. Ther. 2019, 10, 359. [Google Scholar] [CrossRef]
- Samanta, S.; Rajasingh, S.; Drosos, N.; Zhou, Z.; Dawn, B.; Rajasingh, J. Exosomes: New molecular targets of diseases. Acta Pharmacol. Sin. 2017, 39, 501–513. [Google Scholar] [CrossRef]
- Menck, K.; Sivaloganathan, S.; Bleckmann, A.; Binder, C. Microvesicles in Cancer: Small Size, Large Potential. Int. J. Mol. Sci. 2020, 21, 5373. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Li, G.; Liu, M.-L. Microvesicles as Emerging Biomarkers and Therapeutic Targets in Cardiometabolic Diseases. Genom. Proteom. Bioinform. 2018, 16, 50–62. [Google Scholar] [CrossRef] [PubMed]
- Kaid, C.; Assoni, A.; Marçola, M.; Semedo-Kuriki, P.; Bortolin, R.H.; Carvalho, V.M.; Okamoto, O.K. Proteome and miRNome profiling of microvesicles derived from medulloblastoma cell lines with stem-like properties reveals biomarkers of poor prognosis. Brain Res. 2020, 1730, 146646. [Google Scholar] [CrossRef] [PubMed]
- Serrano-Heras, G.; Díaz-Maroto, I.; Castro-Robles, B.; Carrión, B.; Perona-Moratalla, A.B.; Gracia, J.; Arteaga, S.; Hernández-Fernández, F.; García-García, J.; Ayo-Martín, O.; et al. Isolation and Quantification of Blood Apoptotic Bodies, a Non-invasive Tool to Evaluate Apoptosis in Patients with Ischemic Stroke and Neurodegenerative Diseases. Biol. Proced. Online 2020, 22, 17. [Google Scholar] [CrossRef]
- Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.-J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caruso, S.; Poon, I.K.H.; Caruso, S.; Poon, I.K.H. Apoptotic Cell-Derived Extracellular Vesicles: More Than Just Debris. Front. Immunol. 2018, 9, 1486. [Google Scholar] [CrossRef] [Green Version]
- Pant, S.; Hilton, H.; Burczynski, M.E. The multifaceted exosome: Biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem. Pharmacol. 2012, 83, 1484–1494. [Google Scholar] [CrossRef] [PubMed]
- Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
- Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bang, O.Y.; Kim, E.H. Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Stroke: Challenges and Progress. Front. Neurol. 2019, 10, 211. [Google Scholar] [CrossRef] [Green Version]
- Abreu, S.C.; Lopes-Pacheco, M.; Weiss, D.J.; Rocco, P.R.M. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Lung Diseases: Current Status and Perspectives. Front. Cell Dev. Biol. 2021, 9, 97. [Google Scholar] [CrossRef]
- Eirin, A.; Lerman, L.O. Mesenchymal Stem/Stromal Cell–Derived Extracellular Vesicles for Chronic Kidney Disease: Are We There Yet? Hypertension 2021, 78, 261–269. [Google Scholar] [CrossRef]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Ma, X.; Huang, M.; Zheng, M.; Dai, C.; Song, Q.; Zhang, Q.; Li, Q.; Gu, X.; Chen, H.; Jiang, G.; et al. ADSCs-derived extracellular vesicles alleviate neuronal damage, promote neurogenesis and rescue memory loss in mice with Alzheimer’s disease. J. Control. Release 2020, 327, 688–702. [Google Scholar] [CrossRef]
- Chen, Y.-A.; Lu, C.-H.; Ke, C.-C.; Chiu, S.-J.; Jeng, F.-S.; Chang, C.-W.; Yang, B.-H.; Liu, R.-S. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Alzheimer’s Disease Pathology and Improve Cognitive Deficits. Biomedicines 2021, 9, 594. [Google Scholar] [CrossRef]
- Gratpain, V.; Mwema, A.; Labrak, Y.; Muccioli, G.G.; van Pesch, V.; Rieux, A.D. Extracellular vesicles for the treatment of central nervous system diseases. Adv. Drug Deliv. Rev. 2021, 174, 535–552. [Google Scholar] [CrossRef]
- Matsumoto, J.; Stewart, T.; Sheng, L.; Li, N.; Bullock, K.; Song, N.; Shi, M.; Banks, W.A.; Zhang, J. Transmission of α-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: Another mechanism for initiation and progression of Parkinson’s disease? Acta Neuropathol. Commun. 2017, 5, 71. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.C.; Liu, L.; Ma, F.; Wong, C.W.; Guo, X.E.; Chacko, J.V.; Farhoodi, H.; Zhang, S.; Zimak, J.; Ségaliny, A.; et al. Elucidation of Exosome Migration Across the Blood–Brain Barrier Model In Vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef]
- 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]
- Reza-Zaldivar, E.E.; Hernández-Sapiéns, M.A.; Gutiérrez-Mercado, Y.K.; Sandoval-Ávila, S.; Gomez-Pinedo, U.; Márquez-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. [Google Scholar] [CrossRef] [PubMed]
- De Godoy, M.A.; Saraiva, L.M.; de Carvalho, L.R.; Vasconcelos-Dos-Santos, A.; Beiral, H.J.; Ramos, A.B.; Silva, L.R.D.P.; Leal, R.B.; Monteiro, V.H.S.; Braga, C.V.; et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J. Biol. Chem. 2018, 293, 1957–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bodart-Santos, V.; De Carvalho, L.R.P.; De Godoy, M.A.; Batista, A.F.; Saraiva, L.M.; Lima, L.G.; Abreu, C.A.; De Felice, F.G.; Galina, A.; Mendez-Otero, R.; et al. Extracellular vesicles derived from human Wharton’s jelly mesenchymal stem cells protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. Stem Cell Res. Ther. 2019, 10, 332. [Google Scholar] [CrossRef]
- Wang, S.-S.; Jia, J.; Wang, Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles Suppresses iNOS Expression and Ameliorates Neural Impairment in Alzheimer’s Disease Mice. J. Alzheimer’s Dis. 2018, 61, 1005–1013. [Google Scholar] [CrossRef]
- Losurdo, M.; Pedrazzoli, M.; D’Agostino, C.; Elia, C.A.; Massenzio, F.; Lonati, E.; Mauri, M.; Rizzi, L.; Molteni, L.; Bresciani, E.; et al. Intranasal delivery of mesenchymal stem cell-derived extracellular vesicles exerts immunomodulatory and neuroprotective effects in a 3xTg model of Alzheimer’s disease. Stem Cells Transl. Med. 2020, 9, 1068–1084. [Google Scholar] [CrossRef] [PubMed]
- Cone, A.S.; Yuan, X.; Sun, L.; Duke, L.C.; Vreones, M.P.; Carrier, A.N.; Kenyon, S.M.; Carver, S.R.; Benthem, S.D.; Stimmell, A.C.; et al. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model. Theranostics 2021, 11, 8129–8142. [Google Scholar] [CrossRef]
- Katsuda, T.; Tsuchiya, R.; Kosaka, N.; Yoshioka, Y.; Takagaki, K.; Oki, K.; Takeshita, F.; Sakai, Y.; Kuroda, M.; Ochiya, T. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci. Rep. 2013, 3, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Yang, L.; Zhai, Y.; Hao, Y.; Zhu, Z.; Cheng, G. The Regulatory Functionality of Exosomes Derived from hUMSCs in 3D Culture for Alzheimer’s Disease Therapy. Small 2019, 16, e1906273. [Google Scholar] [CrossRef]
- Lee, M.; Ban, J.-J.; Yang, S.; Im, W.; Kim, M. The exosome of adipose-derived stem cells reduces β-amyloid pathology and apoptosis of neuronal cells derived from the transgenic mouse model of Alzheimer’s disease. Brain Res. 2018, 1691, 87–93. [Google Scholar] [CrossRef]
- Ding, M.; Shen, Y.; Wang, P.; Xie, Z.; Xu, S.; Zhu, Z.; Wang, Y.; Lyu, Y.; Wang, D.; Xu, L.; et al. Exosomes Isolated From Human Umbilical Cord Mesenchymal Stem Cells Alleviate Neuroinflammation and Reduce Amyloid-Beta Deposition by Modulating Microglial Activation in Alzheimer’s Disease. Neurochem. Res. 2018, 43, 2165–2177. [Google Scholar] [CrossRef] [PubMed]
- Cui, G.; Wu, J.; Mou, F.; Xie, W.; Wang, F.; Wang, Q.; Fang, J.; Xu, Y.; Dong, Y.; Liu, J.; 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. [Google Scholar] [CrossRef] [Green Version]
- Cui, G.-H.; Guo, H.-D.; Li, H.; Zhai, Y.; Gong, Z.-B.; Wu, J.; Liu, J.-S.; Dong, Y.-R.; Hou, S.-X. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun. Ageing 2019, 16, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Elia, C.A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M.; et al. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Aβ Plaque Burden in Early Stages of a Preclinical Model of Alzheimer’s Disease. Cells 2019, 8, 1059. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Yang, G. Bone marrow mesenchymal stem cells-derived exosomes reduce Aβ deposition and improve cognitive function recovery in mice with Alzheimer’s disease by activating sphingosine kinase/sphingosine-1-phosphate signaling pathway. Cell Biol. Int. 2020, 45, 775–784. [Google Scholar] [CrossRef] [PubMed]
- Zhai, L.; Shen, H.; Sheng, Y.; Guan, Q. ADMSC Exo-MicroRNA-22 improve neurological function and neuroinflammation in mice with Alzheimer’s disease. J. Cell. Mol. Med. 2021, 25, 7513–7523. [Google Scholar] [CrossRef] [PubMed]
- Apodaca, L.A.; Baddour, A.A.D.; Garcia, C.; Alikhani, L.; Giedzinski, E.; Ru, N.; Agrawal, A.; Acharya, M.M.; Baulch, J.E. Human neural stem cell-derived extracellular vesicles mitigate hallmarks of Alzheimer’s disease. Alzheimer’s Res. Ther. 2021, 13, 57. [Google Scholar] [CrossRef]
- Sha, S.; Shen, X.; Cao, Y.; Qu, L. Mesenchymal stem cells-derived extracellular vesicles ameliorate Alzheimer’s disease in rat models via the microRNA-29c-3p/BACE1 axis and the Wnt/β-catenin pathway. Aging 2021, 13, 15285–15306. [Google Scholar] [CrossRef]
- Van Es, M.A.; Berg, L.H.V.D. Alzheimer’s disease beyond APOE. Nat. Genet. 2009, 41, 1047–1048. [Google Scholar] [CrossRef]
- Huang, L.; Su, X.; Federoff, H.J. Single-Chain Fragment Variable Passive Immunotherapies for Neurodegenerative Diseases. Int. J. Mol. Sci. 2013, 14, 19109–19127. [Google Scholar] [CrossRef] [PubMed]
- Dahlgren, K.N.; Manelli, A.M.; Stine, W.B.; Baker, L.; Krafft, G.A.; LaDu, M.J. Oligomeric and Fibrillar Species of Amyloid-β Peptides Differentially Affect Neuronal Viability. J. Biol. Chem. 2002, 277, 32046–32053. [Google Scholar] [CrossRef] [Green Version]
- Haass, C.; Selkoe, D.J. Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 2007, 8, 101–112. [Google Scholar] [CrossRef] [PubMed]
- Shankar, G.M.; Li, S.; Mehta, T.H.; Garcia-Munoz, A.; Shepardson, N.E.; Smith, I.; Brett, F.M.; Farrell, M.A.; Rowan, M.J.; Lemere, C.A.; et al. Amyloid-β protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 2008, 14, 837–842. [Google Scholar] [CrossRef] [Green Version]
- Tarasoff-Conway, J.M.; Carare, R.O.; Osorio, R.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B.V.; et al. Clearance systems in the brain—implications for Alzheimer disease. Nat. Rev. Neurol. 2015, 11, 457–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erickson, M.; Banks, W.A. Blood–Brain Barrier Dysfunction as a Cause and Consequence of Alzheimer’s Disease. Br. J. Pharmacol. 2013, 33, 1500–1513. [Google Scholar] [CrossRef] [Green Version]
- Iwata, N.; Tsubuki, S.; Takaki, Y.; Shirotani, K.; Lu, B.; Gerard, N.P.; Gerard, C.; Hama, E.; Lee, H.-J.; Saido, T.C. Metabolic Regulation of Brain Aβ by Neprilysin. Science 2001, 292, 1550–1552. [Google Scholar] [CrossRef]
- Iwata, N.; Tsubuki, S.; Takaki, Y.; Watanabe, K.; Sekiguchi, M.; Hosoki, E.; Kawashima-Morishima, M.; Lee, H.-J.; Hama, E.; Sekine-Aizawa, Y.; et al. Identification of the major Aβ1–42-degrading catabolic pathway in brain parenchyma: Suppression leads to biochemical and pathological deposition. Nat. Med. 2000, 6, 143–150. [Google Scholar] [CrossRef]
- Yasojima, K.; Akiyama, H.; McGeer, E.G.; McGeer, P.L. Reduced neprilysin in high plaque areas of Alzheimer brain: A possible relationship to deficient degradation of β-amyloid peptide. Neurosci. Lett. 2001, 297, 97–100. [Google Scholar] [CrossRef]
- Mufson, E.; Mahady, L.; Waters, D.; Counts, S.; Perez, S.; DeKosky, S.; Ginsberg, S.; Ikonomovic, M.; Scheff, S.; Binder, L. Hippocampal plasticity during the progression of Alzheimer’s disease. Neuroscience 2015, 309, 51–67. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
- Chasaide, C.N.; Lynch, M.A. The role of the immune system in driving neuroinflammation. Brain Neurosci. Adv. 2020, 4, 2398212819901082. [Google Scholar] [CrossRef] [Green Version]
- Xie, M.; Xiong, W.; She, Z.; Wen, Z.; Abdirahman, A.S.; Wan, W.; Wen, C. Immunoregulatory Effects of Stem Cell-Derived Extracellular Vesicles on Immune Cells. Front. Immunol. 2020, 11, 13. [Google Scholar] [CrossRef] [Green Version]
- Harting, M.T.; Srivastava, A.; Zhaorigetu, S.; Bair, H.; Prabhakara, K.S.; Furman, N.E.T.; Vykoukal, J.V.; Ruppert, K.A.; Cox, C.S.; Olson, S.D. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem Cells 2017, 36, 79–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Togo, T.; Katsuse, O.; Iseki, E. Nitric oxide pathways in Alzheimer’s disease and other neurodegenerative dementias. Neurol. Res. 2004, 26, 563–566. [Google Scholar] [CrossRef] [PubMed]
- Cosenza, S.; Ruiz, M.; Toupet, K.; Jorgensen, C.; Noël, D. Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef]
- De Strooper, B.; Karran, E. The Cellular Phase of Alzheimer’s Disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef] [Green Version]
- Vizoso, F.; Fernández-Francos, S.; Eiro, N. Mesenchymal Stem/Stromal Cells and Their Derivates in Acute Diseases: Emergency in the Post-COVID-19 Times. Int. J. Mol. Sci. 2021, 22, 8395. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Zhang, X.; Chen, X.; Wang, L.; Yang, G. Exosome Mediated Delivery of miR-124 Promotes Neurogenesis after Ischemia. Mol. Ther. Nucleic Acids 2017, 7, 278–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendt, M.; Rezvani, K.; Shpall, E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant. 2019, 54, 789–792. [Google Scholar] [CrossRef] [PubMed]
- Fu, S.; Wang, Y.; Xia, X.; Zheng, J.C. Exosome engineering: Current progress in cargo loading and targeted delivery. NanoImpact 2020, 20, 100261. [Google Scholar] [CrossRef]
- Seo, Y.; Kim, H.-S.; Hong, I.-S. Stem Cell-Derived Extracellular Vesicles as Immunomodulatory Therapeutics. Stem Cells Int. 2019, 2019, 5126156. [Google Scholar] [CrossRef]
- Grange, C.; Tapparo, M.; Bruno, S.; Chatterjee, D.; Quesenberry, P.J.; Tetta, C.; Camussi, G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int. J. Mol. Med. 2014, 33, 1055–1063. [Google Scholar] [CrossRef] [Green Version]
- Shao, J.; Zaro, J.; Shen, Y. Advances in Exosome-Based Drug Delivery and Tumor Targeting: From Tissue Distribution to Intracellular Fate. Int. J. Nanomed. 2020, 15, 9355–9371. [Google Scholar] [CrossRef]
- Lu, C.-H.; Chen, Y.-A.; Ke, C.-C.; Chiu, S.-J.; Chen, C.-C.; Hsieh, Y.-J.; Yang, B.-H.; Liu, R.-S. Preclinical Characterization and In Vivo Imaging of 111In-Labeled Mesenchymal Stem Cell–Derived Extracellular Vesicles. Mol. Imaging Biol. 2020, 23, 361–371. [Google Scholar] [CrossRef]
- Takahashi, Y.; Nishikawa, M.; Shinotsuka, H.; Matsui, Y.; Ohara, S.; Imai, T.; Takakura, Y. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J. Biotechnol. 2013, 165, 77–84. [Google Scholar] [CrossRef]
- Armstrong, J.P.K.; Holme, M.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
- Piffoux, M.; Nicolás-Boluda, A.; Mulens-Arias, V.; Richard, S.; Rahmi, G.; Gazeau, F.; Wilhelm, C.; Silva, A.K. Extracellular vesicles for personalized medicine: The input of physically triggered production, loading and theranostic properties. Adv. Drug Deliv. Rev. 2018, 138, 247–258. [Google Scholar] [CrossRef]
- Jurgielewicz, B.; Stice, S.; Yao, Y. Therapeutic Potential of Nucleic Acids when Combined with Extracellular Vesicles. Aging Dis. 2021, 12, 1476–1493. [Google Scholar] [CrossRef]
- Izco, M.; Blesa, J.; Schleef, M.; Schmeer, M.; Porcari, R.; Al-Shawi, R.; Ellmerich, S.; De Toro, M.; Gardiner, C.; Seow, Y.; et al. Systemic Exosomal Delivery of shRNA Minicircles Prevents Parkinsonian Pathology. Mol. Ther. 2019, 27, 2111–2122. [Google Scholar] [CrossRef]
- Ren, X.; Zhao, Y.; Xue, F.; Zheng, Y.; Huang, H.; Wang, W.; Chang, Y.; Yang, H.; Zhang, J. Exosomal DNA Aptamer Targeting α-Synuclein Aggregates Reduced Neuropathological Deficits in a Mouse Parkinson’s Disease Model. Mol. Ther. Nucleic Acids 2019, 17, 726–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, T.; Zhang, H.-X.; He, C.-P.; Fan, S.; Zhu, Y.-L.; Qi, C.; Huang, N.-P.; Xiao, Z.-D.; Lu, Z.-H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2017, 150, 137–149. [Google Scholar] [CrossRef]
- Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018, 178, 302–316. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Xiong, G.; Tsang, W.C.; Schätzlein, A.G.; Uchegbu, I.F. Nose-to-Brain Delivery. J. Pharmacol. Exp. Ther. 2019, 370, 593–601. [Google Scholar] [CrossRef] [Green Version]
- Bai, L.; Liu, Y.; Guo, K.; Zhang, K.; Liu, Q.; Wang, P.; Wang, X. Ultrasound Facilitates Naturally Equipped Exosomes Derived from Macrophages and Blood Serum for Orthotopic Glioma Treatment. ACS Appl. Mater. Interfaces 2019, 11, 14576–14587. [Google Scholar] [CrossRef]
- Yuan, D.; Zhao, Y.; Banks, W.A.; Bullock, K.M.; Haney, M.; Batrakova, E.; Kabanov, A.V. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials 2017, 142, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Kim, G.; Kim, M.; Lee, Y.; Byun, J.W.; Hwang, D.W.; Lee, M. Systemic delivery of microRNA-21 antisense oligonucleotides to the brain using T7-peptide decorated exosomes. J. Control. Release 2020, 317, 273–281. [Google Scholar] [CrossRef]
- Ye, Z.; Zhang, T.; He, W.; Jin, H.; Liu, C.; Yang, Z.; Ren, J. Methotrexate-Loaded Extracellular Vesicles Functionalized with Therapeutic and Targeted Peptides for the Treatment of Glioblastoma Multiforme. ACS Appl. Mater. Interfaces 2018, 10, 12341–12350. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of Brain Inflammatory Diseases by Delivering Exosome Encapsulated Anti-inflammatory Drugs From the Nasal Region to the Brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
Model | Source of EVs | Protocol | Administration Route | Reported Effects | Ref. |
---|---|---|---|---|---|
In vitro models | |||||
N2a cells | ADSCs | 500 µg/well, 24 h | Co-culture | Decreased extracellular and intracellular Aβs levels | [82] |
SH-SY5Y-APPswe cells | UC-MSC | 2 μg/well for 24 h | Co-culture | Decreased extracellular and intracellular Aβs levels | [83] |
SH-SY5Y-APP(S/L) cells | WJ-MSCs | 50 µg/well, twice a week for 1 week | Co-culture | Decreased Aβs expression and restored the expression of neuronal memory/synaptic plasticity-related genes | [71] |
NSCs isolated from Tg2576 mice | ADSCs | 200 μg/mL for 24 or 48 h | Co-culture | Reduced Aβ levels and the Aβ 42/40 ratio, increased neurite growth and alleviated cell apoptosis | [84] |
Cortical neuron culture from newborn APP/PS1 mice | BM-MSCs | 100 μg/mL for 12 h | Co-culture | Reduced Aβs induced iNOS expression | [79] |
Hippocampal neuron culture from rat embryos (E18) | BM-MSCs isolated from Wistar rats | 2.4 × 108 particles for 22 h | Co-culture (Pretreatment with 500 nM of AβOs for 2 h) | Protected neurons from AβOs-induced oxidative stress and synapse damage | [77] |
Hippocampal neuron culture from rat embryos (E18) | WJ-MSCs | 6 × 108 particles for 22 h | Co-culture (With 500 nM of AβOs for 2 h) | Protected neurons from AβOs-induced oxidative stress and synapse damage | [78] |
Cortical neurons culture from C57BL/6 mice embryos (E13–15) | ADSCs | 0.05, 0.1, 1 μg/mL for 24 h | Co-culture (Pretreatment with 20 μM of AβOs) | Alleviated AβOs-induced neuronal toxicity | [70] |
In vivo models | |||||
APP/PS1 mice | |||||
No age indicate | BM-MSCs | 100 μg/5 μL, once per 2 days for 2 weeks | i.c.v. | Improved cognitive behavior, rescued impairment of CA1 synaptic transmission and LTP | [79] |
7-month-old | UC-MSC | 30 μg/100 μL, every 2 weeks, four times | i.v. | Reduced Aβ deposition, improved cognitive behavior; enhanced expression of IDE and NEP; modulated the activation of microglia | [85] |
7-month-old | PC-BM-MSCs | 150 μg/80 μL, biweekly for 4 months | i.v. | Improved cognitive behavior, reduced Aβ deposition; decreased proinflammatory factors and increased anti-inflammatory factors | [86] |
7-month-old | RVG-BM-MSCs | 5 × 1011 particles/100 μL, monthly for 4 months | i.v. | Improved cognitive behavior, reduced Aβ deposition, and restored the levels of inflammatory cytokines | [87] |
5-month-old | BM-MSCs | 22.4 μg/4 μL | i.c.v | Reduced Aβ deposition and the amount of dystrophic neurons in both the cortex and hippocampus | [88] |
9-month-old | UC-MSC | 2 mg/mL, continuously at 0.25 µL/h for 14 days | i.c.v. | Reduced Aβ deposition, improved cognitive behavior and inhibited the inflammatory and oxidative stress | [83] |
7-month-old | BM-MSCs | 50 μg/80 μL, every 2 weeks for 16 weeks | i.v. | Reduced Aβ deposition, promoted cognitive function recovery and increased NeuN expression | [89] |
4-month-old | miRNA-22-loaded mouse ADSCs | 100 μg/mL, every 7 days until 30 days | i.v. | Improved cognitive behavior, inhibited the inflammatory factors expression and reduced the nerve cell damage | [90] |
9-month-old | ADSCs | 1 mg/kg in 10 μL, every two days for 2 weeks | IN | Ameliorated neurologic damage in the whole brain areas, increased neurogenesis, reduced Aβ deposition and decreased microglia activation | [70] |
J20 mice | |||||
9-month-old | WJ-MSCs | 50 µg/100 µL, once a week for 4 weeks | i.v. | Restored the expression of neuronal memory/synaptic plasticity-related genes, improved brain glucose metabolism and cognitive function; inhibited astrocyte and microglia activation | [71] |
3 × Tg | |||||
7-month-old | Cytokine-preconditioned BM-MSCs | 30 μg/100 µL | IN | Decreased microglia activation and increased dendritic spine density | [80] |
5 × FAD | |||||
2-month-old | BM-MSCs | 20 × 108 particles in 5 µL every 4 days until 4 months of age | IN | Improved cognitive behavior, reduced Aβ deposition in the hippocampus and decreased colocalization between GFAP and Aβ plaques | [81] |
1.5–2.5-month-old 5.0–6.5-month-old | hNSC | 2.25 × 107 particles in 50 μL hibernation buffer | i.v. via RO injection | Restored fear extinction memory consolidation and reduced anxiety related behaviors; reduced the dense core Aβ plaque number and microglial activation; restored synaptophysin in the AD brain and homeostatic levels of pro-inflammatory cytokines | [91] |
Administration of Aβ peptides into the dentate gyrus of C57BL/6 mice 8-week-old | MSCs (No source indicated) | 10 µg/2 µL of PBS | i.c.v. into the dentate gyrus | Promoted neurogenesis in the SVZ and alleviated Aβ1–42-induced cognitive impairment | [76] |
Administration of Aβ peptides into the lateral ventricle of SD rats (7-week-old) | BM-MSCs isolated from SD rats | 30 μg/100 µL, once a month for 2 months | i.c.v. into the lateral ventricle | Reduced Aβ deposition, reduced the levels of inflammatory cytokines, elevated NEP and IDE expressions, increased neuron viability and reduced apoptosis rate | [92] |
Disease/Condition | Clinical Trial Number | Title | Sponsor |
---|---|---|---|
“Exosome” used in title | |||
Alzheimer’s disease | NCT04388982 | The Safety and the Efficacy Evaluation of Allogenic Adipose MSC-Exos in Patients with Alzheimer’s Disease (Adipose MSC-derived exosomes) | Ruijin Hospital |
Cerebrovascular disorders | NCT03384433 | Allogenic Mesenchymal Stem Cell Derived Exosome in Patients with Acute Ischemic Stroke | Isfahan University of Medical Sciences |
Acute respiratory distress syndrome (ARDS) | NCT04602104 | A Clinical Study of Mesenchymal Stem Cell Exosomes Nebulizer for the Treatment of ARDS | Ruijin Hospital |
Coronavirus | NCT04276987 | A Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus Pneumonia | Ruijin Hospital |
Healthy | NCT04313647 | A Tolerance Clinical Study on Aerosol Inhalation of Mesenchymal Stem Cells Exosomes in Healthy Volunteers | Ruijin Hospital |
Macular holes | NCT03437759 | MSC-Exos Promote Healing of MHs | Tianjin Medical University |
Multiple organ failure | NCT04356300 | Exosome of Mesenchymal Stem Cells for Multiple Organ Dysfuntion Syndrome After Surgical Repaire of Acute Type A Aortic Dissection | Fujian Medical University |
Dry eye | NCT04213248 | Effect of UMSCs Derived Exosomes on Dry Eye in Patients With cGVHD (Umbilical MSCs derived exosomes) | Zhongshan Ophthalmic Center, Sun Yat-sen University |
Drug-resistant | NCT04544215 | A Clinical Study of Mesenchymal Progenitor Cell Exosomes Nebulizer for the Treatment of Pulmonary Infection | Ruijin Hospital |
Sepsis Critical illness | NCT04850469 | Study of MSC-Exo on the Therapy for Intensively Ill Children | Children’s Hospital of Fudan University |
Periodontitis | NCT04270006 | Effect of Adipose Derived Stem Cells Exosomes as an Adjunctive Therapy to Scaling and Root Planning in the Treatment of Periodontitis: A Human Clinical Trial (Adipose derived stem cells exosomes) | Beni-Suef University |
COVID-19 SARS-CoV-2 Pneumonia | NCT04491240 | Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia | State-Financed Health Facility “Samara Regional Medical Center Dinasty” |
“Extracellular vesicle” used in title | |||
Bronchopulmonary Dysplasia | NCT03857841 | A Safety Study of IV Stem Cell derived Extracellular Vesicles (UNEX-42) in Preterm Neonates at High Risk for BPD | United Therapeutics |
Dystrophic epidermolysis Bullosa | NCT04173650 | MSC Evs in Dystrophic Epidermolysis Bullosa | Aegle Therapeutics |
COVID-19 ARDS Hypoxia Cytokine storm | NCT04657458 | Expanded Access Protocol on Bone Marrow Mesenchymal Stem Cell Derived Extracellular Vesicle Infusion Treatment for Patients With COVID-19 Associated ARDS (BM-MSC derived EVs) | Direct Biologics, LLC |
COVID-19 ARDS Pneumonia, Viral | NCT04493242 | Extracellular Vesicle Infusion Treatment for COVID-19 Associated ARDS (BM-MSC derived EVs) | Direct Biologics, LLC |
Strategies | Cargo-Loaded Molecules | Source of EVs | Disease | Reported Effects | Ref. |
---|---|---|---|---|---|
Peptide-tagged | |||||
Rabies viral glycoprotein (RVG) | Nucleic acid siRNAs specific to BACE1 | Dendritic cells | AD | Significant knockdown of BACE1 in mRNA and protein levels | [69] |
Naturally production | BM-MSCs | AD | Improved cognitive behavior, reduced Aβ deposition, and restored the levels of inflammatory cytokines | [87] | |
miR-124 | Mouse BM-MSCs | Ischemic stroke | Promoted cortical neurogenesis | [113] | |
siRNAs specific to α-synuclein | Dendritic cells | PD | Decreased α-synuclein aggregation and rescued the loss of dopaminergic neurons | [124] | |
DNA aptamers that recognize the α-synuclein | HEK293T | PD | Reduced α-synuclein aggregation and improved motor impairments | [125] | |
RGD peptides | Drug loaded Curcumin | Mouse BM-MSCs | Ischemic stroke | Strong suppression of the inflammatory response and cellular apoptosis | [126] |
T7 peptide | Antisense miRNA oligonucleotides against miR-21 (AMO-21) | HEK293T | Glioblastoma | Reduction of tumor sizes | [131] |
NRP-1-targeted RGE peptide | Superparamagnetic iron oxide nanoparticles (SPIONs) and curcumin | Raw264.7 cells, a macrophage cell line | Glioma | Delayed tumor recurrence, extended the survival of tumor-bearing mice and had targeted-imaging ability | [127] |
Low-density lipoprotein (LDL) | Drug loaded Methotrexate | L929, a mouse fibroblastic cell line | Glioma | Prolonged the median survival period | [132] |
Natural production | |||||
LFA-1 expression | BDNF | Macrophage | PD | Enhanced delivery and accumulation in inflamed brain | [130] |
Unidentified | Paclitaxel and doxorubicin | Brain endothelial cells | Brain cancer | Induction of cytotoxic effects against brain cancer | [133] |
Administration route | |||||
IN | Unmodified | ADSCs | AD | Decreased AβOs-induced neuronal toxicity | [70,81] |
Cytokine-stimulated | BM-MSCs | AD | Increased dendritic spine density, reduced Aβ deposition and microglia activation | [80] | |
Drug loaded Curcumin, JSI-124, a Stat3 inhibitor | EL-4, a T cell line | Inflammation-mediated disease models, including LPS-induced brain inflammation model, EAE model and a GL26 brain tumor model | Selectively taken up by microglia and induced apoptosis | [134] | |
Disruption of BBB by pFUS | Unmodified | Blood serum | Glioma | Suppressed glioma growth with no obvious side effects | [129] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chen, Y.-A.; Lu, C.-H.; Ke, C.-C.; Liu, R.-S. Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapy for Alzheimer’s Disease: Progress and Opportunity. Membranes 2021, 11, 796. https://doi.org/10.3390/membranes11100796
Chen Y-A, Lu C-H, Ke C-C, Liu R-S. Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapy for Alzheimer’s Disease: Progress and Opportunity. Membranes. 2021; 11(10):796. https://doi.org/10.3390/membranes11100796
Chicago/Turabian StyleChen, Yi-An, Cheng-Hsiu Lu, Chien-Chih Ke, and Ren-Shyan Liu. 2021. "Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapy for Alzheimer’s Disease: Progress and Opportunity" Membranes 11, no. 10: 796. https://doi.org/10.3390/membranes11100796