Extracellular Vesicle-Based Therapy for COVID-19: Promises, Challenges and Future Prospects
Abstract
:1. Introduction
2. COVID-19-Associated Multiple Organ Failure and EV-Mediated Recovery
2.1. COVID-19-Associated Lung Damage and Its Recovery by EVs
2.2. COVID-19-Associated Cardiovascular Disease and Its Recovery by EVs
2.3. COVID-19 Associated Kidney Diseases and Their Recovery by EVs
2.4. COVID-19-Associated Liver Disease and Its Recovery by EVs
2.5. COVID-19 Associated Neurological Diseases and Their EV-Mediated Recovery
2.6. EV-Mediated Recovery of COVID-19-Associated Hematological Disorders
3. Translational Potential of EVs in COVID-19 Management
3.1. MSC-Derived EVs as Promising Medications
3.2. Platelet-Derived EV-Based Therapy
3.3. EV-Based Vaccines for COVID-19 Prevention
3.4. Engineered EVs as Delivery Vehicles for COVID-19 Therapy
4. Clinical Trials on EVs for COVID-19 Treatments
5. Challenges in Designing EVs as Therapeutic Candidates
6. Concluding Remarks and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Du, J.; Dong, L.; Wang, T.; Yuan, C.; Fu, R.; Zhang, L.; Liu, B.; Zhang, M.; Yin, Y.; Qin, J.; et al. Psychological symptoms among frontline healthcare workers during COVID-19 outbreak in Wuhan. Gen. Hosp. Psychiatry 2020, 67, 144–145. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Nagoor Meeran, M.F.; Javed, H.; Sharma, C.; Goyal, S.N.; Kumar, S.; Jha, N.K.; Ojha, S. Can Echinacea Be a Potential Candidate to Target Immunity, Inflammation, and Infection-The Trinity of Coronavirus Disease 2019. Heliyon 2021, 7, e05990. [Google Scholar] [CrossRef] [PubMed]
- Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical Course and Risk Factors for Mortality of Adult Inpatients with COVID-19 in Wuhan, China: A Retrospective Cohort Study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
- Meeran, M.F.N.; Sharma, C.; Goyal, S.N.; Kumar, S.; Ojha, S. CB2 Receptor-Selective Agonists as Candidates for Targeting Infection, Inflammation, and Immunity in SARS-CoV-2 Infections. Drug Dev. Res. 2021, 82, 7–11. [Google Scholar] [CrossRef] [PubMed]
- Sahin, A.R.; Erdogan, A.; Mutlu Agaoglu, P.; Dineri, Y.; Cakirci, A.Y.; Senel, M.E.; Okyay, R.A.; Tasdogan, A.M. 2019 Novel Coronavirus (COVID-19) Outbreak: A Review of the Current Literature. EURASIAN J. Med. Oncol. 2020, 4, 1–7. [Google Scholar] [CrossRef]
- Kumar, A.; Prasoon, P.; Sekhawat, P.S.; Pareek, V.; Faiq, M.A.; Kumari, C.; Narayan, R.K.; Kulandhasamy, M.; Kant, K. Pathogenesis guided therapeutic management of COVID-19: An immunological perspective. Int. Rev. Immunol. 2021, 40, 54–71. [Google Scholar] [CrossRef] [PubMed]
- Tsuchiya, A.; Takeuchi, S.; Iwasawa, T.; Kumagai, M.; Sato, T.; Motegi, S.; Ishii, Y.; Koseki, Y.; Tomiyoshi, K.; Natsui, K.; et al. Therapeutic potential of mesenchymal stem cells and their exosomes in severe novel coronavirus disease 2019 (COVID-19) cases. Inflamm. Regen. 2020, 40, 14. [Google Scholar] [CrossRef]
- Al-Khawaga, S.; Abdelalim, E.M. Potential application of mesenchymal stem cells and their exosomes in lung injury: An emerging therapeutic option for COVID-19 patients. Stem Cell Res. Ther. 2020, 11, 437. [Google Scholar] [CrossRef]
- Sanyaolu, A.; Okorie, C.; Marinkovic, A.; Patidar, R.; Younis, K.; Desai, P.; Hosein, Z.; Padda, I.; Mangat, J.; Altaf, M. Comorbidity and Its Impact on Patients with COVID-19. SN Compr. Clin. Med. 2020, 1–8. [Google Scholar] [CrossRef]
- 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]
- Fujita, Y.; Hoshina, T.; Matsuzaki, J.; Kadota, T.; Fujimoto, S.; Kawamoto, H.; Watanabe, N.; Sawaki, K.; Sakamoto, Y.; Miyajima, M.; et al. Early Prediction of COVID-19 Severity Using Extracellular Vesicles and Extracellular RNAs. medRxiv 2020. [Google Scholar] [CrossRef]
- Jabbari, N.; Karimipour, M.; Khaksar, M.; Akbariazar, E.; Heidarzadeh, M.; Mojarad, B.; Aftab, H.; Rahbarghazi, R.; Rezaie, J. Tumor-derived extracellular vesicles: Insights into bystander effects of exosomes after irradiation. Lasers Med. Sci. 2020, 35, 531–545. [Google Scholar] [CrossRef]
- Kowal, J.; Tkach, M.; Théry, C. Biogenesis and Secretion of Exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef] [Green Version]
- Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef] [Green Version]
- Hurley, J.H. ESCRTs Are Everywhere. EMBO J. 2015, 34, 2398–2407. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Kumar, S.; Jayachandran, M.; Herrera Hernandez, L.P.; Wang, S.; Wilson, E.M.; Lieske, J.C. Excretion of Urine Extracellular Vesicles Bearing Markers of Activated Immune Cells and Calcium/Phosphorus Physiology Differ between Calcium Kidney Stone Formers and Non-Stone Formers. BMC Nephrol. 2021, 22, 204. [Google Scholar] [CrossRef] [PubMed]
- Jayachandran, M.; Yuzhakov, S.V.; Kumar, S.; Larson, N.B.; Enders, F.T.; Milliner, D.S.; Rule, A.D.; Lieske, J.C. Specific Populations of Urinary Extracellular Vesicles and Proteins Differentiate Type 1 Primary Hyperoxaluria Patients without and with Nephrocalcinosis or Kidney Stones. Orphanet J. Rare Dis. 2020, 15, 319. [Google Scholar] [CrossRef]
- Nabhan, J.F.; Hu, R.; Oh, R.S.; Cohen, S.N.; Lu, Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc. Natl. Acad. Sci. USA 2012, 109, 4146–4151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Q.; Lu, Q. Plasma Membrane-Derived Extracellular Microvesicles Mediate Non-Canonical Intercellular NOTCH Signaling. Nat. Commun. 2017, 8, 709. [Google Scholar] [CrossRef]
- Li, B.; Antonyak, M.A.; Zhang, J.; Cerione, R.A. RhoA Triggers a Specific Signaling Pathway That Generates Transforming Microvesicles in Cancer Cells. Oncogene 2012, 31, 4740–4749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.-M.; Gould, S.J. The Cis-Acting Signals That Target Proteins to Exosomes and Microvesicles. Biochem. Soc. Trans. 2013, 41, 277–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Dong, M.; Zhang, J.; Ma, X.; Tan, J.; Chen, L.; Liu, S.; Xin, Y.; Zhuang, L. ACE2, TMPRSS2 distribution and extrapulmonary organ injury in patients with COVID-19. Biomed. Pharmacother. 2020, 131, 110678. [Google Scholar] [CrossRef]
- Kumar, A.; Prasoon, P.; Kumari, C.; Pareek, V.; Faiq, M.A.; Narayan, R.K.; Kulandhasamy, M.; Kant, K. SARS-CoV-2-specific virulence factors in COVID-19. J. Med. Virol. 2021, 93, 1343–1350. [Google Scholar] [CrossRef] [PubMed]
- Bhaskar, S.; Sinha, A.; Banach, M.; Mittoo, S.; Weissert, R.; Kass, J.S.; Rajagopal, S.; Pai, A.R.; Kutty, S. Cytokine Storm in COVID-19—Immunopathological Mechanisms, Clinical Considerations, and Therapeutic Approaches: The REPROGRAM Consortium Position Paper. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhou, W.; Yang, L.; You, R. Physiological and pathological regulation of ACE2, the SARS-CoV-2 receptor. Pharmacol. Res. 2020, 157, 104833. [Google Scholar] [CrossRef]
- Chatterjee, S. Understanding the Nature of Variations in Structural Sequences Coding for Coronavirus Spike, Envelope, Membrane and Nucleocapsid Proteins of SARS-CoV-2; Social Science Research Network: Rochester, NY, USA, 2020. [Google Scholar]
- Hussain, A.; Kaler, J.; Tabrez, E.; Tabrez, S.; Tabrez, S.S.M. Novel COVID-19: A Comprehensive Review of Transmission, Manifestation, and Pathogenesis. Cureus 2020, 12. [Google Scholar] [CrossRef]
- Amawi, H.; Abu Deiab, G.I.; Aljabali, A.A.A.; Dua, K.; Tambuwala, M.M. COVID-19 pandemic: An overview of epidemiology, pathogenesis, diagnostics and potential vaccines and therapeutics. Ther. Deliv. 2020, 11, 245–268. [Google Scholar] [CrossRef]
- Moll, G.; Rasmusson-Duprez, I.; von Bahr, L.; Connolly-Andersen, A.-M.; Elgue, G.; Funke, L.; Hamad, O.A.; Lönnies, H.; Magnusson, P.U.; Sanchez, J.; et al. Are Therapeutic Human Mesenchymal Stromal Cells Compatible with Human Blood? Stem Cells 2012, 30, 1565–1574. [Google Scholar] [CrossRef] [Green Version]
- Pacienza, N.; Lee, R.H.; Bae, E.-H.; Kim, D.-K.; Liu, Q.; Prockop, D.J.; Yannarelli, G. In Vitro Macrophage Assay Predicts the In Vivo Anti-inflammatory Potential of Exosomes from Human Mesenchymal Stromal Cells. Mol. Ther. Methods Clin. Dev. 2019, 13, 67–76. [Google Scholar] [CrossRef] [Green Version]
- Campagnoli, C.; Roberts, I.A.; Kumar, S.; Bennett, P.R.; Bellantuono, I.; Fisk, N.M. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001, 98, 2396–2402. [Google Scholar] [CrossRef]
- Neudecker, V.; Brodsky, K.S.; Clambey, E.T.; Schmidt, E.P.; Packard, T.A.; Davenport, B.; Standiford, T.J.; Weng, T.; Fletcher, A.A.; Barthel, L.; et al. Neutrophil transfer of miR-223 to lung epithelial cells dampens acute lung injury in mice. Sci. Transl. Med. 2017, 9, eaah5360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef]
- Fox, S.E.; Li, G.; Akmatbekov, A.; Harbert, J.L.; Lameira, F.S.; Brown, J.Q.; Vander Heide, R.S. Unexpected Features of Cardiac Pathology in COVID-19 Infection. Circulation 2020, 142, 1123–1125. [Google Scholar] [CrossRef]
- Bradley, B.T.; Maioli, H.; Johnston, R.; Chaudhry, I.; Fink, S.L.; Xu, H.; Najafian, B.; Deutsch, G.; Lacy, J.M.; Williams, T.; et al. Histopathology and ultrastructural findings of fatal COVID-19 infections in Washington State: A case series. Lancet 2020, 396, 320–332. [Google Scholar] [CrossRef]
- Wu, C.; Hu, X.; Song, J.; Du, C.; Xu, J.; Yang, D.; Chen, D.; Zhong, M.; Jiang, J.; Xiong, W.; et al. Heart injury signs are associated with higher and earlier mortality in coronavirus disease 2019 (COVID-19). medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Libby, P. The Heart in COVID-19: Primary Target or Secondary Bystander? JACC Basic Transl. Sci. 2020, 5, 537–542. [Google Scholar] [CrossRef] [PubMed]
- Rapkiewicz, A.V.; Mai, X.; Carsons, S.E.; Pittaluga, S.; Kleiner, D.E.; Berger, J.S.; Thomas, S.; Adler, N.M.; Charytan, D.M.; Gasmi, B.; et al. Megakaryocytes and platelet-fibrin thrombi characterize multi-organ thrombosis at autopsy in COVID-19: A case series. EClinicalMedicine 2020, 24, 100434. [Google Scholar] [CrossRef]
- Long, B.; Brady, W.J.; Koyfman, A.; Gottlieb, M. Cardiovascular complications in COVID-19. Am. J. Emerg. Med. 2020, 38, 1504–1507. [Google Scholar] [CrossRef]
- Wiklander, O.P.B.; Brennan, M.Á.; Lötvall, J.; Breakefield, X.O.; Andaloussi, S.E. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, 11. [Google Scholar] [CrossRef] [PubMed]
- 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.; et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arslan, F.; Lai, R.C.; Smeets, M.B.; Akeroyd, L.; Choo, A.; Aguor, E.N.E.; Timmers, L.; van Rijen, H.V.; Doevendans, P.A.; Pasterkamp, G.; et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013, 10, 301–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teng, X.; Chen, L.; Chen, W.; Yang, J.; Yang, Z.; Shen, Z. Mesenchymal Stem Cell-Derived Exosomes Improve the Microenvironment of Infarcted Myocardium Contributing to Angiogenesis and Anti-Inflammation. Cell. Physiol. Biochem. 2015, 37, 2415–2424. [Google Scholar] [CrossRef]
- Bian, S.; Zhang, L.; Duan, L.; Wang, X.; Min, Y.; Yu, H. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J. Mol. Med. 2014, 92, 387–397. [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. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Zhao, Y.; Sun, X.; Cao, W.; Ma, J.; Sun, L.; Qian, H.; Zhu, W.; Xu, W. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Relieve Acute Myocardial Ischemic Injury. Stem Cells Int. 2015, 2015, 761643. [Google Scholar] [CrossRef]
- Cheng, Y.; Luo, R.; Wang, K.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Yao, Y.; Ge, S.; Xu, G. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. 2020, 97, 829–838. [Google Scholar] [CrossRef]
- Naicker, S.; Yang, C.-W.; Hwang, S.-J.; Liu, B.-C.; Chen, J.-H.; Jha, V. The Novel Coronavirus 2019 epidemic and kidneys. Kidney Int. 2020, 97, 824–828. [Google Scholar] [CrossRef]
- Diao, B.; Wang, C.; Wang, R.; Feng, Z.; Tan, Y.; Wang, H.; Wang, C.; Liu, L.; Liu, Y.; Liu, Y.; et al. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nat. Commun. 2021, 12, 2506. [Google Scholar] [CrossRef]
- Pitt, J.M.; Kroemer, G.; Zitvogel, L. Extracellular Vesicles: Masters of Intercellular Communication and Potential Clinical Interventions. J. Clin. Investig. 2021, 126, 1139–1143. [Google Scholar] [CrossRef] [Green Version]
- Camussi, G.; Deregibus, M.C.; Bruno, S.; Cantaluppi, V.; Biancone, L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010, 78, 838–848. [Google Scholar] [CrossRef] [Green Version]
- Aghajani Nargesi, A.; Lerman, L.O.; Eirin, A. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Kidney Repair: Current Status and Looming Challenges. Stem Cell Res. Ther. 2017, 8. [Google Scholar] [CrossRef]
- Tsuji, K.; Kitamura, S.; Wada, J. Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Renal Diseases. Int. J. Mol. Sci. 2020, 21, 756. [Google Scholar] [CrossRef] [Green Version]
- Lv, L.; Wu, W.; Feng, Y.; Li, Z.; Tang, T.; Liu, B. Therapeutic application of extracellular vesicles in kidney disease: Promises and challenges. J. Cell. Mol. Med. 2018, 22, 728–737. [Google Scholar] [CrossRef] [PubMed]
- Lv, L.L.; Tang, P.M.-K.; Li, C.J.; You, Y.K.; Li, J.; Huang, X.-R.; Ni, J.; Feng, M.; Liu, B.C.; Lan, H.-Y. The pattern recognition receptor, Mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation. Kidney Int. 2017, 91, 587–602. [Google Scholar] [CrossRef] [PubMed]
- Meng, X.-M.; Tang, P.M.-K.; Li, J.; Lan, H.Y. Macrophage Phenotype in Kidney Injury and Repair. Kidney Dis. 2015, 1, 138–146. [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, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phipps, M.M.; Barraza, L.H.; LaSota, E.D.; Sobieszczyk, M.E.; Pereira, M.R.; Zheng, E.X.; Fox, A.N.; Zucker, J.; Verna, E.C. Acute Liver Injury in COVID-19: Prevalence and Association with Clinical Outcomes in a Large U.S. Cohort. Hepatology 2020, 72, 807–817. [Google Scholar] [CrossRef]
- Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
- Chai, X.; Hu, L.; Zhang, Y.; Han, W.; Lu, Z.; Ke, A.; Zhou, J.; Shi, G.; Fang, N.; Fan, J.; et al. Specific ACE2 Expression in Cholangiocytes May Cause Liver Damage After 2019-nCoV Infection. biorxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Robinson, M.W.; Harmon, C.; O’Farrelly, C. Liver Immunology and Its Role in Inflammation and Homeostasis. Cell Mol. Immunol. 2016, 13, 267–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chand, N.; Sanyal, A.J. Sepsis-induced cholestasis. Hepatology 2007, 45, 230–241. [Google Scholar] [CrossRef] [PubMed]
- Fix, O.K.; Hameed, B.; Fontana, R.J.; Kwok, R.M.; McGuire, B.M.; Mulligan, D.C.; Pratt, D.S.; Russo, M.W.; Schilsky, M.L.; Verna, E.C.; et al. Clinical Best Practice Advice for Hepatology and Liver Transplant Providers During the COVID-19 Pandemic: AASLD Expert Panel Consensus Statement. Hepatology 2020, 72, 287–304. [Google Scholar] [CrossRef] [PubMed]
- Tan, C.Y.; Lai, R.C.; Wong, W.; Dan, Y.Y.; Lim, S.-K.; Ho, H.K. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Res. Ther. 2014, 5, 76. [Google Scholar] [CrossRef] [Green Version]
- Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W.; et al. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Liver Fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiore, E.; Domínguez, L.M.; Bayo, J.; Malvicini, M.; Atorrasagasti, C.; Rodriguez, M.; Cantero, M.J.; García, M.; Yannarelli, G.; Mazzolini, G. Human umbilical cord perivascular cells-derived extracellular vesicles mediate the transfer of IGF-I to the liver and ameliorate hepatic fibrogenesis in mice. Gene Ther. 2020, 27, 62–73. [Google Scholar] [CrossRef] [PubMed]
- Ohara, M.; Ohnishi, S.; Hosono, H.; Yamamoto, K.; Yuyama, K.; Nakamura, H.; Fu, Q.; Maehara, O.; Suda, G.; Sakamoto, N. Extracellular Vesicles from Amnion-Derived Mesenchymal Stem Cells Ameliorate Hepatic Inflammation and Fibrosis in Rats. Stem Cells Int. 2018, 2018, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Lou, G.; Chen, Z.; Zheng, M.; Liu, Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp. Mol. Med. 2017, 49, e346. [Google Scholar] [CrossRef]
- Herrera, M.B.; Fonsato, V.; Gatti, S.; Deregibus, M.C.; Sordi, A.; Cantarella, D.; Calogero, R.; Bussolati, B.; Tetta, C.; Camussi, G. Human liver stem cell-derived microvesicles accelerate hepatic regeneration in hepatectomized rats. J. Cell. Mol. Med. 2010, 14, 1605–1618. [Google Scholar] [CrossRef] [Green Version]
- Caress, J.B.; Castoro, R.J.; Simmons, Z.; Scelsa, S.N.; Lewis, R.A.; Ahlawat, A.; Narayanaswami, P. COVID-19-associated Guillain-Barré syndrome: The early pandemic experience. Muscle Nerve 2020, 62, 485–491. [Google Scholar] [CrossRef]
- Paniz-Mondolfi, A.; Bryce, C.; Grimes, Z.; Gordon, R.E.; Reidy, J.; Lednicky, J.; Sordillo, E.M.; Fowkes, M. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). J. Med. Virol. 2020, 92, 699–702. [Google Scholar] [CrossRef] [Green Version]
- Lersy, F.; Benotmane, I.; Helms, J.; Collange, O.; Schenck, M.; Brisset, J.-C.; Chammas, A.; Willaume, T.; Lefebvre, N.; Solis, M.; et al. Cerebrospinal Fluid Features in Patients With Coronavirus Disease 2019 and Neurological Manifestations: Correlation with Brain Magnetic Resonance Imaging Findings in 58 Patients. J. Infect. Dis. 2021, 223, 600–609. [Google Scholar] [CrossRef]
- Matschke, J.; Lütgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schröder, A.S.; Edler, C.; Mushumba, H.; Fitzek, A.; Allweiss, L.; Dandri, M.; et al. Neuropathology of patients with COVID-19 in Germany: A post-mortem case series. Lancet Neurol. 2020, 19, 919–929. [Google Scholar] [CrossRef]
- Zubair, A.S.; McAlpine, L.S.; Gardin, T.; Farhadian, S.; Kuruvilla, D.E.; Spudich, S. Neuropathogenesis and Neurologic Manifestations of the Coronaviruses in the Age of Coronavirus Disease 2019 A Review. JAMA Neurol. 2020, 77, 1018–1027. [Google Scholar] [CrossRef] [PubMed]
- Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
- Nascimento Conde, J.; Schutt, W.R.; Gorbunova, E.E.; Mackow, E.R. Recombinant ACE2 Expression Is Required for SARS-CoV-2 To Infect Primary Human Endothelial Cells and Induce Inflammatory and Procoagulative Responses. mBio 2020, 11, e03185-20. [Google Scholar] [CrossRef] [PubMed]
- Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Prado, A.V.; Skriabine, S.; Lu, P.; Weizman, O.-E.; Liu, F.; Dai, Y.; et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. bioRxiv 2020. [Google Scholar] [CrossRef]
- Perrin, P.; Collongues, N.; Baloglu, S.; Bedo, D.; Bassand, X.; Lavaux, T.; Gautier-Vargas, G.; Keller, N.; Kremer, S.; Fafi-Kremer, S.; et al. Cytokine release syndrome-associated encephalopathy in patients with COVID-19. Eur. J. Neurol. 2021, 28, 248–258. [Google Scholar] [CrossRef]
- Pilotto, A.; Masciocchi, S.; Volonghi, I.; De Giuli, V.; Caprioli, F.; Mariotto, S.; Ferrari, S.; Bozzetti, S.; Imarisio, A.; Risi, B.; et al. SARS-CoV-2 encephalitis is a cytokine release syndrome: Evidences from cerebrospinal fluid analyses. Clin. Infect. Dis 2021. [Google Scholar] [CrossRef] [PubMed]
- Erickson, M.A.; Wilson, M.L.; Banks, W.A. In vitro modeling of blood–brain barrier and interface functions in neuroimmune communication. Fluids Barriers CNS 2020, 17, 26. [Google Scholar] [CrossRef] [PubMed]
- Hsuchou, H.; Kastin, A.J.; Mishra, P.K.; Pan, W. C-reactive protein increases BBB permeability: Implications for obesity and neuroinflammation. Cell. Physiol. Biochem. 2012, 30, 1109–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erickson, M.A.; Banks, W.A. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol. Rev. 2018, 70, 278–314. [Google Scholar] [CrossRef]
- Banks, W.A.; Freed, E.O.; Wolf, K.M.; Robinson, S.M.; Franko, M.; Kumar, V.B. Transport of human immunodeficiency virus type 1 pseudoviruses across the blood-brain barrier: Role of envelope proteins and adsorptive endocytosis. J. Virol. 2001, 75, 4681–4691. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Li, M.; Wang, M.; Zhou, Y.; Chang, J.; Xian, Y.; Wang, D.; Mao, L.; Jin, H.; Hu, B. Acute cerebrovascular disease following COVID-19: A single center, retrospective, observational study. Stroke Vasc. Neurol. 2020, 5, 279–284. [Google Scholar] [CrossRef]
- Jin, Y.; Ji, W.; Yang, H.; Chen, S.; Zhang, W.; Duan, G. Endothelial activation and dysfunction in COVID-19: From basic mechanisms to potential therapeutic approaches. Signal. Transduct. Target. Ther. 2020, 5, 1–13. [Google Scholar] [CrossRef]
- Andjus, P.; Kosanović, M.; Milićević, K.; Gautam, M.; Vainio, S.J.; Jagečić, D.; Kozlova, E.N.; Pivoriūnas, A.; Chachques, J.-C.; Sakaj, M.; et al. Extracellular Vesicles as Innovative Tool for Diagnosis, Regeneration and Protection against Neurological Damage. Int. J. Mol. Sci. 2020, 21, 6859. [Google Scholar] [CrossRef]
- Jin, Q.; Wu, P.; Zhou, X.; Qian, H.; Xu, W. Extracellular Vesicles: Novel Roles in Neurological Disorders. Stem Cells Int. 2021, 2021, e6640836. [Google Scholar] [CrossRef]
- Huang, J.-H.; Yin, X.-M.; Xu, Y.; Xu, C.-C.; Lin, X.; Ye, F.-B.; Cao, Y.; Lin, F.-Y. Systemic Administration of Exosomes Released from Mesenchymal Stromal Cells Attenuates Apoptosis, Inflammation, and Promotes Angiogenesis after Spinal Cord Injury in Rats. J. Neurotrauma 2017, 34, 3388–3396. [Google Scholar] [CrossRef]
- Li, C.; Jiao, G.; Wu, W.; Wang, H.; Ren, S.; Zhang, L.; Zhou, H.; Liu, H.; Chen, Y. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/β-catenin Signaling Pathway. Cell Transplant. 2019, 28, 1373–1383. [Google Scholar] [CrossRef] [PubMed]
- Lambert, C.; Cisternas, P.; Inestrosa, N.C. Role of Wnt Signaling in Central Nervous System Injury. Mol. Neurobiol. 2016, 53, 2297–2311. [Google Scholar] [CrossRef] [PubMed]
- Xian, P.; Hei, Y.; Wang, R.; Wang, T.; Yang, J.; Li, J.; Di, Z.; Liu, Z.; Baskys, A.; Liu, W.; et al. Mesenchymal stem cell-derived exosomes as a nanotherapeutic agent for amelioration of inflammation-induced astrocyte alterations in mice. Theranostics 2019, 9, 5956–5975. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.-B.; Yang, H.; Manaenko, A.; Lu, J.; Mei, Q.; Hu, Q. Potential of Exosomes for the Treatment of Stroke. Cell Transplant. 2019, 28, 662–670. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Cheng, Q.; Hu, G.; Deng, T.; Wang, Q.; Zhou, J.; Su, X. Extracellular vesicles in mesenchymal stromal cells: A novel therapeutic strategy for stroke. Exp. Ther. Med. 2018, 15, 4067–4079. [Google Scholar] [CrossRef]
- Araya, S.; Wordofa, M.; Mamo, M.A.; Tsegay, Y.G.; Hordofa, A.; Negesso, A.E.; Fasil, T.; Berhanu, B.; Begashaw, H.; Atlaw, A.; et al. The Magnitude of Hematological Abnormalities Among COVID-19 Patients in Addis Ababa, Ethiopia. JMDH 2021, 14, 545–554. [Google Scholar] [CrossRef]
- Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020, 12, 8. [Google Scholar] [CrossRef]
- Unsinger, J.; McDonough, J.S.; Shultz, L.D.; Ferguson, T.A.; Hotchkiss, R.S. Sepsis-induced human lymphocyte apoptosis and cytokine production in “humanized” mice. J. Leukoc. Biol. 2009, 86, 219–227. [Google Scholar] [CrossRef] [Green Version]
- Lefrançais, E.; Ortiz-Muñoz, G.; Caudrillier, A.; Mallavia, B.; Liu, F.; Sayah, D.M.; Thornton, E.E.; Headley, M.B.; David, T.; Coughlin, S.R.; et al. The lung is a site of platelet biogenesis and a reservoir for hematopoietic progenitors. Nature 2017, 544, 105–109. [Google Scholar] [CrossRef]
- Liu, X.; Zhang, R.; He, G. Hematological findings in coronavirus disease 2019: Indications of progression of disease. Ann. Hematol. 2020, 99, 1421–1428. [Google Scholar] [CrossRef]
- The Lancet Haematology. COVID-19 coagulopathy: An evolving story. Lancet Haematol. 2020, 7, e425.
- Escher, R.; Breakey, N.; Lämmle, B. Severe COVID-19 infection associated with endothelial activation. Thromb. Res. 2020, 190, 62. [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]
- Ware, L.B.; Matthay, M.A. The acute respiratory distress syndrome. N. Engl. J. Med. 2000, 342, 1334–1349. [Google Scholar] [CrossRef] [PubMed]
- Phillipson, M.; Kubes, P. The neutrophil in vascular inflammation. Nat. Med. 2011, 17, 1381–1390. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Morel, O.; Toti, F.; Morel, N.; Freyssinet, J.-M. Microparticles in endothelial cell and vascular homeostasis: Are they really noxious? Haematologica 2009, 94, 313–317. [Google Scholar] [CrossRef] [PubMed]
- Lacroix, R.; Sabatier, F.; Mialhe, A.; Basire, A.; Pannell, R.; Borghi, H.; Robert, S.; Lamy, E.; Plawinski, L.; Camoin-Jau, L.; et al. Activation of plasminogen into plasmin at the surface of endothelial microparticles: A mechanism that modulates angiogenic properties of endothelial progenitor cells in vitro. Blood 2007, 110, 2432–2439. [Google Scholar] [CrossRef] [PubMed]
- Gholizadeh-Ghaleh Aziz, S.; Alipour, S.; Ranjbarvan, P.; Azari, A.; Babaei, G.; Golchin, A. Critical Roles of TLRs on the Polarization of Mesenchymal Stem Cells for Cell Therapy of Viral Infections: A Notice for COVID-19 Treatment. Comp. Clin. Pathol. 2021, 1–10. [Google Scholar] [CrossRef]
- Raicevic, G.; Najar, M.; Stamatopoulos, B.; De Bruyn, C.; Meuleman, N.; Bron, D.; Toungouz, M.; Lagneaux, L. The source of human mesenchymal stromal cells influences their TLR profile as well as their functional properties. Cell. Immunol. 2011, 270, 207–216. [Google Scholar] [CrossRef]
- Cruz, T.; Rojas, M. Preclinical Evidence for the Role of Stem/Stromal Cells in Targeting ARDS. In Stem Cell-Based Therapy for Lung Disease; Burgess, J.K., Heijink, I.H., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 199–217. ISBN 978-3-030-29403-8. [Google Scholar]
- Zanoni, M.; Cortesi, M.; Zamagni, A.; Tesei, A. The Role of Mesenchymal Stem Cells in Radiation-Induced Lung Fibrosis. Int. J. Mol. Sci. 2019, 20, 3876. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, Y.; Tsuchiya, A.; Seino, S.; Kawata, Y.; Kojima, Y.; Ikarashi, S.; Starkey Lewis, P.J.; Lu, W.-Y.; Kikuta, J.; Kawai, H.; et al. Mesenchymal Stem Cells and Induced Bone Marrow-Derived Macrophages Synergistically Improve Liver Fibrosis in Mice. Stem Cells Transl. Med. 2019, 8, 271–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kojima, Y.; Tsuchiya, A.; Ogawa, M.; Nojiri, S.; Takeuchi, S.; Watanabe, T.; Nakajima, K.; Hara, Y.; Yamashita, J.; Kikuta, J.; et al. Mesenchymal stem cells cultured under hypoxic conditions had a greater therapeutic effect on mice with liver cirrhosis compared to those cultured under normal oxygen conditions. Regen. Ther. 2019, 11, 269–281. [Google Scholar] [CrossRef]
- Ikarashi, S.; Tsuchiya, A.; Kawata, Y.; Kojima, Y.; Watanabe, T.; Takeuchi, S.; Igarashi, K.; Ideta-Otsuka, M.; Oki, K.; Takamura, M.; et al. Effects of Human Adipose Tissue-Derived and Umbilical Cord Tissue-Derived Mesenchymal Stem Cells in a Dextran Sulfate Sodium-Induced Mouse Model. BioRes. Open Access 2019, 8, 185–199. [Google Scholar] [CrossRef] [PubMed]
- Kawata, Y.; Tsuchiya, A.; Seino, S.; Watanabe, Y.; Kojima, Y.; Ikarashi, S.; Tominaga, K.; Yokoyama, J.; Yamagiwa, S.; Terai, S. Early injection of human adipose tissue-derived mesenchymal stem cell after inflammation ameliorates dextran sulfate sodium-induced colitis in mice through the induction of M2 macrophages and regulatory T cells. Cell Tissue Res. 2019, 376, 257–271. [Google Scholar] [CrossRef]
- Yáñez-Mó, M.; Siljander, P.R.-M.; 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] [Green Version]
- Kim, D.; Nishida, H.; An, S.Y.; Shetty, A.K.; Bartosh, T.J.; Prockop, D.J. Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc. Natl. Acad. Sci. USA 2016, 113, 170–175. [Google Scholar] [CrossRef] [Green Version]
- Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [Green Version]
- Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Feng, X.; Abbott, J.; Fang, X.; Hao, Q.; Monsel, A.; Qu, J.; Matthay, M.A.; Lee, J.W. Human Mesenchymal Stem Cell Microvesicles for Treatment of E.coli Endotoxin-Induced Acute Lung Injury in Mice. Stem Cells 2014, 32, 116–125. [Google Scholar] [CrossRef] [Green Version]
- Xia, X.; Wang, Y.; Huang, Y.; Zhang, H.; Lu, H.; Zheng, J.C. Exosomal miRNAs in central nervous system diseases: Biomarkers, pathological mediators, protective factors and therapeutic agents. Prog. Neurobiol. 2019, 183, 101694. [Google Scholar] [CrossRef] [PubMed]
- Ti, D.; Hao, H.; Tong, C.; Liu, J.; Dong, L.; Zheng, J.; Zhao, Y.; Liu, H.; Fu, X.; Han, W. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J. Transl. Med. 2015, 13, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Li, J.W.; Wei, L.; Han, Z.; Chen, Z. Mesenchymal Stromal Cells-Derived Exosomes Alleviate Ischemia/Reperfusion Injury in Mouse Lung by Transporting Anti-Apoptotic MiR-21-5p. Eur. J. Pharmacol. 2019, 852, 68–76. [Google Scholar] [CrossRef]
- Hao, Q.; Gudapati, V.; Monsel, A.; Park, J.H.; Hu, S.; Kato, H.; Lee, J.H.; Zhou, L.; He, H.; Lee, J.W. Mesenchymal Stem Cell-Derived Extracellular Vesicles Decrease Lung Injury in Mice. J. Immunol. 2019, 203, 1961–1972. [Google Scholar] [CrossRef]
- Iqbal Yatoo, M.; Hamid, Z.; Rather, I.; Nazir, Q.U.A.; Bhat, R.A.; Ul Haq, A.; Magray, S.N.; Haq, Z.; Sah, R.; Tiwari, R.; et al. Immunotherapies and Immunomodulatory Approaches in Clinical Trials—A Mini Review. Hum. Vaccin. Immunother. 2020, 17, 1897–1909. [Google Scholar] [CrossRef]
- Tao, S.-C.; Guo, S.-C.; Zhang, C.-Q. Platelet-derived Extracellular Vesicles: An Emerging Therapeutic Approach. Int. J. Biol. Sci. 2017, 13, 828–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torreggiani, E.; Perut, F.; Roncuzzi, L.; Zini, N.; Baglìo, S.R.; Baldini, N. Exosomes: Novel effectors of human platelet lysate activity. Eur. Cell. Mater. 2014, 28, 137–151; discussion 151. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.-C.; Tao, S.-C.; Yin, W.-J.; Qi, X.; Yuan, T.; Zhang, C.-Q. Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model. Theranostics 2017, 7, 81–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Q.; Fan, Q.; Xu, J.; Bai, J.; Han, X.; Dong, Z.; Zhou, X.; Liu, Z.; Gu, Z.; Wang, C. Calming Cytokine Storm in Pneumonia by Targeted Delivery of TPCA-1 Using Platelet-Derived Extracellular Vesicles. Matter 2020, 3, 287–301. [Google Scholar] [CrossRef] [PubMed]
- Yan, Y.; Zhou, W.; Wang, Y.; Guo, Q.; Zhao, F.; Zhu, Z.; Xing, Y.; Zhang, H.; Aljofan, M.; Jarrahi, A.M.; et al. The Potential Role of Extracellular Vesicles in COVID-19 Treatment: Opportunity and Challenge. Front Mol. Biosci. 2021, 8, 699929. [Google Scholar] [CrossRef]
- Varon, D.; Shai, E. Role of Platelet-Derived Microparticles in Angiogenesis and Tumor Progression. Discov. Med. 2009, 8, 237–241. [Google Scholar] [PubMed]
- Hayon, Y.; Dashevsky, O.; Shai, E.; Brill, A.; Varon, D.; Leker, R.R. Platelet Microparticles Induce Angiogenesis and Neurogenesis after Cerebral Ischemia. Curr. Neurovasc. Res. 2012, 9, 185–192. [Google Scholar] [CrossRef] [PubMed]
- Hayon, Y.; Dashevsky, O.; Shai, E.; Varon, D.; Leker, R.R. Platelet Microparticles Promote Neural Stem Cell Proliferation, Survival and Differentiation. J. Mol. Neurosci. 2012, 47, 659–665. [Google Scholar] [CrossRef] [PubMed]
- Hayon, Y.; Shai, E.; Varon, D.; Leker, R.R. The Role of Platelets and Their Microparticles in Rehabilitation of Ischemic Brain Tissue. CNS Neurol. Disord. Drug Targets 2012, 11, 921–925. [Google Scholar] [CrossRef]
- Sabanovic, B.; Piva, F.; Cecati, M.; Giulietti, M. Promising Extracellular Vesicle-Based Vaccines against Viruses, Including SARS-CoV-2. Biology 2021, 10, 94. [Google Scholar] [CrossRef]
- Kaur, S.P.; Gupta, V. COVID-19 Vaccine: A Comprehensive Status Report. Virus Res. 2020, 288, 198114. [Google Scholar] [CrossRef]
- Jeyanathan, M.; Afkhami, S.; Smaill, F.; Miller, M.S.; Lichty, B.D.; Xing, Z. Immunological considerations for COVID-19 vaccine strategies. Nat. Rev. Immunol. 2020, 20, 615–632. [Google Scholar] [CrossRef] [PubMed]
- E Pecq, J.-B. Dexosomes as a Therapeutic Cancer Vaccine: From Bench to Bedside. Blood Cells Mol. Dis. 2005, 35, 129–135. [Google Scholar] [CrossRef] [PubMed]
- Tan, A.; De La Peña, H.; Seifalian, A.M. The Application of Exosomes as a Nanoscale Cancer Vaccine. Int. J. Nanomed. 2010, 5, 889–900. [Google Scholar] [CrossRef] [Green Version]
- Kuate, S.; Cinatl, J.; Doerr, H.W.; Überla, K. Exosomal vaccines containing the S protein of the SARS coronavirus induce high levels of neutralizing antibodies. Virology 2007, 362, 26. [Google Scholar] [CrossRef] [Green Version]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef]
- Polak, K.; Greze, N.; Lachat, M.; Merle, D.; Chiumento, S.; Bertrand-Gaday, C.; Trentin, B.; Mamoun, R.Z. Extracellular Vesicle-Based Vaccine Platform Displaying Native Viral Envelope Proteins Elicits a Robust Anti-SARS-CoV-2 Response in Mice. bioRxiv 2020. [Google Scholar] [CrossRef]
- Kowalewski, M.; Fina, D.; Słomka, A.; Raffa, G.M.; Martucci, G.; Lo Coco, V.; De Piero, M.E.; Ranucci, M.; Suwalski, P.; Lorusso, R. COVID-19 and ECMO: The Interplay between Coagulation and Inflammation—A Narrative Review. Crit. Care 2020, 24. [Google Scholar] [CrossRef]
- Millar, J.E.; von Bahr, V.; Malfertheiner, M.V.; Ki, K.K.; Redd, M.A.; Bartnikowski, N.; Suen, J.Y.; McAuley, D.F.; Fraser, J.F. Administration of mesenchymal stem cells during ECMO results in a rapid decline in oxygenator performance. Thorax 2019, 74, 194–196. [Google Scholar] [CrossRef] [Green Version]
- De Jong, B.; Barros, E.R.; Hoenderop, J.G.J.; Rigalli, J.P. Recent Advances in Extracellular Vesicles as Drug Delivery Systems and Their Potential in Precision Medicine. Pharmaceutics 2020, 12, 1006. [Google Scholar] [CrossRef]
- Duong, N.; Curley, K.; Brown, A.; Campanelli, A.; Do, M.A.; Levy, D.; Tantry, A.; Marriott, G.; Lu, B. Decoy exosomes as a novel biologic reagent to antagonize inflammation. Int. J. Nanomed. 2019, 14, 3413–3425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Lee, H.; Wang, X.; Groot, M.; Sharma, L.; Cruz, C.S.D.; Jin, Y. A potential role of microvesicle-containing miR-223/142 in lung inflammation. Thorax 2019, 74, 865. [Google Scholar] [CrossRef] [PubMed]
- Zhang, D.; Lee, H.; Wang, X.; Rai, A.; Groot, M.; Jin, Y. Exosome-Mediated Small RNA Delivery: A Novel Therapeutic Approach for Inflammatory Lung Responses. Mol. Ther. 2018, 26, 2119–2130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, S.; Wang, Y.; Xia, X.; Zheng, J.C. Exosome Engineering: Current Progress in Cargo Loading and Targeted Delivery. NanoImpact 2020, 20, 100261. [Google Scholar] [CrossRef]
- 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]
- Harcourt, J.; Tamin, A.; Lu, X.; Kamili, S.; Sakthivel, S.K.; Wang, L.; Murray, J.; Queen, K.; Lynch, B.; Whitaker, B.; et al. Isolation and characterization of SARS-CoV-2 from the first US COVID-19 patient. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Sharma, D.; Zhao, F. Updates on clinical trials evaluating the regenerative potential of allogenic mesenchymal stem cells in COVID-19. Npj Regen. Med. 2021, 6, 1–11. [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]
- Lim, S.K.; Giebe, B.; Weiss, D.J.; Witwer, K.W.; Rohde, E. Re: Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19’’ by Sengupta et al. Stem Cells Dev. 2020, 29, 877–878. [Google Scholar] [CrossRef]
- Scarfe, L.; Taylor, A.; Sharkey, J.; Harwood, R.; Barrow, M.; Comenge, J.; Beeken, L.; Astley, C.; Santeramo, I.; Hutchinson, C.; et al. Non-Invasive Imaging Reveals Conditions That Impact Distribution and Persistence of Cells after in Vivo Administration. Stem Cell Res. Ther. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
- Pinky; Gupta, S.; Krishnakumar, V.; Sharma, Y.; Dinda, A.K.; Mohanty, S. Mesenchymal Stem Cell Derived Exosomes: A Nano Platform for Therapeutics and Drug Delivery in Combating COVID-19. Stem Cell Rev. Rep. 2021, 17, 33–43. [Google Scholar] [CrossRef]
- Plava, J.; Cihova, M.; Burikova, M.; Matuskova, M.; Kucerova, L.; Miklikova, S. Recent Advances in Understanding Tumor Stroma-Mediated Chemoresistance in Breast Cancer. Mol. Cancer 2019, 18. [Google Scholar] [CrossRef]
- Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C.; et al. Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 2016, 76, 1770–1780. [Google Scholar] [CrossRef] [Green Version]
- Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and Challenges of Extracellular Vesicle-Based Drug Delivery System: Considering Cell Source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gimona, M.; Brizzi, M.F.; Choo, A.B.H.; Dominici, M.; Davidson, S.M.; Grillari, J.; Hermann, D.M.; Hill, A.F.; de Kleijn, D.; Lai, R.C.; et al. Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles. Cytotherapy 2021, 23, 373–380. [Google Scholar] [CrossRef] [PubMed]
- Bustos, M.L.; Huleihel, L.; Kapetanaki, M.G.; Lino-Cardenas, C.L.; Mroz, L.; Ellis, B.M.; McVerry, B.J.; Richards, T.J.; Kaminski, N.; Cerdenes, N.; et al. Aging Mesenchymal Stem Cells Fail to Protect Because of Impaired Migration and Antiinflammatory Response. Am. J. Respir. Crit. Care Med. 2014, 189, 787–798. [Google Scholar] [CrossRef] [PubMed]
- Kurian, T.K.; Banik, S.; Gopal, D.; Chakrabarti, S.; Mazumder, N. Elucidating Methods for Isolation and Quantification of Exosomes: A Review. Mol. Biotechnol. 2021, 63, 249–266. [Google Scholar] [CrossRef] [PubMed]
Study Identifier | Disease Condition | Source of EVs | Study Phase Study Type Enrolled Patients | Route of Administration | Recruitment Status | Country/Year |
---|---|---|---|---|---|---|
NCT04602442 |
| MSC | Phase II Interventional 90 | Inhalation | Enrolling by invitation | Russia/ 2020 |
NCT04798716 |
| MSCs | Phase I and II Interventional 55 | Intravenous | Not yet recruited | USA/ 2021 |
NCT04747574 |
| Drug: Exosomes-CD24 | Phase Interventional 35 | Intravenous | Recruiting | Israel/ 2021 |
NCT04491240 |
| Drug Exosomes | Phase I and II Interventional 30 | Inhalation | Completed | Russia/ 2020 |
NCT04389385 |
| T-Cells | Phase I Interventional 60 | Intravenous | Recruiting | Turkey/ 2020 |
NCT04276987 |
| Adipose tissue MSCs | Phase I Interventional 24 | Inhalation | Completed | China/ 2020 |
NCT04384445 |
| Biological Zofin from human Amniotic fluids | Phase I and II Interventional 20 | Intravenous | Recruiting | USA/ 2021 |
NCT04657406 |
| Drug Zofin TM from human amniotic fluid (HAF) | Phase –not available Interventional Not available | Intravenous | Available | Not available/ 2020 |
NCT04493242 |
| Bone marrow | Phase II 60 | Intravenous | No yet recruiting | US / 2021 |
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
Karn, V.; Ahmed, S.; Tsai, L.-W.; Dubey, R.; Ojha, S.; Singh, H.N.; Kumar, M.; Gupta, P.K.; Sadhu, S.; Jha, N.K.; et al. Extracellular Vesicle-Based Therapy for COVID-19: Promises, Challenges and Future Prospects. Biomedicines 2021, 9, 1373. https://doi.org/10.3390/biomedicines9101373
Karn V, Ahmed S, Tsai L-W, Dubey R, Ojha S, Singh HN, Kumar M, Gupta PK, Sadhu S, Jha NK, et al. Extracellular Vesicle-Based Therapy for COVID-19: Promises, Challenges and Future Prospects. Biomedicines. 2021; 9(10):1373. https://doi.org/10.3390/biomedicines9101373
Chicago/Turabian StyleKarn, Vamika, Shaista Ahmed, Lung-Wen Tsai, Rajni Dubey, Shreesh Ojha, Himanshu Naryan Singh, Mukesh Kumar, Piyush Kumar Gupta, Soumi Sadhu, Niraj Kumar Jha, and et al. 2021. "Extracellular Vesicle-Based Therapy for COVID-19: Promises, Challenges and Future Prospects" Biomedicines 9, no. 10: 1373. https://doi.org/10.3390/biomedicines9101373
APA StyleKarn, V., Ahmed, S., Tsai, L. -W., Dubey, R., Ojha, S., Singh, H. N., Kumar, M., Gupta, P. K., Sadhu, S., Jha, N. K., Kumar, A., Pandit, S., & Kumar, S. (2021). Extracellular Vesicle-Based Therapy for COVID-19: Promises, Challenges and Future Prospects. Biomedicines, 9(10), 1373. https://doi.org/10.3390/biomedicines9101373