Role of Extracellular Vesicles in Cell Death and Inflammation
Abstract
:1. Extracellular Vesicles: Introduction, Subtypes, and Cargo
2. EVs as Regulators of Cell Death
3. Role of EVs in Attenuating Cell Death
4. Role of EVs in Promoting Cell Death
5. EVs in Inflammation
6. EVs in Sepsis Associated Inflammation
7. EVs in Lung Inflammatory Disorders
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ALI | Acute lung injury |
ApoBDs | Apoptotic bodies |
ApoEVs | Apoptotic EVs |
ApoExos | Apoptotic exosomes |
ApoMVs | Apoptotic microvesicles |
ARDS | Acute respiratory distress syndrome |
BALF | Bronchoalveolar lavage fluid |
CD | Cluster of differentiation |
CLP | Cecal ligation and puncture |
COPD | Chronic obstructive pulmonary disorder |
EVs | Extracellular vesicles |
HSP HIV | Heat shock protein Human immunodeficiency virus |
IECs | Intestinal epithelial cells |
LPS | Lipopolysaccharide |
MEVs | Milk-derived EVs |
MHC | Major histocompatibility complex |
miR | micro-RNA |
MSCs | Mesenchymal stem cells |
MVB | Multivesicular bodies |
MVs | Microvesicles |
PAMPs | Pathogen associated molecular patters |
TNF | Tumor necrosis factor |
References
- Kalra, H.; Drummen, G.P.C.; Mathivanan, S. Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int. J. Mol. Sci. 2016, 17, 170. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Pan, B.T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Biol. 1985, 101, 942–948. [Google Scholar] [CrossRef] [Green Version]
- Gangoda, L.; Boukouris, S.; Liem, M.; Kalra, H.; Mathivanan, S. Extracellular vesicles including exosomes are mediators of signal transduction: Are they protective or pathogenic? Proteomics 2015, 15, 260–271. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef] [PubMed]
- Howitt, J.; Hill, A.F. Exosomes in the Pathology of Neurodegenerative Diseases. J. Biol. Chem. 2016, 291, 26589–26597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samuel, M.; Fonseka, P.; Sanwlani, R.; Gangoda, L.; Chee, S.H.; Keerthikumar, S.; Spurling, A.; Chitti, S.V.; Zanker, D.; Ang, C.-S.; et al. Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer metastasis. Nat. Commun. 2021, 12, 3950. [Google Scholar] [CrossRef]
- Sanwlani, R.; Fonseka, P.; Chitti, S.V.; Mathivanan, S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes 2020, 8, 11. [Google Scholar] [CrossRef]
- Sanwlani, R.; Fonseka, P.; Mathivanan, S. Are Dietary Extracellular Vesicles Bioavailable and Functional in Consuming Organisms? Subcell. Biochem. 2021, 97, 509–521. [Google Scholar] [CrossRef]
- Kang, T.; Atukorala, I.; Mathivanan, S. Biogenesis of Extracellular Vesicles. Subcell. Biochem. 2021, 97, 19–43. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
- Keerthikumar, S.; Gangoda, L.; Liem, M.; Fonseka, P.; Atukorala, I.; Ozcitti, C.; Mechler, A.; Adda, C.G.; Ang, C.S.; Mathivanan, S. Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes. Oncotarget 2015, 6, 15375–15396. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef] [PubMed]
- Anand, S.; Samuel, M.; Mathivanan, S. Exomeres: A New Member of Extracellular Vesicles Family. In New Frontiers: Extracellular Vesicles; Mathivanan, S., Fonseka, P., Nedeva, C., Atukorala, I., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 89–97. [Google Scholar]
- György, B.; Szabó, T.G.; Pásztói, M.; Pál, Z.; Misják, P.; Aradi, B.; László, V.; Pállinger, E.; Pap, E.; Kittel, A.; et al. Membrane vesicles, current state-of-the-art: Emerging role of extracellular vesicles. Cell. Mol. Life Sci. CMLS 2011, 68, 2667–2688. [Google Scholar] [CrossRef] [Green Version]
- Minciacchi, V.R.; You, S.; Spinelli, C.; Morley, S.; Zandian, M.; Aspuria, P.-J.; Cavallini, L.; Ciardiello, C.; Reis Sobreiro, M.; Morello, M.; et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget 2015, 6, 11327–11341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zijlstra, A.; Di Vizio, D. Size matters in nanoscale communication. Nat. Cell Biol. 2018, 20, 228–230. [Google Scholar] [CrossRef]
- Tavano, S.; Heisenberg, C.-P. Migrasomes take center stage. Nat. Cell Biol. 2019, 21, 918–920. [Google Scholar] [CrossRef]
- 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.H. 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] [PubMed]
- Kerr, J.F.R.; Wyllie, A.H.; Currie, A.R. Apoptosis: A Basic Biological Phenomenon with Wideranging Implications in Tissue Kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [Green Version]
- Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.-J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schiller, M.; Parcina, M.; Heyder, P.; Foermer, S.; Ostrop, J.; Leo, A.; Heeg, K.; Herrmann, M.; Lorenz, H.-M.; Bekeredjian-Ding, I. Induction of Type I IFN Is a Physiological Immune Reaction to Apoptotic Cell-Derived Membrane Microparticles. J. Immunol. 2012, 189, 1747–1756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tucher, C.; Bode, K.; Schiller, P.; Claßen, L.; Birr, C.; Souto-Carneiro, M.M.; Blank, N.; Lorenz, H.-M.; Schiller, M. Extracellular Vesicle Subtypes Released from Activated or Apoptotic T-Lymphocytes Carry a Specific and Stimulus-Dependent Protein Cargo. Front. Immunol. 2018, 9, 534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sirois, I.; Raymond, M.A.; Brassard, N.; Cailhier, J.F.; Fedjaev, M.; Hamelin, K.; Londono, I.; Bendayan, M.; Pshezhetsky, A.V.; Hébert, M.J. Caspase-3-dependent export of TCTP: A novel pathway for antiapoptotic intercellular communication. Cell Death Differ. 2011, 18, 549–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sirois, I.; Groleau, J.; Pallet, N.; Brassard, N.; Hamelin, K.; Londono, I.; Pshezhetsky, A.V.; Bendayan, M.; Hébert, M.J. Caspase activation regulates the extracellular export of autophagic vacuoles. Autophagy 2012, 8, 927–937. [Google Scholar] [CrossRef] [Green Version]
- Pathan, M.; Fonseka, P.; Chitti, S.V.; Kang, T.; Sanwlani, R.; Van Deun, J.; Hendrix, A.; Mathivanan, S. Vesiclepedia 2019: A compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2019, 47, D516–D519. [Google Scholar] [CrossRef] [Green Version]
- Mori, Y.; Koike, M.; Moriishi, E.; Kawabata, A.; Tang, H.; Oyaizu, H.; Uchiyama, Y.; Yamanishi, K. Human Herpesvirus-6 Induces MVB Formation, and Virus Egress Occurs by an Exosomal Release Pathway. Traffic 2008, 9, 1728–1742. [Google Scholar] [CrossRef] [Green Version]
- Kalra, H.; Gangoda, L.; Fonseka, P.; Chitti, S.V.; Liem, M.; Keerthikumar, S.; Samuel, M.; Boukouris, S.; Al Saffar, H.; Collins, C.; et al. Extracellular vesicles containing oncogenic mutant β-catenin activate Wnt signalling pathway in the recipient cells. J. Extracell. Vesicles 2019, 8, 1690217. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Boukouris, S.; Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics. Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef] [Green Version]
- Mathivanan, S.; Lim, J.W.E.; Tauro, B.J.; Ji, H.; Moritz, R.L.; Simpson, R.J. Proteomics Analysis of A33 Immunoaffinity-purified Exosomes Released from the Human Colon Tumor Cell Line LIM1215 Reveals a Tissue-specific Protein Signature. Mol. Cell. Proteom. 2010, 9, 197–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Srivastava, P. Interaction of Heat Shock Proteins with Peptides and Antigen Presenting Cells: Chaperoning of the Innate and Adaptive Immune Responses. Annu. Rev. Immunol. 2002, 20, 395–425. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. Rev. Bras. Pesqui. Med. E Biol. 2013, 46, 824–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poon, I.K.; Lucas, C.D.; Rossi, A.G.; Ravichandran, K.S. Apoptotic cell clearance: Basic biology and therapeutic potential. Nat. Rev. Immunol. 2014, 14, 166–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, S.J.; Kim, J.M.; Kim, J.; Hur, J.; Park, S.; Kim, K.; Shin, H.J.; Chwae, Y.J. Molecular mechanisms of biogenesis of apoptotic exosome-like vesicles and their roles as damage-associated molecular patterns. Proc. Natl. Acad. Sci. USA 2018, 115, E11721–E11730. [Google Scholar] [CrossRef] [Green Version]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Kalluri, R.; LeBleu, V.S. Discovery of Double-Stranded Genomic DNA in Circulating Exosomes. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 275–280. [Google Scholar] [CrossRef] [Green Version]
- Kurywchak, P.; Kalluri, R. An evolving function of DNA-containing exosomes in chemotherapy-induced immune response. Cell Res. 2017, 27, 722–723. [Google Scholar] [CrossRef] [Green Version]
- Llorente, A.; Skotland, T.; Sylvänne, T.; Kauhanen, D.; Róg, T.; Orłowski, A.; Vattulainen, I.; Ekroos, K.; Sandvig, K. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2013, 1831, 1302–1309. [Google Scholar] [CrossRef]
- Lydic, T.A.; Townsend, S.; Adda, C.G.; Collins, C.; Mathivanan, S.; Reid, G.E. Rapid and comprehensive ‘shotgun’ lipidome profiling of colorectal cancer cell derived exosomes. Methods 2015, 87, 83–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skotland, T.; Sandvig, K.; Llorente, A. Lipids in exosomes: Current knowledge and the way forward. Prog. Lipid Res. 2017, 66, 30–41. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.W.; Lee, H.M.; Lee, T.H.; Kang, C.; Kleinman, H.K.; Gho, Y.S. Extracellular Membrane Vesicles from Tumor Cells Promote Angiogenesis via Sphingomyelin. Cancer Res. 2002, 62, 6312–6317. [Google Scholar] [PubMed]
- Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
- Reinhardt, T.A.; Sacco, R.E.; Nonnecke, B.J.; Lippolis, J.D. Bovine milk proteome: Quantitative changes in normal milk exosomes, milk fat globule membranes and whey proteomes resulting from Staphylococcus aureus mastitis. J. Proteom. 2013, 82, 141–154. [Google Scholar] [CrossRef]
- Miyake, H.; Lee, C.; Chusilp, S.; Bhalla, M.; Li, B.; Pitino, M.; Seo, S.; O’Connor, D.L.; Pierro, A. Human breast milk exosomes attenuate intestinal damage. Pediatric Surg. Int. 2020, 36, 155–163. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Xie, M.-Y.; Sun, J.-J.; Ye, R.-S.; Cheng, X.; Sun, R.-P.; Wei, L.-M.; Li, M.; Lin, D.-L.; Jiang, Q.-Y.; et al. Porcine milk-derived exosomes promote proliferation of intestinal epithelial cells. Sci. Rep. 2016, 6, 33862. [Google Scholar] [CrossRef]
- Gao, H.N.; Guo, H.Y.; Zhang, H.; Xie, X.L.; Wen, P.C.; Ren, F.Z. Yak-milk-derived exosomes promote proliferation of intestinal epithelial cells in an hypoxic environment. J. Dairy Sci. 2019, 102, 985–996. [Google Scholar] [CrossRef] [Green Version]
- Martin, C.; Patel, M.; Williams, S.; Arora, H.; Brawner, K.; Sims, B. Human breast milk-derived exosomes attenuate cell death in intestinal epithelial cells. Innate Immun. 2018, 24, 278–284. [Google Scholar] [CrossRef]
- Melnik, B.C.; Schmitz, G. Milk’s Role as an Epigenetic Regulator in Health and Disease. Diseases 2017, 5, 12. [Google Scholar] [CrossRef] [PubMed]
- Meng, F.; Henson, R.; Wehbe–Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 Regulates Expression of the PTEN Tumor Suppressor Gene in Human Hepatocellular Cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [Green Version]
- Murata, T.; Takayama, K.; Katayama, S.; Urano, T.; Horie-Inoue, K.; Ikeda, K.; Takahashi, S.; Kawazu, C.; Hasegawa, A.; Ouchi, Y.; et al. miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis. 2010, 13, 356–361. [Google Scholar] [CrossRef]
- Fonseka, P.; Liem, M.; Ozcitti, C.; Adda, C.G.; Ang, C.-S.; Mathivanan, S. Exosomes from N-Myc amplified neuroblastoma cells induce migration and confer chemoresistance to non-N-Myc amplified cells: Implications of intra-tumour heterogeneity. J. Extracell. Vesicles 2019, 8, 1597614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pavlyukov, M.S.; Yu, H.; Bastola, S.; Minata, M.; Shender, V.O.; Lee, Y.; Zhang, S.; Wang, J.; Komarova, S.; Wang, J.; et al. Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma Via Intercellular Transfer of Splicing Factors. Cancer Cell 2018, 34, 119–135.e110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gallet, R.; Dawkins, J.; Valle, J.; Simsolo, E.; de Couto, G.; Middleton, R.; Tseliou, E.; Luthringer, D.; Kreke, M.; Smith, R.R.; et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur. Heart J. 2017, 38, 201–211. [Google Scholar] [CrossRef] [Green Version]
- Zhao, G.; Liu, F.; Liu, Z.; Zuo, K.; Wang, B.; Zhang, Y.; Han, X.; Lian, A.; Wang, Y.; Liu, M.; et al. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res. Ther. 2020, 11, 174. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Zhang, L.; Li, Y.; Chen, L.; Wang, X.; Guo, W.; Zhang, X.; Qin, G.; He, S.H.; Zimmerman, A.; et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int. J. Cardiol. 2015, 192, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, L.; Jin, X.; Hu, C.F.; Li, R.; Zhou, Z.; Shen, C.X. Exosomes Derived from Mesenchymal Stem Cells Rescue Myocardial Ischaemia/Reperfusion Injury by Inducing Cardiomyocyte Autophagy Via AMPK and Akt Pathways. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2017, 43, 52–68. [Google Scholar] [CrossRef] [PubMed]
- Yu, B.; Kim, H.W.; Gong, M.; Wang, J.; Millard, R.W.; Wang, Y.; Ashraf, M.; Xu, M. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int. J. Cardiol. 2015, 182, 349–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Liu, S.; Qiu, X.; Yang, X.; Bao, L.; Pu, F.; Liu, X.; Li, C.; Xuan, K.; Zhou, J.; et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy 2020, 16, 2140–2155. [Google Scholar] [CrossRef] [PubMed]
- Gupta, K.H.; Goldufsky, J.W.; Wood, S.J.; Tardi, N.J.; Moorthy, G.S.; Gilbert, D.Z.; Zayas, J.P.; Hahm, E.; Altintas, M.M.; Reiser, J.; et al. Apoptosis and Compensatory Proliferation Signaling Are Coupled by CrkI-Containing Microvesicles. Dev. Cell 2017, 41, 674–684.e675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Winau, F.; Weber, S.; Sad, S.; de Diego, J.; Hoops, S.L.; Breiden, B.; Sandhoff, K.; Brinkmann, V.; Kaufmann, S.H.; Schaible, U.E. Apoptotic vesicles crossprime CD8 T cells and protect against tuberculosis. Immunity 2006, 24, 105–117. [Google Scholar] [CrossRef] [Green Version]
- Kranich, J.; Krautler, N.J.; Falsig, J.; Ballmer, B.; Li, S.; Hutter, G.; Schwarz, P.; Moos, R.; Julius, C.; Miele, G.; et al. Engulfment of cerebral apoptotic bodies controls the course of prion disease in a mouse strain-dependent manner. J. Exp. Med. 2010, 207, 2271–2281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Liao, L.; Tian, W. Extracellular Vesicles Derived from Apoptotic Cells: An Essential Link Between Death and Regeneration. Front. Cell Dev. Biol. 2020, 8, 573511. [Google Scholar] [CrossRef] [PubMed]
- Schorey, J.S.; Cheng, Y.; Singh, P.P.; Smith, V.L. Exosomes and other extracellular vesicles in host–pathogen interactions. EMBO Rep. 2015, 16, 24–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuipers, M.E.; Hokke, C.H.; Smits, H.H.; Nolte-‘t Hoen, E.N.M. Pathogen-Derived Extracellular Vesicle-Associated Molecules That Affect the Host Immune System: An Overview. Front. Microbiol. 2018, 9, 2182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munhoz da Rocha, I.F.; Amatuzzi, R.F.; Lucena, A.C.R.; Faoro, H.; Alves, L.R. Cross-Kingdom Extracellular Vesicles EV-RNA Communication as a Mechanism for Host–Pathogen Interaction. Front. Cell. Infect. Microbiol. 2020, 10, 593160. [Google Scholar] [CrossRef]
- Martins, S.d.T.; Alves, L.R. Extracellular Vesicles in Viral Infections: Two Sides of the Same Coin? Front. Cell. Infect. Microbiol. 2020, 10, 593170. [Google Scholar] [CrossRef] [PubMed]
- Rahimian, P.; He, J.J. Exosome-associated release, uptake, and neurotoxicity of HIV-1 Tat protein. J. Neurovirology 2016, 22, 774–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keryer-Bibens, C.; Pioche-Durieu, C.; Villemant, C.; Souquère, S.; Nishi, N.; Hirashima, M.; Middeldorp, J.; Busson, P. Exosomes released by EBV-infected nasopharyngeal carcinoma cells convey the viral Latent Membrane Protein 1 and the immunomodulatory protein galectin 9. BMC Cancer 2006, 6, 283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balaji, K.N.; Goyal, G.; Narayana, Y.; Srinivas, M.; Chaturvedi, R.; Mohammad, S. Apoptosis triggered by Rv1818c, a PE family gene from Mycobacterium tuberculosis is regulated by mitochondrial intermediates in T cells. Microbes Infect. 2007, 9, 271–281. [Google Scholar] [CrossRef] [PubMed]
- Vanaja, S.K.; Russo, A.J.; Behl, B.; Banerjee, I.; Yankova, M.; Deshmukh, S.D.; Rathinam, V.A.K. Bacterial Outer Membrane Vesicles Mediate Cytosolic Localization of LPS and Caspase-11 Activation. Cell 2016, 165, 1106–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deo, P.; Chow, S.H.; Hay, I.D.; Kleifeld, O.; Costin, A.; Elgass, K.D.; Jiang, J.H.; Ramm, G.; Gabriel, K.; Dougan, G.; et al. Outer membrane vesicles from Neisseria gonorrhoeae target PorB to mitochondria and induce apoptosis. PLoS Pathog. 2018, 14, e1006945. [Google Scholar] [CrossRef] [Green Version]
- Kunsmann, L.; Rüter, C.; Bauwens, A.; Greune, L.; Glüder, M.; Kemper, B.; Fruth, A.; Wai, S.N.; He, X.; Lloubes, R.; et al. Virulence from vesicles: Novel mechanisms of host cell injury by Escherichia coli O104:H4 outbreak strain. Sci. Rep. 2015, 5, 13252. [Google Scholar] [CrossRef] [Green Version]
- Guay, C.; Kruit, J.K.; Rome, S.; Menoud, V.; Mulder, N.L.; Jurdzinski, A.; Mancarella, F.; Sebastiani, G.; Donda, A.; Gonzalez, B.J.; et al. Lymphocyte-Derived Exosomal MicroRNAs Promote Pancreatic β Cell Death and May Contribute to Type 1 Diabetes Development. Cell Metab. 2019, 29, 348–361.e346. [Google Scholar] [CrossRef] [Green Version]
- Wieckowski, E.U.; Visus, C.; Szajnik, M.; Szczepanski, M.J.; Storkus, W.J.; Whiteside, T.L. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J. Immunol. 2009, 183, 3720–3730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mazzeo, C.; Cañas, J.A.; Zafra, M.P.; Rojas Marco, A.; Fernández-Nieto, M.; Sanz, V.; Mittelbrunn, M.; Izquierdo, M.; Baixaulli, F.; Sastre, J.; et al. Exosome secretion by eosinophils: A possible role in asthma pathogenesis. J. Allergy Clin. Immunol. 2015, 135, 1603–1613. [Google Scholar] [CrossRef] [PubMed]
- Cañas, J.A.; Sastre, B.; Rodrigo-Muñoz, J.M.; Fernández-Nieto, M.; Barranco, P.; Quirce, S.; Sastre, J.; Del Pozo, V. Eosinophil-derived exosomes contribute to asthma remodelling by activating structural lung cells. Clin. Exp. Allergy 2018, 48, 1173–1185. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; Exline, M.; Habyarimana, F.; Gavrilin, M.A.; Baker, P.J.; Masters, S.L.; Wewers, M.D.; Sarkar, A. Microparticulate Caspase 1 Regulates Gasdermin D and Pulmonary Vascular Endothelial Cell Injury. Am. J. Respir. Cell Mol. Biol. 2018, 59, 56–64. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; Wewers, M.D.; Sarkar, A. Mononuclear Phagocyte-Derived Microparticulate Caspase-1 Induces Pulmonary Vascular Endothelial Cell Injury. PLoS ONE 2015, 10, e0145607. [Google Scholar] [CrossRef] [Green Version]
- Kubo, H. Extracellular Vesicles in Lung Disease. Chest 2018, 153, 210–216. [Google Scholar] [CrossRef]
- Medzhitov, R. Inflammation 2010: New adventures of an old flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davidovich, P.; Kearney, C.J.; Martin, S.J. Inflammatory outcomes of apoptosis, necrosis and necroptosis. Biol. Chem. 2014, 395, 1163–1171. [Google Scholar] [CrossRef] [PubMed]
- Jorgensen, I.; Miao, E.A. Pyroptotic cell death defends against intracellular pathogens. Immunol. Rev. 2015, 265, 130–142. [Google Scholar] [CrossRef]
- Hannoodee, S.; Nasuruddin, D.N. Acute Inflammatory Response; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
- Heneka, M.T.; Kummer, M.P.; Stutz, A.; Delekate, A.; Schwartz, S.; Vieira-Saecker, A.; Griep, A.; Axt, D.; Remus, A.; Tzeng, T.C.; et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013, 493, 674–678. [Google Scholar] [CrossRef] [PubMed]
- Ising, C.; Venegas, C.; Zhang, S.; Scheiblich, H.; Schmidt, S.V.; Vieira-Saecker, A.; Schwartz, S.; Albasset, S.; McManus, R.M.; Tejera, D.; et al. NLRP3 inflammasome activation drives tau pathology. Nature 2019, 575, 669–673. [Google Scholar] [CrossRef] [PubMed]
- Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nunez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
- Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef]
- Gasser, O.; Schifferli, J.A. Activated polymorphonuclear neutrophils disseminate anti-inflammatory microparticles by ectocytosis. Blood 2004, 104, 2543–2548. [Google Scholar] [CrossRef] [PubMed]
- Hardy, M.P.; Audemard, É.; Migneault, F.; Feghaly, A.; Brochu, S.; Gendron, P.; Boilard, É.; Major, F.; Dieudé, M.; Hébert, M.J.; et al. Apoptotic endothelial cells release small extracellular vesicles loaded with immunostimulatory viral-like RNAs. Sci. Rep. 2019, 9, 7203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mardpour, S.; Hamidieh, A.A.; Taleahmad, S.; Sharifzad, F.; Taghikhani, A.; Baharvand, H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J. Cell Physiol. 2019, 234, 8249–8258. [Google Scholar] [CrossRef] [PubMed]
- Obregon, C.; Rothen-Rutishauser, B.; Gerber, P.; Gehr, P.; Nicod, L.P. Active uptake of dendritic cell-derived exovesicles by epithelial cells induces the release of inflammatory mediators through a TNF-alpha-mediated pathway. Am. J. Pathol. 2009, 175, 696–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [Green Version]
- Park, K.S.; Choi, K.H.; Kim, Y.S.; Hong, B.S.; Kim, O.Y.; Kim, J.H.; Yoon, C.M.; Koh, G.Y.; Kim, Y.K.; Gho, Y.S. Outer membrane vesicles derived from Escherichia coli induce systemic inflammatory response syndrome. PLoS ONE 2010, 5, e11334. [Google Scholar] [CrossRef] [Green Version]
- Lenassi, M.; Cagney, G.; Liao, M.; Vaupotic, T.; Bartholomeeusen, K.; Cheng, Y.; Krogan, N.J.; Plemenitas, A.; Peterlin, B.M. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic 2010, 11, 110–122. [Google Scholar] [CrossRef] [Green Version]
- Nieuwland, R.; Berckmans, R.J.; McGregor, S.; Boing, A.N.; Romijn, F.P.; Westendorp, R.G.; Hack, C.E.; Sturk, A. Cellular origin and procoagulant properties of microparticles in meningococcal sepsis. Blood 2000, 95, 930–935. [Google Scholar] [CrossRef]
- Gao, K.; Jin, J.; Huang, C.; Li, J.; Luo, H.; Li, L.; Huang, Y.; Jiang, Y. Exosomes Derived from Septic Mouse Serum Modulate Immune Responses via Exosome-Associated Cytokines. Front. Immunol. 2019, 10, 1560. [Google Scholar] [CrossRef] [Green Version]
- Fitzgerald, W.; Freeman, M.L.; Lederman, M.M.; Vasilieva, E.; Romero, R.; Margolis, L. A System of Cytokines Encapsulated in ExtraCellular Vesicles. Sci. Rep. 2018, 8, 8973. [Google Scholar] [CrossRef] [Green Version]
- Fujimi, S.; Ogura, H.; Tanaka, H.; Koh, T.; Hosotsubo, H.; Nakamori, Y.; Kuwagata, Y.; Shimazu, T.; Sugimoto, H. Activated polymorphonuclear leukocytes enhance production of leukocyte microparticles with increased adhesion molecules in patients with sepsis. J. Trauma 2002, 52, 443–448. [Google Scholar] [CrossRef]
- Shah, B.; Sullivan, C.J.; Lonergan, N.E.; Stanley, S.; Soult, M.C.; Britt, L.D. Circulating bacterial membrane vesicles cause sepsis in rats. Shock 2012, 37, 621–628. [Google Scholar] [CrossRef]
- Gambim, M.H.; do Carmo Ade, O.; Marti, L.; Verissimo-Filho, S.; Lopes, L.R.; Janiszewski, M. Platelet-derived exosomes induce endothelial cell apoptosis through peroxynitrite generation: Experimental evidence for a novel mechanism of septic vascular dysfunction. Crit. Care 2007, 11, R107. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Yao, X.; Yao, H.; Ji, Q.; Ding, G.; Liu, X. Exosomal miR-103-3p from LPS-activated THP-1 macrophage contributes to the activation of hepatic stellate cells. FASEB J. 2020, 34, 5178–5192. [Google Scholar] [CrossRef] [PubMed]
- Timar, C.I.; Lorincz, A.M.; Csepanyi-Komi, R.; Valyi-Nagy, A.; Nagy, G.; Buzas, E.I.; Ivanyi, Z.; Kittel, A.; Powell, D.W.; McLeish, K.R.; et al. Antibacterial effect of microvesicles released from human neutrophilic granulocytes. Blood 2013, 121, 510–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mostefai, H.A.; Meziani, F.; Mastronardi, M.L.; Agouni, A.; Heymes, C.; Sargentini, C.; Asfar, P.; Martinez, M.C.; Andriantsitohaina, R. Circulating microparticles from patients with septic shock exert protective role in vascular function. Am. J. Respir. Crit. Care Med. 2008, 178, 1148–1155. [Google Scholar] [CrossRef] [Green Version]
- Dalli, J.; Norling, L.V.; Montero-Melendez, T.; Federici Canova, D.; Lashin, H.; Pavlov, A.M.; Sukhorukov, G.B.; Hinds, C.J.; Perretti, M. Microparticle alpha-2-macroglobulin enhances pro-resolving responses and promotes survival in sepsis. EMBO Mol. Med. 2014, 6, 27–42. [Google Scholar] [CrossRef] [PubMed]
- Miksa, M.; Wu, R.; Dong, W.; Komura, H.; Amin, D.; Ji, Y.; Wang, Z.; Wang, H.; Ravikumar, T.S.; Tracey, K.J.; et al. Immature dendritic cell-derived exosomes rescue septic animals via milk fat globule epidermal growth factor-factor VIII [corrected]. J. Immunol. 2009, 183, 5983–5990. [Google Scholar] [CrossRef]
- Kesimer, M.; Scull, M.; Brighton, B.; DeMaria, G.; Burns, K.; O’Neal, W.; Pickles, R.J.; Sheehan, J.K. Characterization of exosome-like vesicles released from human tracheobronchial ciliated epithelium: A possible role in innate defense. FASEB J. 2009, 23, 1858–1868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bourdonnay, E.; Zaslona, Z.; Penke, L.R.; Speth, J.M.; Schneider, D.J.; Przybranowski, S.; Swanson, J.A.; Mancuso, P.; Freeman, C.M.; Curtis, J.L.; et al. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J. Exp. Med. 2015, 212, 729–742. [Google Scholar] [CrossRef]
- Moon, H.G.; Kim, S.H.; Gao, J.; Quan, T.; Qin, Z.; Osorio, J.C.; Rosas, I.O.; Wu, M.; Tesfaigzi, Y.; Jin, Y. CCN1 secretion and cleavage regulate the lung epithelial cell functions after cigarette smoke. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, L326–L337. [Google Scholar] [CrossRef] [Green Version]
- Kulshreshtha, A.; Ahmad, T.; Agrawal, A.; Ghosh, B. Proinflammatory role of epithelial cell-derived exosomes in allergic airway inflammation. J. Allergy Clin. Immunol. 2013, 131, 1194–1203.e14. [Google Scholar] [CrossRef] [PubMed]
- Skokos, D.; Le Panse, S.; Villa, I.; Rousselle, J.C.; Peronet, R.; Namane, A.; David, B.; Mecheri, S. Nonspecific B and T cell-stimulatory activity mediated by mast cells is associated with exosomes. Int. Arch. Allergy Immunol. 2001, 124, 133–136. [Google Scholar] [CrossRef] [PubMed]
- Xie, G.; Yang, H.; Peng, X.; Lin, L.; Wang, J.; Lin, K.; Cui, Z.; Li, J.; Xiao, H.; Liang, Y.; et al. Mast cell exosomes can suppress allergic reactions by binding to IgE. J. Allergy Clin. Immunol. 2018, 141, 788–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Canas, J.A.; Sastre, B.; Mazzeo, C.; Fernandez-Nieto, M.; Rodrigo-Munoz, J.M.; Gonzalez-Guerra, A.; Izquierdo, M.; Barranco, P.; Quirce, S.; Sastre, J.; et al. Exosomes from eosinophils autoregulate and promote eosinophil functions. J. Leukoc. Biol. 2017, 101, 1191–1199. [Google Scholar] [CrossRef] [Green Version]
- Admyre, C.; Johansson, S.M.; Paulie, S.; Gabrielsson, S. Direct exosome stimulation of peripheral human T cells detected by ELISPOT. Eur. J. Immunol. 2006, 36, 1772–1781. [Google Scholar] [CrossRef]
- Admyre, C.; Bohle, B.; Johansson, S.M.; Focke-Tejkl, M.; Valenta, R.; Scheynius, A.; Gabrielsson, S. B cell-derived exosomes can present allergen peptides and activate allergen-specific T cells to proliferate and produce TH2-like cytokines. J. Allergy Clin. Immunol. 2007, 120, 1418–1424. [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]
- Balbi, C.; Burrello, J.; Bolis, S.; Lazzarini, E.; Biemmi, V.; Pianezzi, E.; Burrello, A.; Caporali, E.; Grazioli, L.G.; Martinetti, G.; et al. Circulating extracellular vesicles are endowed with enhanced procoagulant activity in SARS-CoV-2 infection. EBioMedicine 2021, 67, 103369. [Google Scholar] [CrossRef]
- Guervilly, C.; Bonifay, A.; Burtey, S.; Sabatier, F.; Cauchois, R.; Abdili, E.; Arnaud, L.; Lano, G.; Pietri, L.; Robert, T.; et al. Dissemination of extreme levels of extracellular vesicles: Tissue factor activity in patients with severe COVID-19. Blood Adv. 2021, 5, 628–634. [Google Scholar] [CrossRef]
- Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef] [PubMed]
- Hotchkiss, R.S.; Moldawer, L.L.; Opal, S.M.; Reinhart, K.; Turnbull, I.R.; Vincent, J.L. Sepsis and septic shock. Nat. Rev. Dis. Primers 2016, 2, 16045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raeven, P.; Zipperle, J.; Drechsler, S. Extracellular Vesicles as Markers and Mediators in Sepsis. Theranostics 2018, 8, 3348–3365. [Google Scholar] [CrossRef]
- Labbe, K.; Saleh, M. Cell death in the host response to infection. Cell Death Differ. 2008, 15, 1339–1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vincent, J.L.; Sakr, Y.; Sprung, C.L.; Ranieri, V.M.; Reinhart, K.; Gerlach, H.; Moreno, R.; Carlet, J.; Le Gall, J.R.; Payen, D.; et al. Sepsis in European intensive care units: Results of the SOAP study. Crit. Care Med. 2006, 34, 344–353. [Google Scholar] [CrossRef] [PubMed]
- Hotchkiss, R.S.; Nicholson, D.W. Apoptosis and caspases regulate death and inflammation in sepsis. Nat. Rev. Immunol. 2006, 6, 813–822. [Google Scholar] [CrossRef] [PubMed]
- Kim, O.Y.; Choi, S.J.; Jang, S.C.; Park, K.S.; Kim, S.R.; Choi, J.P.; Lim, J.H.; Lee, S.W.; Park, J.; Di Vizio, D.; et al. Bacterial protoplast-derived nanovesicles as vaccine delivery system against bacterial infection. Nano Lett. 2015, 15, 266–274. [Google Scholar] [CrossRef]
- Larsson, A.; Lundahl, T.; Eriksson, M.; Lundkvist, K.; Lindahl, T. Endotoxin induced platelet microvesicle formation measured by flow cytometry. Platelets 1996, 7, 153–158. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Jy, W.; Horstman, L.L.; Janania, J.; Reyes, Y.; Kelley, R.E.; Ahn, Y.S. Elevated platelet microparticles in transient ischemic attacks, lacunar infarcts, and multiinfarct dementias. Thromb. Res. 1993, 72, 295–304. [Google Scholar] [CrossRef]
- Holme, P.A.; Solum, N.O.; Brosstad, F.; Roger, M.; Abdelnoor, M. Demonstration of platelet-derived microvesicles in blood from patients with activated coagulation and fibrinolysis using a filtration technique and western blotting. Thromb. Haemost. 1994, 72, 666–671. [Google Scholar]
- Lundahl, T.H.; Lindahl, T.L.; Fagerberg, I.H.; Egberg, N.; Bunescu, A.; Larsson, A. Activated platelets and impaired platelet function in intensive care patients analyzed by flow cytometry. Blood Coagul. Fibrinolysis 1996, 7, 218–220. [Google Scholar] [CrossRef] [PubMed]
- Denning, N.L.; Aziz, M.; Gurien, S.D.; Wang, P. DAMPs and NETs in Sepsis. Front. Immunol. 2019, 10, 2536. [Google Scholar] [CrossRef]
- Gangoda, L.; Schenk, R.L.; Best, S.A.; Nedeva, C.; Louis, C.; D’Silva, D.B.; Fairfax, K.; Jarnicki, A.G.; Puthalakath, H.; Sutherland, K.D.; et al. Absence of pro-survival A1 has no impact on inflammatory cell survival in vivo during acute lung inflammation and peritonitis. Cell Death Differ. 2021. [Google Scholar] [CrossRef] [PubMed]
- Nedeva, C.; Menassa, J.; Duan, M.; Liu, C.; Doerflinger, M.; Kueh, A.J.; Herold, M.J.; Fonseka, P.; Phan, T.K.; Faou, P.; et al. TREML4 receptor regulates inflammation and innate immune cell death during polymicrobial sepsis. Nat. Immunol. 2020, 21, 1585–1596. [Google Scholar] [CrossRef] [PubMed]
- Herrmann, I.K.; Bertazzo, S.; O’Callaghan, D.J.; Schlegel, A.A.; Kallepitis, C.; Antcliffe, D.B.; Gordon, A.C.; Stevens, M.M. Differentiating sepsis from non-infectious systemic inflammation based on microvesicle-bacteria aggregation. Nanoscale 2015, 7, 13511–13520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Essandoh, K.; Yang, L.; Wang, X.; Huang, W.; Qin, D.; Hao, J.; Wang, Y.; Zingarelli, B.; Peng, T.; Fan, G.C. Blockade of exosome generation with GW4869 dampens the sepsis-induced inflammation and cardiac dysfunction. Biochim. Biophys. Acta 2015, 1852, 2362–2371. [Google Scholar] [CrossRef] [Green Version]
- Ren, C.; Yao, R.Q.; Zhang, H.; Feng, Y.W.; Yao, Y.M. Sepsis-associated encephalopathy: A vicious cycle of immunosuppression. J. Neuroinflammation 2020, 17, 14. [Google Scholar] [CrossRef]
- Xu, Y.; Ku, X.; Wu, C.; Cai, C.; Tang, J.; Yan, W. Exosomal proteome analysis of human plasma to monitor sepsis progression. Biochem. Biophys. Res. Commun. 2018, 499, 856–861. [Google Scholar] [CrossRef] [PubMed]
- Englert, J.A.; Bobba, C.; Baron, R.M. Integrating molecular pathogenesis and clinical translation in sepsis-induced acute respiratory distress syndrome. JCI Insight 2019, 4, e124061. [Google Scholar] [CrossRef] [Green Version]
- Fujita, Y.; Kosaka, N.; Araya, J.; Kuwano, K.; Ochiya, T. Extracellular vesicles in lung microenvironment and pathogenesis. Trends Mol. Med. 2015, 21, 533–542. [Google Scholar] [CrossRef] [PubMed]
- Mohan, A.; Agarwal, S.; Clauss, M.; Britt, N.S.; Dhillon, N.K. Extracellular vesicles: Novel communicators in lung diseases. Respir. Res. 2020, 21, 175. [Google Scholar] [CrossRef]
- Letsiou, E.; Sammani, S.; Zhang, W.; Zhou, T.; Quijada, H.; Moreno-Vinasco, L.; Dudek, S.M.; Garcia, J.G. Pathologic mechanical stress and endotoxin exposure increases lung endothelial microparticle shedding. Am. J. Respir. Cell Mol. Biol. 2015, 52, 193–204. [Google Scholar] [CrossRef] [PubMed]
- Li, C.J.; Liu, Y.; Chen, Y.; Yu, D.; Williams, K.J.; Liu, M.L. Novel proteolytic microvesicles released from human macrophages after exposure to tobacco smoke. Am. J. Pathol. 2013, 182, 1552–1562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moon, H.G.; Zheng, Y.; An, C.H.; Kim, Y.K.; Jin, Y. CCN1 secretion induced by cigarette smoking extracts augments IL-8 release from bronchial epithelial cells. PLoS ONE 2013, 8, e68199. [Google Scholar] [CrossRef] [PubMed]
- Schutze, N.; Rucker, N.; Muller, J.; Adamski, J.; Jakob, F. 5’ flanking sequence of the human immediate early responsive gene ccn1 (cyr61) and mapping of polymorphic CA repeat sequence motifs in the human ccn1 (cyr61) locus. Mol. Pathol. 2001, 54, 170–175. [Google Scholar] [CrossRef]
- Hamid, Q.; Tulic, M.K.; Liu, M.C.; Moqbel, R. Inflammatory cells in asthma: Mechanisms and implications for therapy. J. Allergy Clin. Immunol. 2003, 111, S5–S12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, K.Y. Pneumonia, Acute Respiratory Distress Syndrome, and Early Immune-Modulator Therapy. Int. J. Mol. Sci. 2017, 18, 388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsushima, K.; King, L.S.; Aggarwal, N.R.; De Gorordo, A.; D’Alessio, F.R.; Kubo, K. Acute lung injury review. Intern. Med. 2009, 48, 621–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matthay, M.A.; Ware, L.B.; Zimmerman, G.A. The acute respiratory distress syndrome. J. Clin. Invest. 2012, 122, 2731–2740. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Zhang, D.; Laskin, D.L.; Jin, Y. Functional Evidence of Pulmonary Extracellular Vesicles in Infectious and Noninfectious Lung Inflammation. J. Immunol. 2018, 201, 1500–1509. [Google Scholar] [CrossRef] [PubMed]
- Jiang, K.; Yang, J.; Guo, S.; Zhao, G.; Wu, H.; Deng, G. Peripheral Circulating Exosome-Mediated Delivery of miR-155 as a Novel Mechanism for Acute Lung Inflammation. Mol. Ther. 2019, 27, 1758–1771. [Google Scholar] [CrossRef] [PubMed]
- Pocsfalvi, G.; Mammadova, R.; Ramos Juarez, A.P.; Bokka, R.; Trepiccione, F.; Capasso, G. COVID-19 and Extracellular Vesicles: An Intriguing Interplay. Kidney Blood Press Res. 2020, 45, 661–670. [Google Scholar] [CrossRef]
- Rodrigues, T.S.; de Sa, K.S.G.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Goncalves, A.V.; Perucello, D.B.; Andrade, W.A.; Castro, R.; et al. Inflammasomes are activated in response to SARS-CoV-2 infection and are associated with COVID-19 severity in patients. J. Exp. Med. 2021, 218, e20201707. [Google Scholar] [CrossRef]
- Xiao, K.; Hou, F.; Huang, X.; Li, B.; Qian, Z.R.; Xie, L. Mesenchymal stem cells: Current clinical progress in ARDS and COVID-19. Stem Cell Res. Ther. 2020, 11, 305. [Google Scholar] [CrossRef]
- Barberis, E.; Vanella, V.V.; Falasca, M.; Caneapero, V.; Cappellano, G.; Raineri, D.; Ghirimoldi, M.; De Giorgis, V.; Puricelli, C.; Vaschetto, R.; et al. Circulating Exosomes Are Strongly Involved in SARS-CoV-2 Infection. Front. Mol. Biosci. 2021, 8, 632290. [Google Scholar] [CrossRef] [PubMed]
- Song, J.W.; Lam, S.M.; Fan, X.; Cao, W.J.; Wang, S.Y.; Tian, H.; Chua, G.H.; Zhang, C.; Meng, F.P.; Xu, Z.; et al. Omics-Driven Systems Interrogation of Metabolic Dysregulation in COVID-19 Pathogenesis. Cell Metab. 2020, 32, 188–202.e185. [Google Scholar] [CrossRef]
- Cocozza, F.; Nevo, N.; Piovesana, E.; Lahaye, X.; Buchrieser, J.; Schwartz, O.; Manel, N.; Tkach, M.; Thery, C.; Martin-Jaular, L. Extracellular vesicles containing ACE2 efficiently prevent infection by SARS-CoV-2 Spike protein-containing virus. J. Extracell. Vesicles 2020, 10, e12050. [Google Scholar] [CrossRef]
- Tsiapalis, D.; O’Driscoll, L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020, 9, 911. [Google Scholar] [CrossRef] [Green Version]
- O’Driscoll, L. Extracellular vesicles from mesenchymal stem cells as a Covid-19 treatment. Drug Discov. Today 2020, 25, 1124–1125. [Google Scholar] [CrossRef] [PubMed]
- Khalaj, K.; Figueira, R.L.; Antounians, L.; Lauriti, G.; Zani, A. Systematic review of extracellular vesicle-based treatments for lung injury: Are EVs a potential therapy for COVID-19? J. Extracell. Vesicles 2020, 9, 1795365. [Google Scholar] [CrossRef]
- Xia, X.; Yuan, P.; Liu, Y.; Wang, Y.; Cao, W.; Zheng, J.C. Emerging roles of extracellular vesicles in COVID-19, a double-edged sword? Immunology 2021, 163, 416–430. [Google Scholar] [CrossRef] [PubMed]
Cargo | EV Source | Function | Reference |
---|---|---|---|
NS | Milk (bovine, porcine, human) | Attenuate cell death in IECs | [47,48,49,50] |
miR-148a, miR-21 | Milk (bovine) | OncomiRs, inhibition of tumor suppressor genes | [51,52,53] |
NS | N-myc amplified neuroblastoma cells | Chemoresistance and enhanced survival in non N-myc amplified tumor cells | [54] |
Spliceosome components | Apoptotic glioblastoma cells | Enhanced survival in recipient tumor cells | [55] |
NS | MSCs (exosomes) | Enhance proliferation and survival of keratinocytes, cardiomyocytes and endotheliocytes | [57,58,59,60] |
NS | MSCs (ApoEVs) | Regulation of cardiac function and autophagy, promote nephron repair | [61,62] |
NS | Pathogen infected host cells | Immune regulation and enhanced pathogen clearance to prevent spread of infection | [63,64] |
Cargo | EV Source | Function | Reference |
---|---|---|---|
HIV Tat protein | HIV infected astrocytes | Neuronal cell death | [70] |
Latent membrane protein-1, galectin-9 | Epsetien-Barr virus infected nasopharyngeal carcinoma cells | T lymphocyte cell death | [71] |
NS | Mycobacterium tuberculosis infected T-lymphocytes | Apoptosis in Jurkat T cells | [72] |
PAMPs | Pathogen infected macrophages | Pro-inflammatory cytokine release in uninfected macrophages | [66] |
LPS | Gram negative bacteria | Caspase-11 mediated pyroptosis in | [73] |
PorB | Neisseria gonorrhoeae | Apoptosis in macrophages | [74] |
Shiga toxin 2a | Escherichia coli O104:H4 | Cell death in IECs | [75] |
miR-142-3p, miR-142-5p and miR-155 | T-lymphocytes | Selective targeting and apoptosis of pancreatic β cells leading to type-1 diabetes | [76] |
PD-L1 | Metastatic melanomas | Exhaustion of CD8+ T cells leading to immune response evasion | [30] |
Fas ligand | Serum from patients with oral squamous cell carcinoma | CD8+ T cell apoptosis | [77] |
NS | Eosinophils | Airway inflammation and apoptosis in primary alveolar epithelial cells | [78] |
Caspase-1 | Monocytes | Apoptosis of pulmonary microvascular endothelial cells in ALI and ARDS | [80,81] |
NS | Apoptotic endothelial cells | Apoptosis in healthy endothelial cells in COPD | [82] |
Cargo | EV Source | Identified in | Function | Reference |
---|---|---|---|---|
MHC | Dendritic cells, B lymphocytes | NS | antigen specific immune response | [34,91,92] |
NS | Neutrophils | NS | anti-inflammatory effects | [93] |
Sphingosine 1-Phosphate Receptors 1 and SPR1/3 | Bone marrow-derived macrophages | NS | pro-inflammatory effects | [37] |
Non-coding RNA | Endothelial cells | NS | pro-inflammatory effects | [94] |
Chemokines and inflammatory cytokines: TNF, IL-1β, CXCL2, CXCL8 | Dendritic cells, Mesenchymal stromal cells | NS | immune modulatory functions | [95,96,97] |
Bacterial virulence factors | Bacteria | Sepsis | pro-inflammatory effects | [98] |
Nef protein | HIV type 1 infected cells | HIV type 1 infection | anti-inflammatory effects | [99] |
NS | platelets, granulocytes | Meningococcal sepsis | pro-coagulation activity | [100] |
IL-12, IL-15, IL-17, IFN-γ | NS | sepsis | pro-inflammatory effects | [101,102] |
IL-4, IL-10 | NS | sepsis | anti-inflammatory effects | [101,102] |
Adhesion molecules | activated polymorphonuclear leukocytes | sepsis | organ damage | [103,104] |
NS | platelet | sepsis | pro-apoptotic | [105] |
miR-103-3p | activated macrophages | sepsis | organ damage | [106] |
NS | granulocyte | sepsis | anti-bacterial effects | [107] |
NS | sepsis | protecting from vascular dysfunction | [108] | |
alpha-2-macroglobulin | granulocytes | sepsis | bacterial clearance, anti-inflammatory effects | [109] |
NS | Immature dendritic cells | sepsis | anti-inflammatory effects | [110] |
membrane tethered mucins | Lung epithelial cells | - | innate defense | [111] |
SOCS1 and 3 | Alveolar macrophages | cigarette smoke exposure | modulation of inflammatory signalling | [112] |
CCN1 | lung epithelial cells | COPD | pro-inflammatory effects | [113] |
NS | lung epithelial cells | Asthma | pro-inflammatory effects | [114] |
MHC class II, CD86, LFA-1 and ICAM-1 | mast cells | Asthma | pro-inflammatory effects | [115] |
FcεRI | mast cells | Asthma | anti-inflammatory effects | [116] |
NS | eosinophils | Asthma | pro-inflammatory effects | [79,117] |
NS | Dendritic cells | NS | pro-inflammatory effects | [118] |
NS | B cell | Allergy | pro-inflammatory effects | [119] |
Caspase-1 | activated macrophages | NS | pro-apoptotic | [81] |
ACE2, CD9 | SARS-CoV-2 infected cells | COVID-19 | promote infection | [120] |
NS | NS | COVID-19 | pro-coagulation activity | [121,122] |
NS | mesenchymal stem cells | NS | anti-inflammatory | [123] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sanwlani, R.; Gangoda, L. Role of Extracellular Vesicles in Cell Death and Inflammation. Cells 2021, 10, 2663. https://doi.org/10.3390/cells10102663
Sanwlani R, Gangoda L. Role of Extracellular Vesicles in Cell Death and Inflammation. Cells. 2021; 10(10):2663. https://doi.org/10.3390/cells10102663
Chicago/Turabian StyleSanwlani, Rahul, and Lahiru Gangoda. 2021. "Role of Extracellular Vesicles in Cell Death and Inflammation" Cells 10, no. 10: 2663. https://doi.org/10.3390/cells10102663
APA StyleSanwlani, R., & Gangoda, L. (2021). Role of Extracellular Vesicles in Cell Death and Inflammation. Cells, 10(10), 2663. https://doi.org/10.3390/cells10102663