Extracellular Vesicles as “Very Important Particles” (VIPs) in Aging
Abstract
:1. Extracellular Vesicles: From Cellular Debris to Essential Particles in Cellular Communication
- (1)
- Exosome (30–100 nm, the smallest extracellular vesicle) formation and release occur through the endosomal pathway and into the extracellular medium after fusion with the plasma membrane. Its content corresponds to that existing in the endosomal compartment [23].
- (2)
- Ectosomes (100–350 nm) are vesicles found everywhere in organisms and released from the plasma membrane. Their function is analogous to exosomes [24].
- (3)
- Microvesicles (MVs; formerly called microparticles or MPs) have a size from 100 nm–1 µm. They are secreted outside the cell by the process of evagination or sprouting of the plasma membrane, which involves: (a) relocation of phospholipids in the outer membrane so that the phosphatidylserine (PS), generally located on the inner side of the membrane, is exposed on the surface of the vesicle, (b) rearrangement of the cytoskeleton, (c) generation of the curvature of the membrane, and (d) liberation of the vesicle [25,26,27].
- (4)
- Apoptotic bodies (1–5 µm) are released as vesicles after cellular apoptosis, followed by increased membrane permeability, DNA fragmentation, and changes in mitochondrial membrane potential. Apoptotic bodies also expose PS on their surface and contain cellular organelles and genetic material [26,28,29].
2. Methods of EV Isolation and Characterization
3. Extracellular Vesicles as Biomarkers in Aging and Age-Associated Diseases
3.1. Types of Senescence
3.2. EVs as Biomarkers in Biological and Premature Diseases
3.3. EVs Cargoes, Concentrations, and Sizes in Aging and Age-Related Pathologies
3.4. EV Modulation by Environmental Factors
3.5. Limitations/Challenges of Using EVs as Biomarkers
4. Extracellular Vesicles as Therapeutic Agents in Aging and Age-Associated Diseases
4.1. Strategies Targeting Senescence
4.2. EV as Therapeutic Mediator in Biological and Premature Aging
5. Conclusions
6. Contribution to the Field Statement
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Yáñez-Mó, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carracedo, J.; Ramírez-Carracedo, R.; Alique, M.; Ramírez-Chamond, R. Endothelial Cell Senescence in the Pathogenesis of Endothelial Dysfunction; Intech: Singapore, 2018. [Google Scholar]
- Alique, M.; Ramírez-Carracedo, R.; Bodega, G.; Carracedo, J.; Ramírez, R. Senescent Microvesicles: A Novel Advance in Molecular Mechanisms of Atherosclerotic Calcification. Int. J. Mol. Sci. 2018, 19, 2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buzas, E.I.; György, B.; Nagy, G.; Falus, A.; Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev. Rheumatol. 2014, 10, 356–364. [Google Scholar] [CrossRef] [PubMed]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
- D’Anca, M.; Fenoglio, C.; Serpente, M.; Arosio, B.; Cesari, M.; Scarpini, E.A.; Galimberti, D. Exosome Determinants of Physiological Aging and Age-Related Neurodegenerative Diseases. Front. Aging Neurosci. 2019, 11, 232. [Google Scholar] [CrossRef] [Green Version]
- Yin, Y.; Chen, H.; Wang, Y.; Zhang, L.; Wang, X. Roles of extracellular vesicles in the aging microenvironment and age-related diseases. J. Extracell. Vesicles 2021, 10, e12154. [Google Scholar] [CrossRef]
- Greening, D.W.; Simpson, R.J. Understanding extracellular vesicle diversity—Current status. Expert Rev. Proteom. 2018, 15, 887–910. [Google Scholar] [CrossRef]
- Hartjes, T.A.; Mytnyk, S.; Jenster, G.W.; van Steijn, V.; van Royen, M.E. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering 2019, 6, 7. [Google Scholar] [CrossRef] [Green Version]
- Bonsergent, E.; Grisard, E.; Buchrieser, J.; Schwartz, O.; Théry, C.; Lavieu, G. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat. Commun. 2021, 12, 1864. [Google Scholar] [CrossRef]
- van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef]
- Chargaff, E.; West, R. The biological significance of the thromboplastic protein of blood. J. Biol. Chem. 1946, 166, 189–197. [Google Scholar] [CrossRef]
- Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef]
- Harding, C.; Heuser, J.; Stahl, P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: Demonstration of a pathway for receptor shedding. Eur. J. Cell Biol. 1984, 35, 256–263. [Google Scholar]
- Bazzan, E.; Tinè, M.; Casara, A.; Biondini, D.; Semenzato, U.; Cocconcelli, E.; Balestro, E.; Damin, M.; Radu, C.M.; 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]
- Stranford, D.M.; Leonard, J.N. Delivery of Biomolecules via Extracellular Vesicles: A Budding Therapeutic Strategy. Adv. Genet. 2017, 98, 155–175. [Google Scholar] [CrossRef]
- Couch, Y.; Buzas, E.I.; Di Vizio, D.; Gho, Y.S.; Harrison, P.; Hill, A.F.; Lotvall, J.; Raposo, G.; Stahl, P.D.; Thery, C.; et al. A brief history of nearly EV-erything—The rise and rise of extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12144. [Google Scholar] [CrossRef]
- Yoon, Y.J.; Kim, O.Y.; Gho, Y.S. Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 2014, 47, 531–539. [Google Scholar] [CrossRef]
- Monti, P.; Solazzo, G.; Ferrari, L.; Bollati, V. Extracellular Vesicles: Footprints of environmental exposures in the aging process? Curr. Environ. Health Rep. 2021, 8, 309–322. [Google Scholar] [CrossRef]
- 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]
- 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.; Goberdhan, D.C.; O’Driscoll, L.; Théry, C.; Welsh, J.A.; Blenkiron, C.; Buzás, E.I.; Di Vizio, D.; Erdbrügger, U.; Falcón-Pérez, J.M.; et al. Updating MISEV: Evolving the minimal requirements for studies of extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12182. [Google Scholar] [CrossRef] [PubMed]
- Bonjoch, L.; Gironella, M.; Iovanna, J.L.; Closa, D. REG3β modifies cell tumor function by impairing extracellular vesicle uptake. Sci. Rep. 2017, 7, 3143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef] [PubMed]
- Bodega, G.; Alique, M.; Puebla, L.; Carracedo, J.; Ramírez, R.M. Microvesicles: ROS scavengers and ROS producers. J. Extracell. Vesicles 2019, 8, 1626654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef] [Green Version]
- Xiong, J.; Miller, V.M.; Li, Y.; Jayachandran, M. Microvesicles at the crossroads between infection and cardiovascular diseases. J. Cardiovasc. Pharmacol. 2012, 59, 124–132. [Google Scholar] [CrossRef] [Green Version]
- Göran Ronquist, K. Extracellular vesicles and energy metabolism. Clin. Chim. Acta 2019, 488, 116–121. [Google Scholar] [CrossRef]
- Jayachandran, M.; Miller, V.M.; Heit, J.A.; Owen, W.G. Methodology for isolation, identification and characterization of microvesicles in peripheral blood. J. Immunol. Methods 2012, 375, 207–214. [Google Scholar] [CrossRef] [Green Version]
- Meehan, B.; Rak, J.; Di Vizio, D. Oncosomes—Large and small: What are they, where they came from? J. Extracell. Vesicles 2016, 5, 33109. [Google Scholar] [CrossRef]
- Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [Green Version]
- Verweij, F.J.; Balaj, L.; Boulanger, C.M.; Carter, D.R.F.; Compeer, E.B.; D’Angelo, G.; El Andaloussi, S.; Goetz, J.G.; Gross, J.C.; Hyenne, V.; et al. The power of imaging to understand extracellular vesicle biology in vivo. Nat. Methods 2021, 18, 1013–1026. [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] [Green Version]
- Zhang, Q.; Jeppesen, D.K.; Higginbotham, J.N.; Graves-Deal, R.; Trinh, V.Q.; Ramirez, M.A.; Sohn, Y.; Neininger, A.C.; Taneja, N.; McKinley, E.T.; et al. Supermeres are functional extracellular nanoparticles replete with disease biomarkers and therapeutic targets. Nat. Cell Biol. 2021, 23, 1240–1254. [Google Scholar] [CrossRef]
- Jeppesen, D.K.; Zhang, Q.; Franklin, J.L.; Coffey, R.J. Extracellular vesicles and nanoparticles: Emerging complexities. Trends Cell Biol. 2023. ahead of print. [Google Scholar] [CrossRef]
- Clancy, J.W.; Boomgarden, A.C.; D’Souza-Schorey, C. Profiling and promise of supermeres. Nat. Cell Biol. 2021, 23, 1217–1219. [Google Scholar] [CrossRef]
- Anand, S.; Samuel, M.; Mathivanan, S. Exomeres: A New Member of Extracellular Vesicles Family. Subcell. Biochem. 2021, 97, 89–97. [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] [Green Version]
- Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef] [Green Version]
- Torres-Bautista, A.; Torres-Acosta, M.A.; González-Valdez, J. Characterization and optimization of polymer-polymer aqueous two-phase systems for the isolation and purification of CaCo2 cell-derived exosomes. PLoS ONE 2022, 17, e0273243. [Google Scholar] [CrossRef]
- Macías, M.; Rebmann, V.; Mateos, B.; Varo, N.; Perez-Gracia, J.L.; Alegre, E.; González, Á. Comparison of six commercial serum exosome isolation methods suitable for clinical laboratories. Effect in cytokine analysis. Clin. Chem. Lab. Med. 2019, 57, 1539–1545. [Google Scholar] [CrossRef]
- Kanwar, S.S.; Dunlay, C.J.; Simeone, D.M.; Nagrath, S. Microfluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomes. Lab. Chip 2014, 14, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
- Poupardin, R.; Wolf, M.; Strunk, D. Adherence to minimal experimental requirements for defining extracellular vesicles and their functions. Adv. Drug Deliv. Rev. 2021, 176, 113872. [Google Scholar] [CrossRef] [PubMed]
- Schaum, N.; Lehallier, B.; Hahn, O.; Pálovics, R.; Hosseinzadeh, S.; Lee, S.E.; Sit, R.; Lee, D.P.; Losada, P.M.; Zardeneta, M.E.; et al. Ageing hallmarks exhibit organ-specific temporal signatures. Nature 2020, 583, 596–602. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
- Thane, K.E.; Davis, A.M.; Hoffman, A.M. Improved methods for fluorescent labeling and detection of single extracellular vesicles using nanoparticle tracking analysis. Sci. Rep. 2019, 9, 12295. [Google Scholar] [CrossRef] [Green Version]
- Bağcı, C.; Sever-Bahcekapili, M.; Belder, N.; Bennett, A.P.S.; Erdener, Ş.; Dalkara, T. Overview of extracellular vesicle characterization techniques and introduction to combined reflectance and fluorescence confocal microscopy to distinguish extracellular vesicle subpopulations. Neurophotonics 2022, 9, 021903. [Google Scholar] [CrossRef]
- Robbins, P.D. Extracellular vesicles and aging. Stem Cell Investig. 2017, 4, 98. [Google Scholar] [CrossRef] [Green Version]
- Breitwieser, K.; Koch, L.F.; Tertel, T.; Proestler, E.; Burgers, L.D.; Lipps, C.; Adjaye, J.; Fürst, R.; Giebel, B.; Saul, M.J. Detailed Characterization of Small Extracellular Vesicles from Different Cell Types Based on Tetraspanin Composition by ExoView R100 Platform. Int. J. Mol. Sci. 2022, 23, 8544. [Google Scholar] [CrossRef]
- Melling, G.E.; Conlon, R.; Pantazi, P.; Dellar, E.R.; Samuel, P.; Baena-Lopez, L.A.; Simpson, J.C.; Carter, D.R.F. Confocal microscopy analysis reveals that only a small proportion of extracellular vesicles are successfully labelled with commonly utilised staining methods. Sci. Rep. 2022, 12, 262. [Google Scholar] [CrossRef]
- Silva, A.M.; Lázaro-Ibáñez, E.; Gunnarsson, A.; Dhande, A.; Daaboul, G.; Peacock, B.; Osteikoetxea, X.; Salmond, N.; Friis, K.P.; Shatnyeva, O.; et al. Quantification of protein cargo loading into engineered extracellular vesicles at single-vesicle and single-molecule resolution. J. Extracell. Vesicles 2021, 10, e12130. [Google Scholar] [CrossRef]
- Rejuvenome Project. Available online: https://astera.org/rejuvenome/ (accessed on 14 February 2023).
- Fafián-Labora, J.; Lesende-Rodriguez, I.; Fernández-Pernas, P.; Sangiao-Alvarellos, S.; Monserrat, L.; Arntz, O.J.; van de Loo, F.J.; Mateos, J.; Arufe, M.C. Effect of age on pro-inflammatory miRNAs contained in mesenchymal stem cell-derived extracellular vesicles. Sci. Rep. 2017, 7, 43923. [Google Scholar] [CrossRef] [Green Version]
- Bulterijs, S.; Hull, R.S.; Björk, V.C.; Roy, A.G. It is time to classify biological aging as a disease. Front. Genet. 2015, 6, 205. [Google Scholar] [CrossRef] [Green Version]
- Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef] [Green Version]
- Mas-Bargues, C.; Alique, M.; Barrús-Ortiz, M.T.; Borrás, C.; Rodrigues-Díez, R. Exploring New Kingdoms: The Role of Extracellular Vesicles in Oxi-Inflamm-Aging Related to Cardiorenal Syndrome. Antioxidants 2021, 11, 78. [Google Scholar] [CrossRef]
- Hayflick, L. The cell biology of aging. J. Investig. Derm. 1979, 73, 8–14. [Google Scholar] [CrossRef] [Green Version]
- Shay, J.W.; Wright, W.E. Hayflick, his limit, and cellular ageing. Nat. Rev. Mol. Cell Biol. 2000, 1, 72–76. [Google Scholar] [CrossRef]
- Vulliamy, T.; Marrone, A.; Goldman, F.; Dearlove, A.; Bessler, M.; Mason, P.J.; Dokal, I. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 2001, 413, 432–435. [Google Scholar] [CrossRef]
- Goligorsky, M.S.; Chen, J.; Patschan, S. Stress-induced premature senescence of endothelial cells: A perilous state between recovery and point of no return. Curr. Opin. Hematol. 2009, 16, 215–219. [Google Scholar] [CrossRef]
- Schosserer, M.; Grillari, J.; Breitenbach, M. The Dual Role of Cellular Senescence in Developing Tumors and Their Response to Cancer Therapy. Front. Oncol. 2017, 7, 278. [Google Scholar] [CrossRef] [Green Version]
- Nyunoya, T.; Monick, M.M.; Klingelhutz, A.; Yarovinsky, T.O.; Cagley, J.R.; Hunninghake, G.W. Cigarette smoke induces cellular senescence. Am. J. Respir. Cell Mol. Biol. 2006, 35, 681–688. [Google Scholar] [CrossRef] [Green Version]
- Mijit, M.; Caracciolo, V.; Melillo, A.; Amicarelli, F.; Giordano, A. Role of p53 in the Regulation of Cellular Senescence. Biomolecules 2020, 10, 420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Admasu, T.D.; Rae, M.; Stolzing, A. Dissecting primary and secondary senescence to enable new senotherapeutic strategies. Ageing Res. Rev. 2021, 70, 101412. [Google Scholar] [CrossRef] [PubMed]
- Beauséjour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of human cellular senescence: Roles of the p53 and p16 pathways. EMBO J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef] [PubMed]
- Gasek, N.S.; Kuchel, G.A.; Kirkland, J.L.; Xu, M. Strategies for Targeting Senescent Cells in Human Disease. Nat. Aging 2021, 1, 870–879. [Google Scholar] [CrossRef] [PubMed]
- Borgoni, S.; Kudryashova, K.S.; Burka, K.; de Magalhães, J.P. Targeting immune dysfunction in aging. Ageing Res. Rev. 2021, 70, 101410. [Google Scholar] [CrossRef]
- van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [Green Version]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [Green Version]
- Narasimhan, A.; Flores, R.R.; Robbins, P.D.; Niedernhofer, L.J. Role of Cellular Senescence in Type II Diabetes. Endocrinology 2021, 162, bqab136. [Google Scholar] [CrossRef]
- Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
- Yousefzadeh, M.J.; Zhao, J.; Bukata, C.; Wade, E.A.; McGowan, S.J.; Angelini, L.A.; Bank, M.P.; Gurkar, A.U.; McGuckian, C.A.; Calubag, M.F.; et al. Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice. Aging Cell 2020, 19, e13094. [Google Scholar] [CrossRef]
- Yousefzadeh, M.J.; Flores, R.R.; Zhu, Y.; Schmiechen, Z.C.; Brooks, R.W.; Trussoni, C.E.; Cui, Y.; Angelini, L.; Lee, K.A.; McGowan, S.J.; et al. An aged immune system drives senescence and ageing of solid organs. Nature 2021, 594, 100–105. [Google Scholar] [CrossRef]
- Leite, G.; Pimentel, M.; Barlow, G.M.; Chang, C.; Hosseini, A.; Wang, J.; Parodi, G.; Sedighi, R.; Rezaie, A.; Mathur, R. Age and the aging process significantly alter the small bowel microbiome. Cell Rep. 2021, 36, 109765. [Google Scholar] [CrossRef]
- Chung, H.Y.; Kim, D.H.; Lee, E.K.; Chung, K.W.; Chung, S.; Lee, B.; Seo, A.Y.; Chung, J.H.; Jung, Y.S.; Im, E.; et al. Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis. 2019, 10, 367–382. [Google Scholar] [CrossRef] [Green Version]
- Saheera, S.; Potnuri, A.G.; Krishnamurthy, P. Nano-Vesicle (Mis)Communication in Senescence-Related Pathologies. Cells 2020, 9, 1974. [Google Scholar] [CrossRef]
- Roig-Carles, D.; Willms, E.; Fontijn, R.D.; Martinez-Pacheco, S.; Mäger, I.; de Vries, H.E.; Hirst, M.; Sharrack, B.; Male, D.K.; Hawkes, C.A.; et al. Endothelial-Derived Extracellular Vesicles Induce Cerebrovascular Dysfunction in Inflammation. Pharmaceutics 2021, 13, 1525. [Google Scholar] [CrossRef]
- Ovadya, Y.; Landsberger, T.; Leins, H.; Vadai, E.; Gal, H.; Biran, A.; Yosef, R.; Sagiv, A.; Agrawal, A.; Shapira, A.; et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat. Commun. 2018, 9, 5435. [Google Scholar] [CrossRef] [Green Version]
- Boccardi, V.; Mecocci, P. The Importance of Cellular Senescence in Frailty and Cardiovascular Diseases. Adv. Exp. Med. Biol. 2020, 1216, 79–86. [Google Scholar] [CrossRef]
- Kadota, T.; Fujita, Y.; Yoshioka, Y.; Araya, J.; Kuwano, K.; Ochiya, T. Emerging role of extracellular vesicles as a senescence-associated secretory phenotype: Insights into the pathophysiology of lung diseases. Mol. Asp. Med. 2018, 60, 92–103. [Google Scholar] [CrossRef]
- Xu, J.; Chen, Y.; Yu, D.; Zhang, L.; Dou, X.; Wu, G.; Wang, Y.; Zhang, S. Evaluation of the cargo contents and potential role of extracellular vesicles in osteoporosis. Aging 2021, 13, 19282–19292. [Google Scholar] [CrossRef]
- Alibhai, F.J.; Lim, F.; Yeganeh, A.; DiStefano, P.V.; Binesh-Marvasti, T.; Belfiore, A.; Wlodarek, L.; Gustafson, D.; Millar, S.; Li, S.H.; et al. Cellular senescence contributes to age-dependent changes in circulating extracellular vesicle cargo and function. Aging Cell 2020, 19, e13103. [Google Scholar] [CrossRef] [Green Version]
- Tominaga, K. The emerging role of senescent cells in tissue homeostasis and pathophysiology. Pathobiol. Aging Age Relat. Dis. 2015, 5, 27743. [Google Scholar] [CrossRef] [PubMed]
- Gabrielli, M.; Prada, I.; Joshi, P.; Falcicchia, C.; D’Arrigo, G.; Rutigliano, G.; Battocchio, E.; Zenatelli, R.; Tozzi, F.; Radeghieri, A.; et al. Microglial large extracellular vesicles propagate early synaptic dysfunction in Alzheimer’s disease. Brain 2022, 145, 2849–2868. [Google Scholar] [CrossRef] [PubMed]
- Głuchowska, A.; Cysewski, D.; Baj-Krzyworzeka, M.; Szatanek, R.; Węglarczyk, K.; Podszywałow-Bartnicka, P.; Sunderland, P.; Kozłowska, E.; Śliwińska, M.A.; Dąbrowski, M.; et al. Unbiased proteomic analysis of extracellular vesicles secreted by senescent human vascular smooth muscle cells reveals their ability to modulate immune cell functions. Geroscience 2022, 44, 2863–2884. [Google Scholar] [CrossRef] [PubMed]
- Wu, S.F.; Noren Hooten, N.; Freeman, D.W.; Mode, N.A.; Zonderman, A.B.; Evans, M.K. Extracellular vesicles in diabetes mellitus induce alterations in endothelial cell morphology and migration. J. Transl. Med. 2020, 18, 230. [Google Scholar] [CrossRef] [PubMed]
- Cordazzo, C.; Petrini, S.; Neri, T.; Lombardi, S.; Carmazzi, Y.; Pedrinelli, R.; Paggiaro, P.; Celi, A. Rapid shedding of proinflammatory microparticles by human mononuclear cells exposed to cigarette smoke is dependent on Ca2+ mobilization. Inflamm. Res. 2014, 63, 539–547. [Google Scholar] [CrossRef]
- Yuan, D.; Luo, J.; Sun, Y.; Hao, L.; Zheng, J.; Yang, Z. PCOS follicular fluid derived exosomal miR-424-5p induces granulosa cells senescence by targeting CDCA4 expression. Cell Signal. 2021, 85, 110030. [Google Scholar] [CrossRef]
- Chanda, D.; Otoupalova, E.; Hough, K.P.; Locy, M.L.; Bernard, K.; Deshane, J.S.; Sanderson, R.D.; Mobley, J.A.; Thannickal, V.J. Fibronectin on the Surface of Extracellular Vesicles Mediates Fibroblast Invasion. Am. J. Respir. Cell Mol. Biol 2019, 60, 279–288. [Google Scholar] [CrossRef]
- Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef] [Green Version]
- Alique, M.; Bodega, G.; Corchete, E.; García-Menéndez, E.; de Sequera, P.; Luque, R.; Rodríguez-Padrón, D.; Marqués, M.; Portolés, J.; Carracedo, J.; et al. Microvesicles from indoxyl sulfate-treated endothelial cells induce vascular calcification. Comput. Struct. Biotechnol. J. 2020, 18, 953–966. [Google Scholar] [CrossRef]
- Neven, K.Y.; Nawrot, T.S.; Bollati, V. Extracellular Vesicles: How the External and Internal Environment Can Shape Cell-To-Cell Communication. Curr. Environ. Health Rep. 2017, 4, 30–37. [Google Scholar] [CrossRef]
- Fujita, Y.; Araya, J.; Ochiya, T. Extracellular vesicles in smoking-related lung diseases. Oncotarget 2015, 6, 43144–43145. [Google Scholar] [CrossRef]
- Momen-Heravi, F.; Saha, B.; Kodys, K.; Catalano, D.; Satishchandran, A.; Szabo, G. Increased number of circulating exosomes and their microRNA cargos are potential novel biomarkers in alcoholic hepatitis. J. Transl. Med. 2015, 13, 261. [Google Scholar] [CrossRef] [Green Version]
- Buzas, E.I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 2022, 2022, 1–15. [Google Scholar] [CrossRef]
- Sanz-Ros, J.; Mas-Bargues, C.; Romero-Garcia, N.; Huete-Acevedo, J.; Dromant, M.; Borras, C. Therapeutic Potential of Extracellular Vesicles in Aging and Age-Related Diseases. Int. J. Mol. Sci. 2022, 23, 14632. [Google Scholar] [CrossRef]
- Ramirez, M.I.; Amorim, M.G.; Gadelha, C.; Milic, I.; Welsh, J.A.; Freitas, V.M.; Nawaz, M.; Akbar, N.; Couch, Y.; Makin, L.; et al. Technical challenges of working with extracellular vesicles. Nanoscale 2018, 10, 881–906. [Google Scholar] [CrossRef] [Green Version]
- Soekmadji, C.; Li, B.; Huang, Y.; Wang, H.; An, T.; Liu, C.; Pan, W.; Chen, J.; Cheung, L.; Falcon-Perez, J.M.; et al. The future of Extracellular Vesicles as Theranostics—An ISEV meeting report. J. Extracell. Vesicles 2020, 9, 1809766. [Google Scholar] [CrossRef]
- Bæk, R.; Varming, K.; Jørgensen, M.M. Does smoking, age or gender affect the protein phenotype of extracellular vesicles in plasma? Transfus. Apher. Sci. 2016, 55, 44–52. [Google Scholar] [CrossRef] [Green Version]
- Hamdan, Y.; Mazini, L.; Malka, G. Exosomes and Micro-RNAs in Aging Process. Biomedicines 2021, 9, 968. [Google Scholar] [CrossRef]
- O’Loghlen, A. Role for extracellular vesicles in the tumour microenvironment. Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160488. [Google Scholar] [CrossRef]
- Bodega, G.; Alique, M.; Bohórquez, L.; Ciordia, S.; Mena, M.C.; Ramírez, M.R. The Antioxidant Machinery of Young and Senescent Human Umbilical Vein Endothelial Cells and Their Microvesicles. Oxidative Med. Cell. Longev. 2017, 2017, 7094781. [Google Scholar] [CrossRef] [Green Version]
- Bodega, G.; Alique, M.; Bohórquez, L.; Morán, M.; Magro, L.; Puebla, L.; Ciordia, S.; Mena, M.C.; Arza, E.; Ramírez, M.R. Young and Especially Senescent Endothelial Microvesicles Produce NADPH: The Fuel for Their Antioxidant Machinery. Oxidative Med. Cell. Longev. 2018, 2018, 3183794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sung, D.K.; Chang, Y.S.; Sung, S.I.; Ahn, S.Y.; Park, W.S. Thrombin Preconditioning of Extracellular Vesicles Derived from Mesenchymal Stem Cells Accelerates Cutaneous Wound Healing by Boosting Their Biogenesis and Enriching Cargo Content. J. Clin. Med. 2019, 8, 533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiley, C.D.; Campisi, J. The metabolic roots of senescence: Mechanisms and opportunities for intervention. Nat. Metab. 2021, 3, 1290–1301. [Google Scholar] [CrossRef] [PubMed]
- Poblocka, M.; Bassey, A.L.; Smith, V.M.; Falcicchio, M.; Manso, A.S.; Althubiti, M.; Sheng, X.; Kyle, A.; Barber, R.; Frigerio, M.; et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci. Rep. 2021, 11, 20358. [Google Scholar] [CrossRef] [PubMed]
- Johmura, Y.; Yamanaka, T.; Omori, S.; Wang, T.W.; Sugiura, Y.; Matsumoto, M.; Suzuki, N.; Kumamoto, S.; Yamaguchi, K.; Hatakeyama, S.; et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021, 371, 265–270. [Google Scholar] [CrossRef]
- Myrianthopoulos, V.; Evangelou, K.; Vasileiou, P.V.S.; Cooks, T.; Vassilakopoulos, T.P.; Pangalis, G.A.; Kouloukoussa, M.; Kittas, C.; Georgakilas, A.G.; Gorgoulis, V.G. Senescence and senotherapeutics: A new field in cancer therapy. Pharmacol. Ther. 2019, 193, 31–49. [Google Scholar] [CrossRef]
- Ding, Y.N.; Wang, H.Y.; Chen, H.Z.; Liu, D.P. Targeting senescent cells for vascular aging and related diseases. J. Mol. Cell. Cardiol. 2022, 162, 43–52. [Google Scholar] [CrossRef]
- Raffaele, M.; Vinciguerra, M. The costs and benefits of senotherapeutics for human health. Lancet Healthy Longev. 2022, 3, e67–e77. [Google Scholar] [CrossRef]
- Storci, G.; De Carolis, S.; Papi, A.; Bacalini, M.G.; Gensous, N.; Marasco, E.; Tesei, A.; Fabbri, F.; Arienti, C.; Zanoni, M.; et al. Genomic stability, anti-inflammatory phenotype, and up-regulation of the RNAseH2 in cells from centenarians. Cell Death Differ. 2019, 26, 1845–1858. [Google Scholar] [CrossRef] [Green Version]
- Ha, D.H.; Kim, H.K.; Lee, J.; Kwon, H.H.; Park, G.H.; Yang, S.H.; Jung, J.Y.; Choi, H.; Lee, J.H.; Sung, S.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020, 9, 1157. [Google Scholar] [CrossRef]
- He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Xie, X.; Lian, W.; Shi, R.; Han, S.; Zhang, H.; Lu, L.; Li, M. Exosomes from adipose-derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Svolacchia, F.; De Francesco, F.; Trovato, L.; Graziano, A.; Ferraro, G.A. An innovative regenerative treatment of scars with dermal micrografts. J. Cosmet. Derm. 2016, 15, 245–253. [Google Scholar] [CrossRef]
- Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef] [Green Version]
- Deryabin, P.I.; Shatrova, A.N.; Borodkina, A.V. Apoptosis resistance of senescent cells is an intrinsic barrier for senolysis induced by cardiac glycosides. Cell. Mol. Life Sci. 2021, 78, 7757–7776. [Google Scholar] [CrossRef]
- Mato-Basalo, R.; Morente-López, M.; Arntz, O.J.; van de Loo, F.A.J.; Fafián-Labora, J.; Arufe, M.C. Therapeutic Potential for Regulation of the Nuclear Factor Kappa-B Transcription Factor p65 to Prevent Cellular Senescence and Activation of Pro-Inflammatory in Mesenchymal Stem Cells. Int. J. Mol. Sci. 2021, 22, 3367. [Google Scholar] [CrossRef]
- Gorgun, C.; Palamà, M.E.F.; Reverberi, D.; Gagliani, M.C.; Cortese, K.; Tasso, R.; Gentili, C. Role of extracellular vesicles from adipose tissue- and bone marrow-mesenchymal stromal cells in endothelial proliferation and chondrogenesis. Stem Cells Transl. Med. 2021, 10, 1680–1695. [Google Scholar] [CrossRef]
- Tejeda-Mora, H.; Leon, L.G.; Demmers, J.; Baan, C.C.; Reinders, M.E.J.; Bleck, B.; Lombardo, E.; Merino, A.; Hoogduijn, M.J. Proteomic Analysis of Mesenchymal Stromal Cell-Derived Extracellular Vesicles and Reconstructed Membrane Particles. Int. J. Mol. Sci. 2021, 22, 12935. [Google Scholar] [CrossRef]
- Van Hoecke, L.; Van Cauwenberghe, C.; Börger, V.; Bruggeman, A.; Castelein, J.; Van Imschoot, G.; Van Wonterghem, E.; Dittrich, R.; Claeys, W.; Xie, J.; et al. Anti-Inflammatory Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Pathology in Niemann-Pick Type C Disease. Biomedicines 2021, 9, 1864. [Google Scholar] [CrossRef]
- Zhang, X.; Baht, G.S.; Huang, R.; Chen, Y.H.; Molitoris, K.H.; Miller, S.E.; Kraus, V.B. Rejuvenation of neutrophils and their extracellular vesicles is associated with enhanced aged fracture healing. Aging Cell 2022, 21, e13651. [Google Scholar] [CrossRef]
- Bi, Y.; Qiao, X.; Liu, Q.; Song, S.; Zhu, K.; Qiu, X.; Zhang, X.; Jia, C.; Wang, H.; Yang, Z.; et al. Systemic proteomics and miRNA profile analysis of exosomes derived from human pluripotent stem cells. Stem Cell Res. 2022, 13, 449. [Google Scholar] [CrossRef] [PubMed]
- Liao, C.M.; Luo, T.; von der Ohe, J.; de Juan Mora, B.; Schmitt, R.; Hass, R. Human MSC-Derived Exosomes Reduce Cellular Senescence in Renal Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 13562. [Google Scholar] [CrossRef] [PubMed]
- Tan, T.T.; Toh, W.S.; Lai, R.C.; Lim, S.K. Practical considerations in transforming MSC therapy for neurological diseases from cell to EV. Exp. Neurol. 2022, 349, 113953. [Google Scholar] [CrossRef] [PubMed]
- Warnecke, A.; Prenzler, N.; Harre, J.; Köhl, U.; Gärtner, L.; Lenarz, T.; Laner-Plamberger, S.; Wietzorrek, G.; Staecker, H.; Lassacher, T.; et al. First-in-human intracochlear application of human stromal cell-derived extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12094. [Google Scholar] [CrossRef]
- Jeon, O.H.; Wilson, D.R.; Clement, C.C.; Rathod, S.; Cherry, C.; Powell, B.; Lee, Z.; Khalil, A.M.; Green, J.J.; Campisi, J.; et al. Senescence cell-associated extracellular vesicles serve as osteoarthritis disease and therapeutic markers. JCI Insight 2019, 4, e125019. [Google Scholar] [CrossRef] [Green Version]
Method | Quantification | Characterization | Advantages | Disadvantages | References |
---|---|---|---|---|---|
Flow cytometry (FC) or flow cytometry experiment on extracellular vesicles (MIFlowCyt-EV) | Yes | Yes (specific surface cell origin marker) |
|
| [2,39,44] |
Nanoparticle tracking analysis (NTA) | Yes | Yes |
|
| [2,38,39,45,46,47] |
Western blot (WB) | No | Yes (example: CD63 protein = exosome markers) |
|
| [39] |
Transmission electron microscopy (TEM) | No | Yes (size and morphology) |
|
| [38,39] |
Dynamic light scattering (DLS) | No | Yes (range distribution) |
|
| [2,38] |
Tunable resistive pulse sensing (tRPS) | Yes | No |
|
| [2,38,45] |
High-resolution flow cytometry (hFC) | Yes | Yes |
|
| [38,45,48] |
ExoView | Yes | Yes |
|
| [49,50,51] |
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
Mas-Bargues, C.; Alique, M. Extracellular Vesicles as “Very Important Particles” (VIPs) in Aging. Int. J. Mol. Sci. 2023, 24, 4250. https://doi.org/10.3390/ijms24044250
Mas-Bargues C, Alique M. Extracellular Vesicles as “Very Important Particles” (VIPs) in Aging. International Journal of Molecular Sciences. 2023; 24(4):4250. https://doi.org/10.3390/ijms24044250
Chicago/Turabian StyleMas-Bargues, Cristina, and Matilde Alique. 2023. "Extracellular Vesicles as “Very Important Particles” (VIPs) in Aging" International Journal of Molecular Sciences 24, no. 4: 4250. https://doi.org/10.3390/ijms24044250
APA StyleMas-Bargues, C., & Alique, M. (2023). Extracellular Vesicles as “Very Important Particles” (VIPs) in Aging. International Journal of Molecular Sciences, 24(4), 4250. https://doi.org/10.3390/ijms24044250