Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities
Abstract
:1. Introduction
2. EVs Types and Biogenesis
3. EVs Characteristics
3.1. Exosome
3.2. Microvesicles
3.3. Apoptotic Bodies
4. MSC-EVs Components
4.1. Pro-Angiogenic Factors
4.2. Immunomodulatory Factors
miRNA | Role/Function in MSC-EVs | Potential Applications | Reference |
---|---|---|---|
miR-22 | inhibits the inflammatory response and nerve function recovery | inhibit the release of inflammatory factors, reduction of infarct size | [89,90] |
miR-21 | Promotes cell survival and proliferation | Tissue regeneration, wound healing | [102] |
miR-let-7 | Regulates cell proliferation and differentiation | tissue regeneration, angiogenesis | [91] |
miR-29b-3p | Modulates extracellular matrix remodeling | Fibrosis treatment, tissue repair, and fracture healing | [92] |
miR-126 | Promotes angiogenesis and neurogenesis | Cardiovascular regeneration, neurological disorders | [93] |
miR-133 | Reduced inflammation and reversed liver injury | Liver injury | [86] |
miR-146a-5p | Modulates immune responses and inflammation | Autoimmune diseases, inflammation modulation | [94] |
miR-155 | Modulates immune responses and inflammation | Immunomodulation, inflammatory disease therapy | [95] |
miR-210 | Induce vascularization, Promotes cellular adaptation to hypoxia | Ischemic diseases, tissue repair, bone regeneration, selective regeneration of ischemic heart | [96,97] |
miR-223 | Regulates neuronal cell apoptosis and immune cells reactions | Ischemic kidney, myocardial infarction | [98,99,100] |
miR-335 | Promotes osteoblast differentiation | bone fracture recovery | [103] |
miR-486 | Promotes cardiac regeneration and repair | Heart disease treatment, cardiac tissue repair | [104] |
miR-499 | Inhibit endometrial cancer growth and metastasis | anticancer | [105] |
miR-17-92 cluster | Increases neural plasticity an functional recovery | In treating stroke | [101] |
4.3. Anti-Apoptotic Factors
5. MSC-EVs as Diagnostics
Neurological Disorder | Diagnostic Uses and Applications | Examples | Reference |
---|---|---|---|
Alzheimer’s Disease | Biomarker Identification: EVs derived from MSCs carry specific proteins, microRNAs, and other molecules that can be analyzed to identify early biomarkers of Alzheimer’s disease. | Exosome-derived tau and amyloid-beta levels in CSF can indicate Alzheimer’s progression | [123,124] |
Detection of Aβ42 in CSF or blood to aid in early Alzheimer’s diagnosis. | Aβ42. | [127] | |
Tracking disease progression, monitoring treatment response, and early detection. | Downregulation of miR-212 and miR-132-enriched EVs in AD | [126] | |
Assessing the effectiveness of therapeutic interventions. | EVs containing Tau protein | [125] | |
Parkinson’s Disease | Biomarker Detection: EVs released by MSCs carry specific proteins and microRNAs that can serve as biomarkers to detect early signs of Parkinson’s disease. | α-synuclein and DJ-1 levels in MSC-EVs from CSF as a potential biomarker for Parkinson’s | [130] |
Monitoring disease progression by assessing miR-34a levels in EVs. | miR-34a over-expression in PD | [129] | |
Biomarkers to detect PD. | miR-124-enriched EVs in PD | [128] | |
Multiple Sclerosis | Quantification of myelin EVs in CSF as a potential biomarker for disease activity and progression. | Myelin-derived EVs | [131] |
Amyotrophic Lateral Sclerosis | Early diagnosis and predicting disease outcome. | ALS-related proteins, including SOD1, TDP-43, pTDP-43, and FUS | [132] |
Huntington’s Disease | Elevated total Huntingtin levels in EVs from plasma of HD groups compared to controls. | Huntingtin protein | [133] |
6. MSC-EVs in Alzheimer’s Disease
7. MSC-EVs in Parkinson’s Diseases
8. MSC-EVs in Multiple Sclerosis
9. MSC-EVs in Amyotrophic Lateral Sclerosis
10. MSC-EVs in Huntington’s Disease
11. Challenge and Future Perspectives
12. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Ding, D.C.; Shyu, W.C.; Lin, S.Z. Mesenchymal stem cells. Cell Transplant. 2011, 20, 5–14. [Google Scholar] [CrossRef]
- Kim, E.-J.; Kim, N.; Cho, S.-G. The potential use of mesenchymal stem cells in hematopoietic stem cell transplantation. Exp. Mol. Med. 2013, 45, e2. [Google Scholar] [CrossRef]
- Appaix, F.; Nissou, M.F.; van der Sanden, B.; Dreyfus, M.; Berger, F.; Issartel, J.P.; Wion, D. Brain mesenchymal stem cells: The other stem cells of the brain? World J. Stem Cells 2014, 6, 134–143. [Google Scholar] [CrossRef]
- Ghaneialvar, H.; Soltani, L.; Rahmani, H.R.; Lotfi, A.S.; Soleimani, M. Characterization and Classification of Mesenchymal Stem Cells in Several Species Using Surface Markers for Cell Therapy Purposes. Indian J. Clin. Biochem. IJCB 2018, 33, 46–52. [Google Scholar] [CrossRef]
- Wang, S.; Qu, X.; Zhao, R.C. Clinical applications of mesenchymal stem cells. J. Hematol. Oncol. 2012, 5, 19. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.-H.; Tseng, W.-C.; Yang, C.-Y.; Tarng, D.-C. The anti-inflammatory, anti-oxidative, and anti-apoptotic benefits of stem cells in acute ischemic kidney injury. Int. J. Mol. Sci. 2019, 20, 3529. [Google Scholar] [CrossRef] [PubMed]
- Harrell, C.R.; Djonov, V.; Volarevic, V. The cross-talk between mesenchymal stem cells and immune cells in tissue repair and regeneration. Int. J. Mol. Sci. 2021, 22, 2472. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, J.K.; Saini, D.; Chaudhary, P.K.; Maurya, A.; Verma, G.K.; Gupta, A.K.; Roshan, R.; Vats, T.K.; Garg, N.; Yadav, D. Exploring the immunomodulatory aspect of mesenchymal stem cells for treatment of severe coronavirus disease 19. Cells 2022, 11, 2175. [Google Scholar] [CrossRef]
- Gao, L.; Xu, W.; Li, T.; Chen, J.; Shao, A.; Yan, F.; Chen, G. Stem cell therapy: A promising therapeutic method for intracerebral hemorrhage. Cell Transplant. 2018, 27, 1809–1824. [Google Scholar] [CrossRef]
- Oliveri, R.S.; Bello, S.; Biering-Sørensen, F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: Systematic review with meta-analyses of rat models. Neurobiol. Dis. 2014, 62, 338–353. [Google Scholar] [CrossRef]
- Hofer, H.R.; Tuan, R.S. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res. Ther. 2016, 7, 131. [Google Scholar] [CrossRef] [PubMed]
- Tahmasebi, F.; Barati, S. Effects of mesenchymal stem cell transplantation on spinal cord injury patients. Cell Tissue Res. 2022, 389, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed]
- He, X.; Zhang, C.; Amirsaadat, S.; Jalil, A.T.; Kadhim, M.M.; Abasi, M.; Pilehvar, Y. Curcumin-Loaded Mesenchymal Stem Cell–Derived Exosomes Efficiently Attenuate Proliferation and Inflammatory Response in Rheumatoid Arthritis Fibroblast-Like Synoviocytes. Appl. Biochem. Biotechnol. 2023, 195, 51–67. [Google Scholar] [CrossRef]
- Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef]
- Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L. 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]
- Wade, D.T. Epidemiology of disabling neurological disease: How and why does disability occur? J. Neurol. Neurosurg. Psychiatry 1997, 63, S11–S18. [Google Scholar] [CrossRef]
- Kennedy-Malone, L. Central and Peripheral Nervous System Disorders. Adv. Pract. Nurs. Care Older Adults 2018, 328. [Google Scholar]
- Lilamand, M.; Hourregue, C.; Paquet, C. Interest of biological biomarkers in the diagnostic approach of neurocognitive disorders in the elderly. Rev. Neurol. 2020, 176, 677–683. [Google Scholar] [CrossRef] [PubMed]
- Thompson, A.G.; Gray, E.; Heman-Ackah, S.M.; Mäger, I.; Talbot, K.; El Andaloussi, S.; Wood, M.J.; Turner, M.R. Extracellular vesicles in neurodegenerative disease—Pathogenesis to biomarkers. Nat. Rev. Neurol. 2016, 12, 346–357. [Google Scholar] [CrossRef]
- Rao, S.M. Neuropsychology of multiple sclerosis. Curr. Opin. Neurol. 1995, 8, 216–220. [Google Scholar] [CrossRef] [PubMed]
- Brown, R.; Marsden, C. How common is dementia in Parkinson’s disease? Lancet 1984, 324, 1262–1265. [Google Scholar] [CrossRef] [PubMed]
- Gaubert, F.; Chainay, H. Decision-Making Competence in Patients with Alzheimer’s Disease: A Review of the Literature. Neuropsychol. Rev. 2021, 31, 267–287. [Google Scholar] [CrossRef]
- Moujalled, D.; Strasser, A.; Liddell, J.R. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 2021, 28, 2029–2044. [Google Scholar] [CrossRef] [PubMed]
- Wilson, D.M.; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of neurodegenerative diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef]
- Wang, W.; Li, M.; Chen, Z.; Xu, L.; Chang, M.; Wang, K.; Deng, C.; Gu, Y.; Zhou, S.; Shen, Y. Biogenesis and function of extracellular vesicles in pathophysiological processes of skeletal muscle atrophy. Biochem. Pharmacol. 2022, 198, 114954. [Google Scholar] [CrossRef] [PubMed]
- Patil, A.A.; Rhee, W.J. Exosomes: Biogenesis, composition, functions, and their role in pre-metastatic niche formation. Biotechnol. Bioprocess Eng. 2019, 24, 689–701. [Google Scholar] [CrossRef]
- Hassanpour, M.; Rezaie, J.; Nouri, M.; Panahi, Y. The role of extracellular vesicles in COVID-19 virus infection. Infect. Genet. Evol. 2020, 85, 104422. [Google Scholar] [CrossRef]
- Tricarico, C.; Clancy, J.; D’Souza-Schorey, C. Biology and biogenesis of shed microvesicles. Small GTPases 2017, 8, 220–232. [Google Scholar] [CrossRef]
- Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef]
- Borges, F.T.; Reis, L.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef] [PubMed]
- Atkin-Smith, G.K.; Tixeira, R.; Paone, S.; Mathivanan, S.; Collins, C.; Liem, M.; Goodall, K.J.; Ravichandran, K.S.; Hulett, M.D.; Poon, I.K. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat. Commun. 2015, 6, 7439. [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]
- Rezaie, J.; Nejati, V.; Mahmoodi, M.; Ahmadi, M. Mesenchymal stem cells derived extracellular vesicles: A promising nanomedicine for drug delivery system. Biochem. Pharmacol. 2022, 203, 115167. [Google Scholar] [CrossRef] [PubMed]
- Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. J. Transl. Med. 2020, 18, 449. [Google Scholar] [CrossRef]
- Benedikter, B.J.; Bouwman, F.G.; Vajen, T.; Heinzmann, A.C.; Grauls, G.; Mariman, E.C.; Wouters, E.F.; Savelkoul, P.H.; Lopez-Iglesias, C.; Koenen, R.R. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci. Rep. 2017, 7, 15297. [Google Scholar] [CrossRef]
- Stranska, R.; Gysbrechts, L.; Wouters, J.; Vermeersch, P.; Bloch, K.; Dierickx, D.; Andrei, G.; Snoeck, R. Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma. J. Transl. Med. 2018, 16, 1. [Google Scholar] [CrossRef]
- Gowen, A.; Shahjin, F.; Chand, S.; Odegaard, K.E.; Yelamanchili, S.V. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Challenges in Clinical Applications. Front. Cell Dev. Biol. 2020, 8, 149. [Google Scholar] [CrossRef]
- Almeria, C.; Kreß, S.; Weber, V.; Egger, D.; Kasper, C. Heterogeneity of mesenchymal stem cell-derived extracellular vesicles is highly impacted by the tissue/cell source and culture conditions. Cell Biosci. 2022, 12, 51. [Google Scholar] [CrossRef]
- Maumus, M.; Rozier, P.; Boulestreau, J.; Jorgensen, C.; Noël, D. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Opportunities and Challenges for Clinical Translation. Front. Bioeng. Biotechnol. 2020, 8, 997. [Google Scholar] [CrossRef]
- Clayton, A.; Boilard, E.; Buzas, E.I.; Cheng, L.; Falcón-Perez, J.M.; Gardiner, C.; Gustafson, D.; Gualerzi, A.; Hendrix, A.; Hoffman, A.; et al. Considerations towards a roadmap for collection, handling and storage of blood extracellular vesicles. J. Extracell. Vesicles 2019, 8, 1647027. [Google Scholar] [CrossRef] [PubMed]
- Liangsupree, T.; Multia, E.; Riekkola, M.-L. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A 2021, 1636, 461773. [Google Scholar] [CrossRef] [PubMed]
- Varderidou-Minasian, S.; Lorenowicz, M.J. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics 2020, 10, 5979–5997. [Google Scholar] [CrossRef] [PubMed]
- Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef]
- Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Shi, X.; Wang, B.; Feng, X.; Xu, Y.; Lu, K.; Sun, M. circRNAs and exosomes: A mysterious frontier for human cancer. Mol. Ther. -Nucleic Acids 2020, 19, 384–392. [Google Scholar] [CrossRef]
- Akers, J.C.; Ramakrishnan, V.; Kim, R.; Skog, J.; Nakano, I.; Pingle, S.; Kalinina, J.; Hua, W.; Kesari, S.; Mao, Y. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): A platform for glioblastoma biomarker development. PLoS ONE 2013, 8, e78115. [Google Scholar] [CrossRef]
- Pisitkun, T.; Shen, R.-F.; Knepper, M.A. Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. USA 2004, 101, 13368–13373. [Google Scholar] [CrossRef]
- Hornick, N.I.; Huan, J.; Doron, B.; Goloviznina, N.A.; Lapidus, J.; Chang, B.H.; Kurre, P. Serum exosome microRNA as a minimally-invasive early biomarker of AML. Sci. Rep. 2015, 5, 11295. [Google Scholar] [CrossRef]
- Yoshida, Y.; Yamamoto, H.; Morita, R.; Oikawa, R.; Matsuo, Y.; Maehata, T.; Nosho, K.; Watanabe, Y.; Yasuda, H.; Itoh, F. Detection of DNA methylation of gastric juice-derived exosomes in gastric cancer. Integr. Mol. Med. 2014, 1, 17–21. [Google Scholar]
- Goto, T.; Fujiya, M.; Konishi, H.; Sasajima, J.; Fujibayashi, S.; Hayashi, A.; Utsumi, T.; Sato, H.; Iwama, T.; Ijiri, M. An elevated expression of serum exosomal microRNA-191,− 21,− 451a of pancreatic neoplasm is considered to be efficient diagnostic marker. BMC Cancer 2018, 18, 116. [Google Scholar] [CrossRef] [PubMed]
- Burtenshaw, D.; Regan, B.; Owen, K.; Collins, D.; McEneaney, D.; Megson, I.L.; Redmond, E.M.; Cahill, P.A. Exosomal Composition, Biogenesis and Profiling Using Point-of-Care Diagnostics—Implications for Cardiovascular Disease. Front. Cell Dev. Biol. 2022, 10, 853451. [Google Scholar] [CrossRef] [PubMed]
- Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.m.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef] [PubMed]
- Bobrie, A.; Colombo, M.; Raposo, G.; Théry, C. Exosome secretion: Molecular mechanisms and roles in immune responses. Traffic 2011, 12, 1659–1668. [Google Scholar] [CrossRef]
- Chaput, N.; Théry, C. Exosomes: Immune properties and potential clinical implementations. Semin. Immunopathol. 2011, 33, 419–440. [Google Scholar] [CrossRef]
- Fauré, J.; Lachenal, G.; Court, M.; Hirrlinger, J.; Chatellard-Causse, C.; Blot, B.; Grange, J.; Schoehn, G.; Goldberg, Y.; Boyer, V. Exosomes are released by cultured cortical neurones. Mol. Cell. Neurosci. 2006, 31, 642–648. [Google Scholar] [CrossRef]
- Lachenal, G.; Pernet-Gallay, K.; Chivet, M.; Hemming, F.J.; Belly, A.; Bodon, G.; Blot, B.; Haase, G.; Goldberg, Y.; Sadoul, R. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell. Neurosci. 2011, 46, 409–418. [Google Scholar] [CrossRef]
- Sprague, D.L.; Elzey, B.D.; Crist, S.A.; Waldschmidt, T.J.; Jensen, R.J.; Ratliff, T.L. Platelet-mediated modulation of adaptive immunity: Unique delivery of CD154 signal by platelet-derived membrane vesicles. Blood J. Am. Soc. Hematol. 2008, 111, 5028–5036. [Google Scholar] [CrossRef]
- Van der Pol, E.; Böing, A.; Gool, E.; Nieuwland, R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J. Thromb. Haemost. 2016, 14, 48–56. [Google Scholar] [CrossRef]
- Balaj, L.; Lessard, R.; Dai, L.; Cho, Y.-J.; Pomeroy, S.L.; Breakefield, X.O.; Skog, J. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2011, 2, 180. [Google Scholar] [CrossRef] [PubMed]
- Hunter, M.P.; Ismail, N.; Zhang, X.; Aguda, B.D.; Lee, E.J.; Yu, L.; Xiao, T.; Schafer, J.; Lee, M.-L.T.; Schmittgen, T.D. Detection of microRNA expression in human peripheral blood microvesicles. PLoS ONE 2008, 3, e3694. [Google Scholar] [CrossRef] [PubMed]
- Castellana, D.; Zobairi, F.; Martinez, M.C.; Panaro, M.A.; Mitolo, V.; Freyssinet, J.-M.; Kunzelmann, C. Membrane microvesicles as actors in the establishment of a favorable prostatic tumoral niche: A role for activated fibroblasts and CX3CL1-CX3CR1 axis. Cancer Res. 2009, 69, 785–793. [Google Scholar] [CrossRef] [PubMed]
- Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef]
- Dhondt, B.; Rousseau, Q.; De Wever, O.; Hendrix, A. Function of extracellular vesicle-associated miRNAs in metastasis. Cell Tissue Res. 2016, 365, 621–641. [Google Scholar] [CrossRef]
- Williams, C.; Royo, F.; Aizpurua-Olaizola, O.; Pazos, R.; Boons, G.-J.; Reichardt, N.-C.; Falcon-Perez, J.M. Glycosylation of extracellular vesicles: Current knowledge, tools and clinical perspectives. J. Extracell. Vesicles 2018, 7, 1442985. [Google Scholar] [CrossRef]
- Hristov, M.; Erl, W.; Linder, S.; Weber, P.C. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 2004, 104, 2761–2766. [Google Scholar] [CrossRef]
- Xu, X.; Lai, Y.; Hua, Z.-C. Apoptosis and apoptotic body: Disease message and therapeutic target potentials. Biosci. Rep. 2019, 39, BSR20180992. [Google Scholar] [CrossRef] [PubMed]
- Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
- Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef]
- Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Gianni-Barrera, R.; Trani, M.; Fontanellaz, C.; Heberer, M.; Djonov, V.; Hlushchuk, R.; Banfi, A. VEGF over-expression in skeletal muscle induces angiogenesis by intussusception rather than sprouting. Angiogenesis 2013, 16, 123–136. [Google Scholar] [CrossRef] [PubMed]
- Gong, M.; Yu, B.; Wang, J.; Wang, Y.; Liu, M.; Paul, C.; Millard, R.W.; Xiao, D.-S.; Ashraf, M.; Xu, M. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget 2017, 8, 45200. [Google Scholar] [CrossRef] [PubMed]
- Yadav, L.; Puri, N.; Rastogi, V.; Satpute, P.; Sharma, V. Tumour angiogenesis and angiogenic inhibitors: A review. J. Clin. Diagn. Res. JCDR 2015, 9, XE01. [Google Scholar] [CrossRef] [PubMed]
- Harrell, C.R.; Fellabaum, C.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Molecular mechanisms responsible for therapeutic potential of mesenchymal stem cell-derived secretome. Cells 2019, 8, 467. [Google Scholar] [CrossRef]
- Carreras-Planella, L.; Monguió-Tortajada, M.; Borràs, F.E.; Franquesa, M. Immunomodulatory effect of MSC on B cells is independent of secreted extracellular vesicles. Front. Immunol. 2019, 10, 1288. [Google Scholar] [CrossRef]
- Luther, K.M.; Haar, L.; McGuinness, M.; Wang, Y.; Lynch IV, T.L.; Phan, A.; Song, Y.; Shen, Z.; Gardner, G.; Kuffel, G. Exosomal miR-21a-5p mediates cardioprotection by mesenchymal stem cells. J. Mol. Cell. Cardiol. 2018, 119, 125–137. [Google Scholar] [CrossRef]
- Ji, W.; Jiang, W.; Li, M.; Li, J.; Li, Z. miR-21 deficiency contributes to the impaired protective effects of obese rat mesenchymal stem cell-derived exosomes against spinal cord injury. Biochimie 2019, 167, 171–178. [Google Scholar] [CrossRef]
- Lou, G.; Yang, Y.; Liu, F.; Ye, B.; Chen, Z.; Zheng, M.; Liu, Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J. Cell. Mol. Med. 2017, 21, 2963–2973. [Google Scholar] [CrossRef]
- Eirin, A.; Zhu, X.-Y.; Puranik, A.S.; Tang, H.; McGurren, K.A.; van Wijnen, A.J.; Lerman, A.; Lerman, L.O. Mesenchymal stem cell–derived extracellular vesicles attenuate kidney inflammation. Kidney Int. 2017, 92, 114–124. [Google Scholar] [CrossRef] [PubMed]
- Chaubey, S.; Thueson, S.; Ponnalagu, D.; Alam, M.A.; Gheorghe, C.P.; Aghai, Z.; Singh, H.; Bhandari, V. Early gestational mesenchymal stem cell secretome attenuates experimental bronchopulmonary dysplasia in part via exosome-associated factor TSG-6. Stem Cell Res. Ther. 2018, 9, 173. [Google Scholar] [CrossRef] [PubMed]
- Shen, B.; Liu, J.; Zhang, F.; Wang, Y.; Qin, Y.; Zhou, Z.; Qiu, J.; Fan, Y. CCR2 positive exosome released by mesenchymal stem cells suppresses macrophage functions and alleviates ischemia/reperfusion-induced renal injury. Stem Cells Int. 2016, 2016, 1240301. [Google Scholar] [CrossRef] [PubMed]
- Reis, M.; Mavin, E.; Nicholson, L.; Green, K.; Dickinson, A.M.; Wang, X.-n. Mesenchymal stromal cell-derived extracellular vesicles attenuate dendritic cell maturation and function. Front. Immunol. 2018, 9, 2538. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Gu, H.; Qin, D.; Yang, L.; Huang, W.; Essandoh, K.; Wang, Y.; Caldwell, C.C.; Peng, T.; Zingarelli, B.; et al. Exosomal miR-223 Contributes to Mesenchymal Stem Cell-Elicited Cardioprotection in Polymicrobial Sepsis. Sci. Rep. 2015, 5, 13721. [Google Scholar] [CrossRef]
- Chen, L.; Lu, F.-b.; Chen, D.-z.; Wu, J.-l.; Xu, L.-m.; Zheng, M.-h.; Li, H.; Huang, Y.; Jin, X.-y.; Gong, Y.-w. BMSCs-derived miR-223-containing exosomes contribute to liver protection in experimental autoimmune hepatitis. Mol. Immunol. 2018, 93, 38–46. [Google Scholar] [CrossRef]
- Martin-Rufino, J.D.; Espinosa-Lara, N.; Osugui, L.; Sanchez-Guijo, F. Targeting the immune system with mesenchymal stromal cell-derived extracellular vesicles: What is the Cargo’s mechanism of action? Front. Bioeng. Biotechnol. 2019, 7, 308. [Google Scholar] [CrossRef]
- Kandeel, M. Oncogenic Viruses-Encoded microRNAs and Their Role in the Progression of Cancer: Emerging Targets for Antiviral and Anticancer Therapies. Pharmaceuticals 2023, 16, 485. [Google Scholar] [CrossRef]
- Sheng, Y.; Zhou, X.; Wang, J.; Shen, H.; Wu, S.; Guo, W.; Yang, Y. MSC derived EV loaded with miRNA-22 inhibits the inflammatory response and nerve function recovery after spinal cord injury in rats. J. Cell. Mol. Med. 2021, 25, 10268–10278. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Gong, M.; Wang, M.; Xu, J.; Yu, B.; Wang, Y.-G.; Liu, M.; Ashraf, M.; Xu, M. Nano-sized extracellular vesicles secreted from GATA-4 modified mesenchymal stem cells promote angiogenesis by delivering Let-7 miRNAs. Cells 2022, 11, 1573. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Gao, J.; Gao, F.; Zhao, Y.; Deng, B.; Mu, X.; Xu, L. Extracellular vesicles-encapsulated microRNA-29b-3p from bone marrow-derived mesenchymal stem cells promotes fracture healing via modulation of the PTEN/PI3K/AKT axis. Exp. Cell Res. 2022, 412, 113026. [Google Scholar] [CrossRef]
- Huang, J.-H.; Xu, Y.; Yin, X.-M.; Lin, F.-Y. Exosomes derived from miR-126-modified MSCs promote angiogenesis and neurogenesis and attenuate apoptosis after spinal cord injury in rats. Neuroscience 2020, 424, 133–145. [Google Scholar] [CrossRef] [PubMed]
- Pei, Z.; Cen, J.; Zhang, X.; Gong, C.; Sun, M.; Meng, W.; Mao, G.; Wan, J.; Hu, B.; He, X. MiR-146a-5p delivered by hucMSC extracellular vesicles modulates the inflammatory response to sulfur mustard-induced acute lung injury. Stem Cell Res. Ther. 2023, 14, 149. [Google Scholar] [CrossRef]
- Pers, Y.-M.; Bony, C.; Duroux-Richard, I.; Bernard, L.; Maumus, M.; Assou, S.; Barry, F.; Jorgensen, C.; Noël, D. miR-155 contributes to the immunoregulatory function of human mesenchymal stem cells. Front. Immunol. 2021, 12, 624024. [Google Scholar] [CrossRef]
- Vieira, J.M.F.; Zamproni, L.N.; Wendt, C.H.; Rocha de Miranda, K.; Lindoso, R.S.; Won Han, S. Overexpression of mir-135b and mir-210 in mesenchymal stromal cells for the enrichment of extracellular vesicles with angiogenic factors. PLoS ONE 2022, 17, e0272962. [Google Scholar] [CrossRef] [PubMed]
- Song, B.-W.; Lee, C.Y.; Kim, R.; Kim, W.J.; Lee, H.W.; Lee, M.Y.; Kim, J.; Jeong, J.-Y.; Chang, W. Multiplexed targeting of miRNA-210 in stem cell-derived extracellular vesicles promotes selective regeneration in ischemic hearts. Exp. Mol. Med. 2021, 53, 695–708. [Google Scholar] [CrossRef]
- Yang, M.; Liao, M.; Liu, R.; Zhang, Q.; Zhang, S.; He, Y.; Jin, J.; Zhang, P.; Zhou, L. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles loaded with miR-223 ameliorate myocardial infarction through P53/S100A9 axis. Genomics 2022, 114, 110319. [Google Scholar] [CrossRef]
- Sun, Z.; Gao, Z.; Wu, J.; Zheng, X.; Jing, S.; Wang, W. MSC-derived extracellular vesicles activate mitophagy to alleviate renal ischemia/reperfusion injury via the miR-223-3p/NLRP3 axis. Stem Cells Int. 2022, 2022, 6852661. [Google Scholar] [CrossRef]
- Wei, H.; Xu, Y.; Chen, Q.; Chen, H.; Zhu, X.; Li, Y. Mesenchymal stem cell-derived exosomal miR-223 regulates neuronal cell apoptosis. Cell Death Dis. 2020, 11, 290. [Google Scholar] [CrossRef]
- Xin, H.; Katakowski, M.; Wang, F.; Qian, J.-Y.; Liu, X.S.; Ali, M.M.; Buller, B.; Zhang, Z.G.; Chopp, M. MicroRNA-17–92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 2017, 48, 747–753. [Google Scholar] [CrossRef]
- Liu, X.; Li, X.; Wu, G.; Qi, P.; Zhang, Y.; Liu, Z.; Li, X.; Yu, Y.; Ye, X.; Li, Y. Umbilical cord mesenchymal stem cell-derived small extracellular vesicles deliver miR-21 to promote corneal epithelial wound healing through PTEN/PI3K/Akt pathway. Stem Cells Int. 2022, 2022, 1252557. [Google Scholar] [CrossRef]
- Hu, H.; Wang, D.; Li, L.; Yin, H.; He, G.; Zhang, Y. Role of microRNA-335 carried by bone marrow mesenchymal stem cells-derived extracellular vesicles in bone fracture recovery. Cell Death Dis. 2021, 12, 156. [Google Scholar] [CrossRef] [PubMed]
- Manu, M.S.; Hohjoh, H.; Yamamura, T. Extracellular vesicles as pro-and anti-inflammatory mediators, biomarkers and potential therapeutic agents in multiple sclerosis. Aging Dis. 2021, 12, 1451. [Google Scholar] [CrossRef] [PubMed]
- Jing, L.; Hua, X.; Yuanna, D.; Rukun, Z.; Junjun, M. Exosomal miR-499a-5p inhibits endometrial cancer growth and metastasis via targeting VAV3. Cancer Manag. Res. 2020, 12, 13541–13552. [Google Scholar] [CrossRef] [PubMed]
- Kossl, J.; Bohacova, P.; Hermankova, B.; Javorkova, E.; Zajicova, A.; Holan, V. Antiapoptotic Properties of Mesenchymal Stem Cells in a Mouse Model of Corneal Inflammation. Stem Cells Dev. 2021, 30, 418–427. [Google Scholar] [CrossRef] [PubMed]
- Bernard, O.; Jeny, F.; Uzunhan, Y.; Dondi, E.; Terfous, R.; Label, R.; Sutton, A.; Larghero, J.; Vanneaux, V.; Nunes, H. Mesenchymal stem cells reduce hypoxia-induced apoptosis in alveolar epithelial cells by modulating HIF and ROS hypoxic signaling. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2018, 314, L360–L371. [Google Scholar] [CrossRef]
- Bari, E.; Ferrarotti, I.; Saracino, L.; Perteghella, S.; Torre, M.L.; Corsico, A.G. Mesenchymal stromal cell secretome for severe COVID-19 infections: Premises for the therapeutic use. Cells 2020, 9, 924. [Google Scholar] [CrossRef]
- Műzes, G.; Sipos, F. Mesenchymal stem cell-derived secretome: A potential therapeutic option for autoimmune and immune-mediated inflammatory diseases. Cells 2022, 11, 2300. [Google Scholar] [CrossRef]
- Shologu, N.; Scully, M.; Laffey, J.G.; O’Toole, D. Human Mesenchymal Stem Cell Secretome from Bone Marrow or Adipose-Derived Tissue Sources for Treatment of Hypoxia-Induced Pulmonary Epithelial Injury. Int. J. Mol. Sci. 2018, 19, 2996. [Google Scholar] [CrossRef]
- González-González, A.; García-Sánchez, D.; Dotta, M.; Rodríguez-Rey, J.C.; Pérez-Campo, F.M. Mesenchymal stem cells secretome: The cornerstone of cell-free regenerative medicine. World J. Stem Cells 2020, 12, 1529–1552. [Google Scholar] [CrossRef]
- Han, Y.; Yang, J.; Fang, J.; Zhou, Y.; Candi, E.; Wang, J.; Hua, D.; Shao, C.; Shi, Y. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct. Target. Ther. 2022, 7, 92. [Google Scholar] [CrossRef]
- An, S.; Anwar, K.; Ashraf, M.; Lee, H.; Jung, R.; Koganti, R.; Ghassemi, M.; Djalilian, A.R. Wound-Healing Effects of Mesenchymal Stromal Cell Secretome in the Cornea and the Role of Exosomes. Pharmaceutics 2023, 15, 1486. [Google Scholar] [CrossRef]
- Jin, Y.; Li, S.; Yu, Q.; Chen, T.; Liu, D. Application of stem cells in regeneration medicine. MedComm 2023, 4, e291. [Google Scholar] [CrossRef]
- Lan, T.; Luo, M.; Wei, X. Mesenchymal stem/stromal cells in cancer therapy. J. Hematol. Oncol. 2021, 14, 195. [Google Scholar] [CrossRef]
- Pang, S.H.M.; D’Rozario, J.; Mendonca, S.; Bhuvan, T.; Payne, N.L.; Zheng, D.; Hisana, A.; Wallis, G.; Barugahare, A.; Powell, D.; et al. Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat. Commun. 2021, 12, 6495. [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]
- Van Poll, D.; Parekkadan, B.; Cho, C.H.; Berthiaume, F.; Nahmias, Y.; Tilles, A.W.; Yarmush, M.L. Mesenchymal stem cell–derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatology 2008, 47, 1634–1643. [Google Scholar] [CrossRef]
- Gazdic, M.; Simovic Markovic, B.; Vucicevic, L.; Nikolic, T.; Djonov, V.; Arsenijevic, N.; Trajkovic, V.; Lukic, M.L.; Volarevic, V. Mesenchymal stem cells protect from acute liver injury by attenuating hepatotoxicity of liver natural killer T cells in an inducible nitric oxide synthase-and indoleamine 2, 3-dioxygenase-dependent manner. J. Tissue Eng. Regen. Med. 2018, 12, e1173–e1185. [Google Scholar] [CrossRef]
- Xagorari, A.; Siotou, E.; Yiangou, M.; Tsolaki, E.; Bougiouklis, D.; Sakkas, L.; Fassas, A.; Anagnostopoulos, A. Protective effect of mesenchymal stem cell-conditioned medium on hepatic cell apoptosis after acute liver injury. Int. J. Clin. Exp. Pathol. 2013, 6, 831. [Google Scholar]
- Milosavljevic, N.; Gazdic, M.; Simovic Markovic, B.; Arsenijevic, A.; Nurkovic, J.; Dolicanin, Z.; Djonov, V.; Lukic, M.L.; Volarevic, V. Mesenchymal stem cells attenuate acute liver injury by altering ratio between interleukin 17 producing and regulatory natural killer T cells. Liver Transplant. 2017, 23, 1040–1050. [Google Scholar] [CrossRef]
- Fabre, T.; Molina, M.F.; Soucy, G.; Goulet, J.-P.; Willems, B.; Villeneuve, J.-P.; Bilodeau, M.; Shoukry, N.H. Type 3 cytokines IL-17A and IL-22 drive TGF-β–dependent liver fibrosis. Sci. Immunol. 2018, 3, eaar7754. [Google Scholar] [CrossRef]
- Hampel, H.; Bürger, K.; Teipel, S.J.; Bokde, A.L.; Zetterberg, H.; Blennow, K. Core candidate neurochemical and imaging biomarkers of Alzheimer’s disease. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2008, 4, 38–48. [Google Scholar] [CrossRef] [PubMed]
- Guix, F.X.; Corbett, G.T.; Cha, D.J.; Mustapic, M.; Liu, W.; Mengel, D.; Chen, Z.; Aikawa, E.; Young-Pearse, T.; Kapogiannis, D. Detection of aggregation-competent tau in neuron-derived extracellular vesicles. Int. J. Mol. Sci. 2018, 19, 663. [Google Scholar] [CrossRef]
- Bell, B.J.; Malvankar, M.M.; Tallon, C.; Slusher, B.S. Sowing the seeds of discovery: Tau-propagation models of Alzheimer’s disease. ACS Chem. Neurosci. 2020, 11, 3499–3509. [Google Scholar] [CrossRef] [PubMed]
- Cha, D.J.; Mengel, D.; Mustapic, M.; Liu, W.; Selkoe, D.J.; Kapogiannis, D.; Galasko, D.; Rissman, R.A.; Bennett, D.A.; Walsh, D.M. miR-212 and miR-132 Are Downregulated in Neurally Derived Plasma Exosomes of Alzheimer’s Patients. Front. Neurosci. 2019, 13, 1208. [Google Scholar] [CrossRef] [PubMed]
- Molinuevo, J.L.; Ayton, S.; Batrla, R.; Bednar, M.M.; Bittner, T.; Cummings, J.; Fagan, A.M.; Hampel, H.; Mielke, M.M.; Mikulskis, A. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 2018, 136, 821–853. [Google Scholar] [CrossRef]
- Angelopoulou, E.; Paudel, Y.N.; Piperi, C. miR-124 and Parkinson’s disease: A biomarker with therapeutic potential. Pharmacol. Res. 2019, 150, 104515. [Google Scholar] [CrossRef]
- Grossi, I.; Radeghieri, A.; Paolini, L.; Porrini, V.; Pilotto, A.; Padovani, A.; Marengoni, A.; Barbon, A.; Bellucci, A.; Pizzi, M. MicroRNA-34a-5p expression in the plasma and in its extracellular vesicle fractions in subjects with Parkinson’s disease: An exploratory study. Int. J. Mol. Med. 2021, 47, 533–546. [Google Scholar] [CrossRef]
- Hong, Z.; Shi, M.; Chung, K.A.; Quinn, J.F.; Peskind, E.R.; Galasko, D.; Jankovic, J.; Zabetian, C.P.; Leverenz, J.B.; Baird, G. DJ-1 and α-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain 2010, 133, 713–726. [Google Scholar] [CrossRef]
- Jagot, F.; Davoust, N. Is it worth considering circulating microRNAs in multiple sclerosis? Front. Immunol. 2016, 7, 129. [Google Scholar] [CrossRef]
- Gagliardi, D.; Bresolin, N.; Comi, G.P.; Corti, S. Extracellular vesicles and amyotrophic lateral sclerosis: From misfolded protein vehicles to promising clinical biomarkers. Cell. Mol. Life Sci. CMLS 2021, 78, 561–572. [Google Scholar] [CrossRef]
- Ananbeh, H.; Novak, J.; Juhas, S.; Juhasova, J.; Klempir, J.; Doleckova, K.; Rysankova, I.; Turnovcova, K.; Hanus, J.; Hansikova, H. Huntingtin co-isolates with small extracellular vesicles from blood plasma of TgHD and KI-HD pig models of Huntington’s disease and human blood plasma. Int. J. Mol. Sci. 2022, 23, 5598. [Google Scholar] [CrossRef]
- Sudar, K.M.; Nagaraj, P.; Nithisaa, S.; Aishwarya, R.; Aakash, M.; Lakshmi, S.I. Alzheimer’s Disease Analysis using Explainable Artificial Intelligence (XAI). In Proceedings of the 2022 International Conference on Sustainable Computing and Data Communication Systems (ICSCDS), Erode, India, 7–9 April 2022; pp. 419–423. [Google Scholar]
- Khan, S.; 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]
- De Strooper, B.; Karran, E. The cellular phase of Alzheimer’s disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef]
- Silvestro, S.; Valeri, A.; Mazzon, E. Aducanumab and its effects on tau pathology: Is this the turning point of amyloid hypothesis? Int. J. Mol. Sci. 2022, 23, 2011. [Google Scholar] [CrossRef]
- Du, X.; Wang, X.; Geng, M. Alzheimer’s disease hypothesis and related therapies. Transl. Neurodegener. 2018, 7, 2. [Google Scholar] [CrossRef]
- Kandeel, M.; Morsy, M.A.; El-lateef, A.; Hany, M.; Marzok, M.; El-Beltagi, H.; Alkhodair, K.; Albokhadaim, I.; Venugopala, K.N. Cognitive-and Memory-Enhancing Effects of Augmentin in Alzheimer’s Rats Through Regulation of Gene Expression and Neuronal Cell Apoptosis. Front. Pharmacol. 2023, 14, 635. [Google Scholar] [CrossRef]
- Birks, J.S.; Harvey, R.J. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst. Rev. 2018, 2018, CD001190. [Google Scholar] [CrossRef]
- Hamlett, E.D.; Goetzl, E.J.; Ledreux, A.; Vasilevko, V.; Boger, H.A.; LaRosa, A.; Clark, D.; Carroll, S.L.; Carmona-Iragui, M.; Fortea, J. Neuronal exosomes reveal Alzheimer’s disease biomarkers in Down syndrome. Alzheimer’s Dement. 2017, 13, 541–549. [Google Scholar] [CrossRef]
- Pusic, K.M.; Grinberg, Y.Y.; Kraig, R.P.; Pusic, A.D. Exosome-Based Therapeutics Against Neurodegenerative Disorders. U.S. Patent US20190160097A1, 30 May 2019. [Google Scholar]
- Abner, E.L.; Elahi, F.M.; Jicha, G.A.; Mustapic, M.; Al-Janabi, O.; Kramer, J.H.; Kapogiannis, D.; Goetzl, E.J. Endothelial-derived plasma exosome proteins in Alzheimer’s disease angiopathy. FASEB J. 2020, 34, 5967–5974. [Google Scholar] [CrossRef]
- Saman, S.; Kim, W.; Raya, M.; Visnick, Y.; Miro, S.; Saman, S.; Jackson, B.; McKee, A.C.; Alvarez, V.E.; Lee, N.C. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J. Biol. Chem. 2012, 287, 3842–3849. [Google Scholar] [CrossRef]
- Lewczuk, P.; Kornhuber, J.; Vanmechelen, E.; Peters, O.; Heuser, I.; Maier, W.; Jessen, F.; Bürger, K.; Hampel, H.; Frölich, L. Amyloid β peptides in plasma in early diagnosis of Alzheimer’s disease: A multicenter study with multiplexing. Exp. Neurol. 2010, 223, 366–370. [Google Scholar] [CrossRef]
- Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimer’s Dement. 2015, 11, 600–607.e601. [Google Scholar] [CrossRef]
- Clark, C.M.; Xie, S.; Chittams, J.; Ewbank, D.; Peskind, E.; Galasko, D.; Morris, J.C.; McKeel, D.W.; Farlow, M.; Weitlauf, S.L. Cerebrospinal fluid tau and β-amyloid: How well do these biomarkers reflect autopsy-confirmed dementia diagnoses? Arch. Neurol. 2003, 60, 1696–1702. [Google Scholar] [CrossRef]
- Riancho, J.; Vázquez-Higuera, J.L.; Pozueta, A.; Lage, C.; Kazimierczak, M.; Bravo, M.; Calero, M.; Gonalezález, A.; Rodríguez, E.; Lleó, A. MicroRNA profile in patients with Alzheimer’s disease: Analysis of miR-9-5p and miR-598 in raw and exosome enriched cerebrospinal fluid samples. J. Alzheimer’s Dis. 2017, 57, 483–491. [Google Scholar] [CrossRef]
- Mattsson, N.; Zetterberg, H.; Janelidze, S.; Insel, P.S.; Andreasson, U.; Stomrud, E.; Palmqvist, S.; Baker, D.; Hehir, C.A.T.; Jeromin, A. Plasma tau in Alzheimer disease. Neurology 2016, 87, 1827–1835. [Google Scholar] [CrossRef]
- Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016, 30, 4141–4148. [Google Scholar] [CrossRef]
- Wang, S.; Cesca, F.; Loers, G.; Schweizer, M.; Buck, F.; Benfenati, F.; Schachner, M.; Kleene, R. Synapsin I is an oligomannose-carrying glycoprotein, acts as an oligomannose-binding lectin, and promotes neurite outgrowth and neuronal survival when released via glia-derived exosomes. J. Neurosci. 2011, 31, 7275–7290. [Google Scholar] [CrossRef]
- 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. Elucidation of exosome migration across the blood–brain barrier model in vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef]
- Katsuda, T.; Oki, K.; Ochiya, T. Potential application of extracellular vesicles of human adipose tissue-derived mesenchymal stem cells in Alzheimer’s disease therapeutics. In Stem Cell Renewal and Cell-Cell Communication; Springer: Berlin/Heidelberg, Germany, 2014; pp. 171–181. [Google Scholar]
- 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. 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] [PubMed]
- 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]
- Elia, C.A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M. 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]
- De Godoy, M.A.; Saraiva, L.M.; de Carvalho, L.R.; Vasconcelos-dos-Santos, A.; Beiral, H.J.; Ramos, A.B.; de Paula Silva, L.R.; Leal, R.B.; Monteiro, V.H.; Braga, C.V. 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]
- 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. [Google Scholar] [CrossRef]
- Wang, H.; Liu, Y.; Li, J.; Wang, T.; Hei, Y.; Li, H.; Wang, X.; Wang, L.; Zhao, R.; Liu, W. Tail-vein injection of MSC-derived small extracellular vesicles facilitates the restoration of hippocampal neuronal morphology and function in APP/PS1 mice. Cell Death Discov. 2021, 7, 230. [Google Scholar] [CrossRef]
- Kim, H.Y.; Kwon, S.; Um, W.; Shin, S.; Kim, C.H.; Park, J.H.; Kim, B.S. Functional Extracellular Vesicles for Regenerative Medicine. Small 2022, 18, e2106569. [Google Scholar] [CrossRef]
- Yin, T.; Liu, Y.; Ji, W.; Zhuang, J.; Chen, X.; Gong, B.; Chu, J.; Liang, W.; Gao, J.; Yin, Y. Engineered mesenchymal stem cell-derived extracellular vesicles: A state-of-the-art multifunctional weapon against Alzheimer’s disease. Theranostics 2023, 13, 1264. [Google Scholar] [CrossRef]
- Jahangard, Y.; Monfared, H.; Moradi, A.; Zare, M.; Mirnajafi-Zadeh, J.; Mowla, S.J. Therapeutic Effects of Transplanted Exosomes Containing miR-29b to a Rat Model of Alzheimer’s Disease. Front. Neurosci. 2020, 14, 564. [Google Scholar] [CrossRef]
- Liu, H.; Jin, M.; Ji, M.; Zhang, W.; Liu, A.; Wang, T. Hypoxic pretreatment of adipose-derived stem cell exosomes improved cognition by delivery of circ-Epc1 and shifting microglial M1/M2 polarization in an Alzheimer’s disease mice model. Aging 2022, 14, 3070–3083. [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.; Vázquez-Méndez, 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. [Google Scholar]
- 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]
- Xin, H.; Li, Y.; Buller, B.; Katakowski, M.; Zhang, Y.; Wang, X.; Shang, X.; Zhang, Z.G.; Chopp, M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 2012, 30, 1556–1564. [Google Scholar] [CrossRef] [PubMed]
- Peng, H.; Li, Y.; Ji, W.; Zhao, R.; Lu, Z.; Shen, J.; Wu, Y.; Wang, J.; Hao, Q.; Wang, J.; et al. Intranasal Administration of Self-Oriented Nanocarriers Based on Therapeutic Exosomes for Synergistic Treatment of Parkinson’s Disease. ACS Nano 2022, 16, 869–884. [Google Scholar] [CrossRef]
- Mokarizadeh, A.; Delirezh, N.; Morshedi, A.; Mosayebi, G.; Farshid, A.-A.; Mardani, K. Microvesicles derived from mesenchymal stem cells: Potent organelles for induction of tolerogenic signaling. Immunol. Lett. 2012, 147, 47–54. [Google Scholar] [CrossRef]
- Li, Z.; Liu, F.; He, X.; Yang, X.; Shan, F.; Feng, J. Exosomes derived from mesenchymal stem cells attenuate inflammation and demyelination of the central nervous system in EAE rats by regulating the polarization of microglia. Int. Immunopharmacol. 2019, 67, 268–280. [Google Scholar] [CrossRef]
- Clark, K.; Zhang, S.; Barthe, S.; Kumar, P.; Pivetti, C.; Kreutzberg, N.; Reed, C.; Wang, Y.; Paxton, Z.; Farmer, D.; et al. Placental Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Myelin Regeneration in an Animal Model of Multiple Sclerosis. Cells 2019, 8, 1497. [Google Scholar] [CrossRef]
- Castelli, V.; Benedetti, E.; Antonosante, A.; Catanesi, M.; Pitari, G.; Ippoliti, R.; Cimini, A.; d’Angelo, M. Neuronal cells rearrangement during aging and neurodegenerative disease: Metabolism, oxidative stress and organelles dynamic. Front. Mol. Neurosci. 2019, 12, 132. [Google Scholar] [CrossRef]
- Kouli, A.; Torsney, K.M.; Kuan, W.-L. Parkinson’s Disease: Etiology, Neuropathology, and Pathogenesis; Codon Publications: Hong Lim Complex, Singapore, 2018; pp. 3–26. [Google Scholar]
- Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T.D. The prevalence of Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2014, 29, 1583–1590. [Google Scholar] [CrossRef]
- Vilaça-Faria, H.; Salgado, A.J.; Teixeira, F.G. Mesenchymal Stem Cells-derived Exosomes: A New Possible Therapeutic Strategy for Parkinson’s Disease? Cells 2019, 8, 118. [Google Scholar] [CrossRef] [PubMed]
- Upadhya, R.; Shetty, A.K. Extracellular Vesicles for the Diagnosis and Treatment of Parkinson’s Disease. Aging Dis. 2021, 12, 1438–1450. [Google Scholar] [CrossRef] [PubMed]
- d’Angelo, M.; Cimini, A.; Castelli, V. Insights into the Effects of Mesenchymal Stem Cell-Derived Secretome in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 5241. [Google Scholar] [CrossRef] [PubMed]
- Heris, R.M.; Shirvaliloo, M.; Abbaspour-Aghdam, S.; Hazrati, A.; Shariati, A.; Youshanlouei, H.R.; Niaragh, F.J.; Valizadeh, H.; Ahmadi, M. The potential use of mesenchymal stem cells and their exosomes in Parkinson’s disease treatment. Stem Cell Res. Ther. 2022, 13, 371. [Google Scholar] [CrossRef] [PubMed]
- Castelli, V.; d’Angelo, M.; Lombardi, F.; Alfonsetti, M.; Antonosante, A.; Catanesi, M.; Benedetti, E.; Palumbo, P.; Cifone, M.G.; Giordano, A. Effects of the probiotic formulation SLAB51 in in vitro and in vivo Parkinson’s disease models. Aging 2020, 12, 4641. [Google Scholar] [CrossRef]
- Caggiu, E.; Arru, G.; Hosseini, S.; Niegowska, M.; Sechi, G.; Zarbo, I.R.; Sechi, L.A. Inflammation, infectious triggers, and Parkinson’s disease. Front. Neurol. 2019, 10, 122. [Google Scholar] [CrossRef]
- Record, M.; Silvente-Poirot, S.; Poirot, M.; Wakelam, M.O. Extracellular vesicles: Lipids as key components of their biogenesis and functions. J. Lipid Res. 2018, 59, 1316–1324. [Google Scholar] [CrossRef]
- Yu, H.; Sun, T.; An, J.; Wen, L.; Liu, F.; Bu, Z.; Cui, Y.; Feng, J. Potential roles of exosomes in Parkinson’s disease: From pathogenesis, diagnosis, and treatment to prognosis. Front. Cell Dev. Biol. 2020, 8, 86. [Google Scholar] [CrossRef]
- Sonntag, K.-C. MicroRNAs and deregulated gene expression networks in neurodegeneration. Brain Res. 2010, 1338, 48–57. [Google Scholar] [CrossRef]
- Elangovan, A.; Venkatesan, D.; Selvaraj, P.; Pasha, M.Y.; Babu, H.W.S.; Iyer, M.; Narayanasamy, A.; Subramaniam, M.D.; Valsala Gopalakrishnan, A.; Kumar, N.S. miRNA in Parkinson’s disease: From pathogenesis to theranostic approaches. J. Cell. Physiol. 2023, 238, 329–354. [Google Scholar] [CrossRef]
- Baglio, S.R.; Rooijers, K.; Koppers-Lalic, D.; Verweij, F.J.; Lanzón, M.P.; Zini, N.; Naaijkens, B.; Perut, F.; Niessen, H.W.; Baldini, N. Human bone marrow-and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res. Ther. 2015, 6, 127. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.-J.; Patel, S.; Lee, S.-J. Intravesicular localization and exocytosis of α-synuclein and its aggregates. J. Neurosci. 2005, 25, 6016–6024. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Erviti, L.; Couch, Y.; Richardson, J.; Cooper, J.M.; Wood, M.J. Alpha-synuclein release by neurons activates the inflammatory response in a microglial cell line. Neurosci. Res. 2011, 69, 337–342. [Google Scholar] [CrossRef]
- Alvarez-Erviti, L.; Seow, Y.; Schapira, A.H.; Gardiner, C.; Sargent, I.L.; Wood, M.J.; Cooper, J.M. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol. Dis. 2011, 42, 360–367. [Google Scholar] [CrossRef]
- Ben Gedalya, T.; Loeb, V.; Israeli, E.; Altschuler, Y.; Selkoe, D.J.; Sharon, R. α-Synuclein and Polyunsaturated Fatty Acids Promote Clathrin-Mediated Endocytosis and Synaptic Vesicle Recycling. Traffic 2009, 10, 218–234. [Google Scholar] [CrossRef] [PubMed]
- Cabin, D.E.; Shimazu, K.; Murphy, D.; Cole, N.B.; Gottschalk, W.; McIlwain, K.L.; Orrison, B.; Chen, A.; Ellis, C.E.; Paylor, R. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking α-synuclein. J. Neurosci. 2002, 22, 8797–8807. [Google Scholar] [CrossRef] [PubMed]
- Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42. [Google Scholar] [CrossRef] [PubMed]
- Grey, M.; Dunning, C.J.; Gaspar, R.; Grey, C.; Brundin, P.; Sparr, E.; Linse, S. Acceleration of α-synuclein aggregation by exosomes. J. Biol. Chem. 2015, 290, 2969–2982. [Google Scholar] [CrossRef]
- Vella, L.J.; Hill, A.F.; Cheng, L. Focus on extracellular vesicles: Exosomes and their role in protein trafficking and biomarker potential in Alzheimer’s and Parkinson’s disease. Int. J. Mol. Sci. 2016, 17, 173. [Google Scholar] [CrossRef]
- Postuma, R.B.; Berg, D.; Stern, M.; Poewe, W.; Olanow, C.W.; Oertel, W.; Obeso, J.; Marek, K.; Litvan, I.; Lang, A.E. MDS clinical diagnostic criteria for Parkinson’s disease. Mov. Disord. 2015, 30, 1591–1601. [Google Scholar] [CrossRef]
- Chaudhuri, K.R.; Healy, D.G.; Schapira, A.H. Non-motor symptoms of Parkinson’s disease: Diagnosis and management. Lancet Neurol. 2006, 5, 235–245. [Google Scholar] [CrossRef]
- Canesi, M.; Giordano, R.; Lazzari, L.; Isalberti, M.; Isaias, I.U.; Benti, R.; Rampini, P.; Marotta, G.; Colombo, A.; Cereda, E.; et al. Finding a new therapeutic approach for no-option Parkinsonisms: Mesenchymal stromal cells for progressive supranuclear palsy. J. Transl. Med. 2016, 14, 127. [Google Scholar] [CrossRef] [PubMed]
- Ocansey, D.K.W.; Pei, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Improved therapeutics of modified mesenchymal stem cells: An update. J. Transl. Med. 2020, 18, 42. [Google Scholar] [CrossRef] [PubMed]
- Fričová, D.; Korchak, J.A.; Zubair, A.C. Challenges and translational considerations of mesenchymal stem/stromal cell therapy for Parkinson’s disease. NPJ Regen. Med. 2020, 5, 20. [Google Scholar] [CrossRef] [PubMed]
- Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release Off. J. Control. Release Soc. 2015, 207, 18–30. [Google Scholar] [CrossRef]
- Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release Off. J. Control. Release Soc. 2018, 287, 156–166. [Google Scholar] [CrossRef]
- Dutta, R.; Trapp, B.D. Mechanisms of neuronal dysfunction and degeneration in multiple sclerosis. Prog. Neurobiol. 2011, 93, 1–12. [Google Scholar] [CrossRef]
- Trapp, B.D.; Bö, L.; Mörk, S.; Chang, A. Pathogenesis of tissue injury in MS lesions. J. Neuroimmunol. 1999, 98, 49–56. [Google Scholar] [CrossRef] [PubMed]
- Weis, S.; Sonnberger, M.; Dunzinger, A.; Voglmayr, E.; Aichholzer, M.; Kleiser, R.; Strasser, P. Demyelinating Diseases: Multiple Sclerosis. In Imaging Brain Diseases; Springer: Berlin/Heidelberg, Germany, 2019; pp. 1071–1095. [Google Scholar]
- Lopez-Diego, R.S.; Weiner, H.L. Novel therapeutic strategies for multiple sclerosis—A multifaceted adversary. Nat. Rev. Drug Discov. 2008, 7, 909–925. [Google Scholar] [CrossRef]
- Lalu, M.M.; Sullivan, K.J.; Mei, S.H.; Moher, D.; Straus, A.; Fergusson, D.A.; Stewart, D.J.; Jazi, M.; MacLeod, M.; Winston, B. Evaluating mesenchymal stem cell therapy for sepsis with preclinical meta-analyses prior to initiating a first-in-human trial. Elife 2016, 5, e17850. [Google Scholar] [CrossRef]
- Rajan, T.S.; Giacoppo, S.; Diomede, F.; Ballerini, P.; Paolantonio, M.; Marchisio, M.; Piattelli, A.; Bramanti, P.; Mazzon, E.; Trubiani, O. The secretome of periodontal ligament stem cells from MS patients protects against EAE. Sci. Rep. 2016, 6, 38743. [Google Scholar] [CrossRef]
- Jafarinia, M.; Alsahebfosoul, F.; Salehi, H.; Eskandari, N.; Azimzadeh, M.; Mahmoodi, M.; Asgary, S.; Ganjalikhani Hakemi, M. Therapeutic effects of extracellular vesicles from human adipose-derived mesenchymal stem cells on chronic experimental autoimmune encephalomyelitis. J. Cell. Physiol. 2020, 235, 8779–8790. [Google Scholar] [CrossRef] [PubMed]
- Shahbaz, S.; Bozorgmehr, N.; Koleva, P.; Namdar, A.; Jovel, J.; Fava, R.A.; Elahi, S. CD71+ VISTA+ erythroid cells promote the development and function of regulatory T cells through TGF-β. PLoS Biol. 2018, 16, e2006649. [Google Scholar] [CrossRef] [PubMed]
- Chu, F.; Shi, M.; Zheng, C.; Shen, D.; Zhu, J.; Zheng, X.; Cui, L. The roles of macrophages and microglia in multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2018, 318, 1–7. [Google Scholar] [CrossRef]
- Song, G.J.; Suk, K. Pharmacological modulation of functional phenotypes of microglia in neurodegenerative diseases. Front. Aging Neurosci. 2017, 9, 139. [Google Scholar] [CrossRef]
- Farinazzo, A.; Angiari, S.; Turano, E.; Bistaffa, E.; Dusi, S.; Ruggieri, S.; Bonafede, R.; Mariotti, R.; Constantin, G.; Bonetti, B. Nanovesicles from adipose-derived mesenchymal stem cells inhibit T lymphocyte trafficking and ameliorate chronic experimental autoimmune encephalomyelitis. Sci. Rep. 2018, 8, 7473. [Google Scholar] [CrossRef] [PubMed]
- Gutiérrez-Fernández, M.; de la Cuesta, F.; Tallón, A.; Cuesta, I.; Fernández-Fournier, M.; Laso-García, F.; Gómez-de Frutos, M.C.; Díez-Tejedor, E.; Otero-Ortega, L. Potential Roles of Extracellular Vesicles as Biomarkers and a Novel Treatment Approach in Multiple Sclerosis. Int. J. Mol. Sci. 2021, 22, 9011. [Google Scholar] [CrossRef]
- Fayazi, N.; Sheykhhasan, M.; Asl, S.S.; Najafi, R. Stem Cell-Derived Exosomes: A New Strategy of Neurodegenerative Disease Treatment. Mol. Neurobiol. 2021, 58, 3494–3514. [Google Scholar] [CrossRef]
- Mohammadzadeh, A.; Pourfathollah, A.A.; Shahrokhi, S.; Fallah, A.; Tahoori, M.T.; Amari, A.; Forouzandeh, M.; Soleimani, M. Evaluation of AD-MSC (adipose-derived mesenchymal stem cells) as a vehicle for IFN-β delivery in experimental autoimmune encephalomyelitis. Clin. Immunol. 2016, 169, 98–106. [Google Scholar] [CrossRef]
- Liao, W.; Pham, V.; Liu, L.; Riazifar, M.; Pone, E.J.; Zhang, S.X.; Ma, F.; Lu, M.; Walsh, C.M.; Zhao, W. Mesenchymal stem cells engineered to express selectin ligands and IL-10 exert enhanced therapeutic efficacy in murine experimental autoimmune encephalomyelitis. Biomaterials 2016, 77, 87–97. [Google Scholar] [CrossRef]
- Talbott, E.O.; Malek, A.M.; Lacomis, D. The epidemiology of amyotrophic lateral sclerosis. Handb. Clin. Neurol. 2016, 138, 225–238. [Google Scholar] [PubMed]
- Comley, L.; Allodi, I.; Nichterwitz, S.; Nizzardo, M.; Simone, C.; Corti, S.; Hedlund, E. Motor neurons with differential vulnerability to degeneration show distinct protein signatures in health and ALS. Neuroscience 2015, 291, 216–229. [Google Scholar] [CrossRef] [PubMed]
- Farinazzo, A.; Turano, E.; Marconi, S.; Bistaffa, E.; Bazzoli, E.; Bonetti, B. Murine adipose-derived mesenchymal stromal cell vesicles: In vitro clues for neuroprotective and neuroregenerative approaches. Cytotherapy 2015, 17, 571–578. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Lee, M.; Ban, J.-J.; Kim, K.Y.; Jeon, G.S.; Im, W.; Sung, J.-J.; Kim, M. Adipose-derived stem cell exosomes alleviate pathology of amyotrophic lateral sclerosis in vitro. Biochem. Biophys. Res. Commun. 2016, 479, 434–439. [Google Scholar] [CrossRef]
- McCluskey, G.; Morrison, K.E.; Donaghy, C.; Rene, F.; Duddy, W.; Duguez, S. Extracellular Vesicles in Amyotrophic Lateral Sclerosis. Life 2023, 13, 121. [Google Scholar] [CrossRef]
- Parfejevs, V.; Sagini, K.; Buss, A.; Sobolevska, K.; Llorente, A.; Riekstina, U.; Abols, A. Adult Stem Cell-Derived Extracellular Vesicles in Cancer Treatment: Opportunities and Challenges. Cells 2020, 9, 1171. [Google Scholar] [CrossRef]
- Wang, K.; Li, Y.; Ren, C.; Wang, Y.; He, W.; Jiang, Y. Extracellular Vesicles as Innovative Treatment Strategy for Amyotrophic Lateral Sclerosis. Front. Cell Dev. Biol. 2021, 9, 754630. [Google Scholar] [CrossRef]
- Schulte, J.; Littleton, J.T. The biological function of the Huntingtin protein and its relevance to Huntington’s Disease pathology. Curr. Trends Neurol. 2011, 5, 65. [Google Scholar]
- Caron, N.S.; Dorsey, E.R.; Hayden, M.R. Therapeutic approaches to Huntington disease: From the bench to the clinic. Nat. Rev. Drug Discov. 2018, 17, 729–750. [Google Scholar] [CrossRef]
- Croese, T.; Furlan, R. Extracellular vesicles in neurodegenerative diseases. Mol. Asp. Med. 2018, 60, 52–61. [Google Scholar] [CrossRef]
- Lee, M.; Liu, T.; Im, W.; Kim, M. Exosomes from adipose-derived stem cells ameliorate phenotype of Huntington’s disease in vitro model. Eur. J. Neurosci. 2016, 44, 2114–2119. [Google Scholar] [CrossRef]
- Lee, S.-T.; Im, W.; Ban, J.-J.; Lee, M.; Jung, K.-H.; Lee, S.K.; Chu, K.; Kim, M. Exosome-based delivery of miR-124 in a Huntington’s disease model. J. Mov. Disord. 2017, 10, 45. [Google Scholar] [CrossRef] [PubMed]
- Yari, H.; Mikhailova, M.V.; Mardasi, M.; Jafarzadehgharehziaaddin, M.; Shahrokh, S.; Thangavelu, L.; Ahmadi, H.; Shomali, N.; Yaghoubi, Y.; Zamani, M.; et al. Emerging role of mesenchymal stromal cells (MSCs)-derived exosome in neurodegeneration-associated conditions: A groundbreaking cell-free approach. Stem Cell Res. Ther. 2022, 13, 423. [Google Scholar] [CrossRef] [PubMed]
- Ebrahimi, M.J.; Aliaghaei, A.; Boroujeni, M.E.; Khodagholi, F.; Meftahi, G.; Abdollahifar, M.A.; Ahmadi, H.; Danyali, S.; Daftari, M.; Sadeghi, Y. Human Umbilical Cord Matrix Stem Cells Reverse Oxidative Stress-Induced Cell Death and Ameliorate Motor Function and Striatal Atrophy in Rat Model of Huntington Disease. Neurotox. Res. 2018, 34, 273–284. [Google Scholar] [CrossRef] [PubMed]
- Didiot, M.C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R.; et al. Exosome-mediated Delivery of Hydrophobically Modified siRNA for Huntingtin mRNA Silencing. Mol. Ther. J. Am. Soc. Gene Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [PubMed]
- Kennedy, T.L.; Russell, A.J.; Riley, P. Experimental limitations of extracellular vesicle-based therapies for the treatment of myocardial infarction. Trends Cardiovasc. Med. 2021, 31, 405–415. [Google Scholar] [CrossRef]
- Lenzini, S. Establishing a working range for effective MSC-EV dose. RoosterBio. Blog. 2021. Available online: https://www.roosterbio.com/blog/establishing-a-working-range-for-effective-msc-ev-dose/ (accessed on 30 June 2023).
- Sun, Z.; Hou, X.; Zhang, J.; Li, J.; Wu, P.; Yan, L.; Qian, H. Diagnostic and Therapeutic Roles of Extracellular Vesicles in Aging-Related Diseases. Oxidative Med. Cell. Longev. 2022, 2022, 6742792. [Google Scholar] [CrossRef]
- Tian, C.-m.; Yang, M.-f.; Xu, H.-m.; Zhu, M.-z.; Zhang, Y.; Yao, J.; Wang, L.-s.; Liang, Y.-j.; Li, D.-f. Mesenchymal Stem Cell-derived Exosomes: Novel Therapeutic Approach for Inflammatory Bowel Diseases. Stem Cells Int. 2023, 2023, 4245704. [Google Scholar] [CrossRef]
- Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef] [PubMed]
- Sun, H.; Zhang, T.; Gao, J. Extracellular Vesicles Derived from Mesenchymal Stem Cells: A Potential Biodrug for Acute Respiratory Distress Syndrome Treatment. BioDrugs 2022, 36, 701–715. [Google Scholar] [CrossRef] [PubMed]
MSC-EV | Structure | Content | Origin | Applications |
---|---|---|---|---|
Exosomes | Small lipid bilayer vesicles (30–150 nm) | miRNAs, mRNAs, proteins, lipids, and signaling molecules | Luminal budding into MVBs; release by fusion of MVB with cell membrane | Therapeutic delivery, tissue regeneration, immunomodulation, and drug delivery |
Microvesicles | Larger vesicles (100–1000 nm) formed by outward budding and shedding from the cell membrane | Proteins, lipids, mRNA, miRNA, and DNA | Outward budding of cell membrane | Tissue repair, wound healing, and immunomodulation |
Apoptotic bodies | Large vesicles (50 nm–1 µm) released during apoptosis | Nucleic acids, histones, and fragmented organelles | Outward blebbing of apoptotic cell membrane | Immunomodulation, tissue regeneration, and biomarkers for cell death |
Disease | Type of EVs and Origin | Outcomes | Ref |
---|---|---|---|
AD | MSCs/exosomes | Enhances neurogenesis, reduces Aβ, and the restoration of cognitive function. | [164,165] |
PD | MSCs/exosome | Transferring of the miR-133b regulates neurite outgrowth. | [166] |
Improved neuronal function and oligodendrogenesis stimulation | [101] | ||
Reduction in α-syn aggregates | [167] | ||
MS | MSCs/exosomes MSCs/EVs | Drive peripheral resistance, activate apoptotic signaling pathway in self-reactive lymphocytes, and stimulate regulatory T cell differentiation by - IL-10 and TGF-β secretion - expression of PD-L1 and TGF-β | [168] |
MSCs/exosomes | Reduce CNS inflammation and demyelination by performing the following: - Shifting microglial polarization toward an M2 phenotype | [169,170] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Kandeel, M.; Morsy, M.A.; Alkhodair, K.M.; Alhojaily, S. Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules 2023, 13, 1250. https://doi.org/10.3390/biom13081250
Kandeel M, Morsy MA, Alkhodair KM, Alhojaily S. Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules. 2023; 13(8):1250. https://doi.org/10.3390/biom13081250
Chicago/Turabian StyleKandeel, Mahmoud, Mohamed A. Morsy, Khalid M. Alkhodair, and Sameer Alhojaily. 2023. "Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities" Biomolecules 13, no. 8: 1250. https://doi.org/10.3390/biom13081250
APA StyleKandeel, M., Morsy, M. A., Alkhodair, K. M., & Alhojaily, S. (2023). Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules, 13(8), 1250. https://doi.org/10.3390/biom13081250