MSC-Derived Exosomes for COVID-19 Treatment: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Alok Raghav.

Exosomes possess anti-inflammatory, pro-angiogenic, and immunomodulatory properties that can be explored in an effort to improve the outcomes of SARS-CoV-2-infected patients. 

  • exosomes
  • SARS-CoV-2
  • COVID-19
  • mesenchymal stem cells
  • anti-inflammatory
  • extracellular vesicles

1. Introduction

The novel coronavirus disease-2019 was also termed COVID-19 by the World Health Organization (WHO) on 11 February 2020 [1]. The International Virus Classification Commission (IVCC) announced the novel coronavirus as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) were also among the deadliest viral infections that have been previously identified [2]. COVID-19 is caused by SARS-CoV-2, and this disease became a devastating global pandemic within only a few months. Currently, COVID-19 cases have been reported in numerous countries throughout the world. According to the latest World Health Organization (WHO) data up to 19 January 2021, the number of confirmed cases is 93,956,883 with confirmed deaths of 2,029,084 individuals in 216 different countries [3]. The WHO also listed COVID-19 as a Public Health Emergency of International Concern (PHEIC). The relative ease of COVID-19 transmission and the mortality, severe illness, and disruption of socioeconomic structures caused by this disease make this illness a source of major concern to the global population. The World Trade Organization (WTO) announced that there was a decline in world trade of between 13% and 32% in 2020 due to the COVID-19 pandemic [4]. Due to the large amount of public interest in the development of new and effective drugs, and therapies for the treatment of COVID-19, a number of researchers are investigating and promoting stem-cell or exosome-based interventions that may ultimately prove useful for the treatment of COVID-19 or SARS-CoV-2. However, the use of stem cell- or exosome-based therapy has not been approved by the US Food and Drug Administration (FDA). A recently published study demonstrated that MSCs mediate the inflammatory response and can be effective for the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) [5]. The literature also suggests that MSCs can be effectively used for the treatment of critical COVID-19 cases [5]. The success of this type of treatment is based on the ability of MSC-derived exosomes to elicit a therapeutic response by balancing inflammation via the paracrine signaling pathway [5].

2. Pathological Features of COVID-19

The primary pathological features of SARS-CoV-2 caused by the COVID-19 pandemic are hypoxemia, chronic pulmonary inflammation and edema, and diffuse alveolar damage with exudates rich in cellular fibromyxoid and hyaline membrane architecture [6]. Previous studies have reported that these pathological changes are similar to those observed in response to ALI, ARDS, severe acute respiratory syndrome (SARS), and Middle Eastern respiratory syndrome (MERS) [6]. However, SARS-CoV-2 or COVID-19 is characterized by more severe inflammatory exudation, inflammatory cytokine storm, pulmonary edema, and milder pulmonary fibrosis [6]. Due to its high transmission properties and high sustainability in the environment, the disease spreads at an immensely rapid rate. Transmission is achieved through aerosols containing mucus and though fecal medium [1,7][1][7]. This virus can survive for up to 3 h in the air if an infected person sneezes without covering their face. Once the pathogen is inoculated into the respiratory system, it binds to alveoli in a manner similar to that of SARS-CoV [8,9][8][9]. The virus exhibits a very high affinity for type 2 pneumocytes within the alveoli. The main role of these type 2 pneumocytes is to release surfactants that reduce the surface tension of the alveoli sac and decrease the collapsing pressure, which is one of the key events in respiration. COVID-19 is further classified into different clinical stages based on the cell types that are likely to be infected [10].

3. Treatment of SARS-CoV-2 with Mesenchymal Stem Cells

Mesenchymal stem cells (MSCs) originate from the bone marrow, umbilical cord, placenta, and other tissues and possess differentiation and programming potential with strong immunomodulation and endogenous repair mechanisms. In regenerative medicine, adult MSCs are known to be protective against pulmonary, neurological, hepatic, renal, and cardiovascular diseases. MSCs are known to exert immunomodulatory effects that impact macrophages and promote polarization towards a therapeutic or reparative phenotype [24][11]. It was evident from a previously published study that CCL2 (a cytokine responsible for the recruitment of macrophages) derived from MSCs exhibited therapeutic effects by assisting macrophage repolarization [24][11]. MSCs act as immune modulators for both the innate and the adaptive immune systems. These cells are also involved in the switching of the phenotypes of inflammatory macrophages (M1) towards a reparative or therapeutic phenotype (M2), and they are related to decreased TNF-α and increased IL-10 production that is mediated by cellular contact [24][11].

Immune modulation of MSCs relies upon the activation of effector functions in immune cells. In a previously published study, MSCs were found to suppress lung infiltration and the dissolution of pulmonary edema. MSCs are multipotent, regenerative, and self-renewable and possess the ability to suppress the immune response and to differentiate into type II alveolar epithelial cells in vitro [25][12]. In a phase 2 clinical trial (NCT03608592) examining patients with ALI and ARDS, MSCs were demonstrated to possess anti-inflammatory activity [26][13]. Based on the outcome of this safety trial, MSCs may alleviate SARS-CoV-2-associated CS and ARDS and may also prove to be a prospective therapy for chronic respiratory dysfunction and lung fibrosis. MSCs are the prime therapeutic approach for treatment of lungs fibrosis, because they are easy to isolate, and have minimum immunogenicity, reparative characteristics along with anti-inflammatory properties. Clinical trials proved that MSCs possess short-term safety and tolerability with some limited studies focusing on adverse pro-inflammatory and myofibroblastic feature. The possible mechanism behind the protective effect of MSCs in lung fibrosis is that, MSCs migrate to the site of action (i.e., site of injury) and release multiple paracrine factors that are largely mediated by their extracellular vesicles that regulate the epithelial and endothelial permeability and cause decrease inflammation along with promotion of tissue repair. MSCs exclusively exhibit three characteristics in SARS-CoV-2-infected patients, including (i) activation of T cells to relieve excessive immune responses, (ii) maintenance of homeostasis in response to specific injuries associated with the lungs while also promoting regeneration, and (iii) inhibition of inflammation caused by the release of cytokines that is mediated by extracellular vesicles (EVs) or exosomes [27,28][14][15]. These exosomes deliver mRNA, microRNA, DNA, proteins, and metabolites to trigger reparative, therapeutic, and regenerative functions in the lungs [29][16].

4. Exosome: A Nanometer-Sized Envelope Possessing Miraculous Properties

Exosomes are nanometer-sized enveloped small extracellular vesicles that are extracted by ultracentrifugation of spent culture media of cells, and they are generated by the processes of the endocytosis pathway [30][17]. Almost all cell types release exosomes that contain varied biological information in the form of microRNAs (miRNAs), nucleic acids, proteins, growth factors, and EVs. In addition to facilitating cell-to-cell communication, exosomes are also helpful in determining the cell state by acting as biomarkers for diagnosis and prognosis [31][18]. Exosomes possess a small cup-shaped spherical morphology with sizes ranging from 30 to 100 nm in diameter. Exosomes also contain conserved proteins, including tetraspanins (CD9, CD63, and CD 81), MHC class I and II, and heat shock proteins (HSP 60, 70, and 90) [32][19]. Primary cells, hematopoietic cells, viral particles, and cancer cells found in biological fluids such as saliva, synovial fluid (SF), serum, plasma, bronchoalveolar lavage (BAL) fluid, urine, amniotic fluid (AF), pleural effusions (PE), semen, were all found in proximity to exosomes.

5. Biogenesis of Exosomes

Exosome biogenesis is a continuous cellular phenomenon that is initiated by inward invagination of the plasma membrane within the cytosol to form early and late endosomes. Late endosomes can further fuse together to form multivesicular bodies (MVBs) that invaginate to form intraluminal vesicles (ILVs). ILVs formed during the episodes of inward invagination are accompanied by several essential proteins, cytoskeletal components, nucleic acids, growth factors, necessary cellular components. After formation, these MVBs containing ILVs fuse together with the plasma membrane of the cells and are released as exosomes by exocytosis into the extracellular spaces. In previous studies, it was speculated that in the MVB biogenesis process, the budding of exosome cargos and their sorting is either endosomal sorting complex required for transport (ESCRT)-dependent or -independent [32][19]. Figure 1 demonstrates the mechanism of EVs biogenesis and its isolation.

It is believed that the ESCRT-mediated cargo-sorting phenomenon includes the identification and sequestration of ubiquitinated proteins to specific sites of the endosomal membranes [32][19]. Furthermore, the sequential association between subunits I, II, and III of ESCRT terminates the complex and initiates the budding process. It was revealed that the sorting protein Vps4 is involved in the detachment of the ESCRT III complex from the MVBs membrane that aids in the formation of ILVs from cleaved buds [32][19]. However, ESCRT-independent processes involve proteins and lipids such as tetraspanins (CD81) and ceramides. These exosomes associate post-release with recipient cells for efficient uptake. The exosome uptake mechanism involves both clathrin-dependent and -independent events, micropinocytosis, phagocytosis, and lipid raft-mediated internalization.

These exosomes are composed of proteins such as heat shock proteins, cell adhesion proteins, tetraspanin membrane proteins, cell signaling proteins, transcription proteins, and trafficking membrane fusion proteins [32][19]. A variety of lipid molecules, including phosphatidylserine (PS), phosphatidic acid, cholesterol, sphingomyelin (SM), arachidonic acid and other fatty acids, prostaglandins, and leukotrienes are also present in these exosomes [32][19]. In addition to these proteins and lipids constituents, exosomes are also rich in non-coding RNAs such as micro-RNA, small nuclear RNA, small nucleolar RNA, long non-coding RNA, piwi-interacting RNA, rRNA, and tRNA [32][19].

6. Mesenchymal Stem Cell-Derived Exosomes

These exosomes play vital autocrine/paracrine intercellular communication roles via the transfer of biological information to recipient cells. Their nature depends on their origin. For example, exosomes derived from MSCs have been shown to promote more therapeutic/regenerative activities compared to those promoted by MSCs alone. Animal model-based studies examining previously recommended that exosomes allow for the identification of better therapeutic candidates and provide a novel alternative to whole MSC cell-based therapy. These benefits may be attributed to the high safety and long shelf lives associated with exosomes that make them suitable and strong candidates for regenerative medicines and therapeutics for a variety of diseases [33][20]. It has been observed that MSC-derived exosomes inhibit pro-inflammatory mechanisms and are also associated with the alleviation of oxidative stress and pulmonary fibrosis and the remodeling that occurs in inflammatory lung disease [33][20].

Exosomes function according to their lineages, where their function is dependent upon the phenotype of their parental cell type. It has been reported that MSC-derived exosomes possess the potential to restore and maintain homeostasis based on their capability for protein and RNA transfer [34][21]. MSC-derived exosomes express several phenotypic markers, including CD29, CD73, CD44, and CD105 [35][22]. In addition to these phenotypic markers, MSC-derived exosomes also possess surface receptors (PDGFRB, EGFR, and PLAUR), cell adhesion molecules (FN1, EZR, IQGAP1, CD47, integrin, and LGALS1/LGALS3), signaling molecules (RRAS/NRAS, MAPK1, GNA13/GNG12, CDC42, and VAV2), and MSC-associated antigens (CD9, CD63, CD81, CD109, CD151, CD248, and CD276) [36][23]. It has been reported that MSC-derived exosomes contain more than 850 unique gene products and more than 150 miRNAs [37,38][24][25].

7. MSC-Derived Exosomes as a Prospective Treatment for COVID-19

MSC-derived exosomes can serve as an attractive target for treating COVID-19 and its associated complications such as lung injury and ARDS. These exosomes of MSC lineages possess immunomodulatory, tissue repair, and antiviral properties. A recent study suggested that MSC-derived exosomes can easily be substituted for MSCs, as these exosomes exert the same effects in the treatment of COVID-19 as do MSCs [61][26]. In another study, it was demonstrated that MSC-derived exosomes could inhibit the influenza virus due to their ability to transfer miRNAs and mRNA into lung epithelial cells to aid in the reduction of cellular apoptosis and viral replication [62][27]. In a study conducted in 2018, it was observed that MSC-derived exosomes induced a reduction in TNF-α, IL-1β, NF-κB, and matrix metalloprotease 9 (MMP-9) levels in the lung and caused decreased expression of IL-6 gene and increased production of IL-10 [63][28]. In another study, it was found that the transfer of miR-146a via exosomes exerted immunomodulatory effects through IL-1β augmentation [64][29].

Researchers are now discovering potential therapies against SARS-CoV-2 infection that include COVID-19 treatment based on MSCs or their derived exosomes [65,66,67,68][30][31][32][33]. As the pathogenesis of SARS-CoV-2 is similar to that of most of the previously encountered viruses that cause ARDS and lung injury, it has been proposed that based upon previous treatment approaches using MSCs or MSC-derived exosomes, the usefulness of exosome therapy should be explored in the context of SARS-CoV-2.MSC-derived exosomes were previously reported to elicit a positive response in ARDS and to inhibit cytokine storms by transferring mRNA and miRNAs to lung tissues [69,70][34][35]. In a preclinical model of non-infectious acute lung injuries or bacterial sepsis in the lungs, MSC-derived exosomes have shown protective effects mediated by miRNAs and inhibitory mRNAs [71][36]. It was suggested that pulmonary fibrosis of the lungs can be prevented in COVID-19-induced pneumonia through the use of MSC-derived exosomes. The rigorous anti-inflammatory activities of MSC exosomes can be used as an infusion in COVID-19 confirmed cases to prevent inflammatory responses.

Exosomes have been evaluated previously in the context of SARS-related coronavirus infection, and it was determined that exosomes containing the SARS-CoV spike protein “S” generate neutralizing antibody titers [72][37]. Kuate injected xenogenic exosomes without any adjuvant into SARS coronavirus-infected subjects and observed the presence of sufficient neutralizing antibodies [72][37]. In one recent clinical trial (NCT04276987), MSC-derived exosomes were found to be capable of treating severe subjects with novel coronavirus-associated pneumonia when used as an aerosol upon inhalation [73][38]. In a recently published prospective non-randomized open-label cohort study, the efficacy of allogenic BM-MSC-derived exosomes in the treatment of severe COVID-19 was tested in 24 SARS-CoV-2-infected subjects with moderate-to-severe acute respiratory distress syndrome [74][39]. A single injection of 15 mL was administered to the enrolled subjects intravenously, and the effects were monitored from days 1 to 14 post exosome injection [74][39]. A survival rate of 83% was observed, and out of 24 patients, 17 recovered, 3 remained critically ill (although stable), and 4 died due to unrelated treatment [74][39]. The authors concluded that exosomes derived from MSC lineages can provide a promising candidate for therapy in SARS-CoV-2 infection [74][39]. In another Chinese clinical trial, it was found that human MSCs and their derived exosomes could aid in the treatment of lung injury associated with novel coronavirus pneumonia (COVID-19) [75][40]. In a recently active clinical trial (NCT04389385), COVID-19-specific T-cell-derived exosomes were used via a metered-dose inhaler to treat pneumonia associated with SARS-CoV-2 infection [76][41]. Table 1 shows the prospective role of mesenchymal stem cell-mediated exosomes in SARS-CoV-2 infection [5,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114][5][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79].

Table 1. Studies showing pro-angiogenic role of mesenchymal stem cells-derived exosomes.

Articles

Conclusion of the study

Reference

Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts

Prostate cancer exosomes dominantly dictate a programme of MSC differentiation generating myofibroblasts with functional properties consistent with disease promotion.

[16]

[80]

Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis

Exosomal transfer of pro-angiogenic miRNAs plays an important role in MSC-mediated angiogenesis and stem cell-to-endothelial cell communication.

[17]

[81]

Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a

adMSC-Exo can transfer miR-125a to endothelial cells and promote angiogenesis by repressing DLL4.

[18]

[82]

Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells

MSC-derived exosomes may serve as a significant mediator of cell-to-cell communication within the tumor microenvironment and suppress angiogenesis by transferring anti-angiogenic molecules.

[19]

[83]

Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment

MSCs have a potential to exert anti-tumor activities, they largely provide service to the tumor using the multidirectional communication system established by exosomes in the TME.

[20]

[84]

Exosomes from cardiomyocyte progenitor cells and mesenchymal stem cells stimulate angiogenesis via EMMPRIN

CMPC and MSC exosomes have powerful pro-angiogenic effects, and this effect is largely mediated via the presence of EMMPRIN on exosomes.

[21]

[85]

Exosomes secreted by hypoxic cardiosphere-derived cells enhance tube formation and increase pro-angiogenic miRNA

Benefits of hypoxic CDC exosomes for the treatment of cardiac diseases by induction of angiogenesis via enrichment of pro-angiogenic exosomal miRNAs.

[22]

[86]

Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro

MSC exosomes were found to activate several signaling pathways important in wound healing (AKT, ERK, and STAT3) and induce the expression of a number of growth factors [hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF1), nerve growth factor (NGF), and stromal-derived growth factor-1 (SDF1)].

[23]

[87]

Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice

Implanted iMSCs-Exo was able to protect limbs from ischemic injury via the promotion of angiogenesis, which indicated that iMSCs-Exo may be a novel therapeutic approach in the treatment of ischemic diseases.

[24]

[11]

Exosomes from hypoxia-treated human adipose-derived mesenchymal stem cells enhance angiogenesis through VEGF/VEGF-R

Exosomes from hypoxia-treated human ADSCs possess a higher capacity to enhance angiogenesis in fat grafting, at least partially, via regulating VEGF/VEGF-R signaling.

[25]

[12]

References

  1. Zhou, P.; Xing-Lou, Y.; Xian-Guang, W.; Ben, H.; Lei, Z.; Wei, Z.S.; Yan, Z.; Bei, L.; Chao-Lin, H.; Hui-Dong, C.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273.
  2. De Wit, E.; van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016, 14, 523–534.
  3. World Health Organization. WHO Coronavirus Disease (COVID-19) Dashboard. 2020. Available online: (accessed on 3 March 2021).
  4. Trade Set to Plunge as COVID-19 Pandemic Upends Global Economy. Available online: (accessed on 3 March 2021).
  5. Shan, L.; Danyi, P.; Huijun, Q.; Ke, Y.; Zhou, F.; Lin, Z. Mesenchymal stem cells as a potential therapy for COVID-19. Stem Cell Res. Ther. 2020, 11, 169.
  6. Mason, R.J. Pathogenesis of COVID-19 from a cell biologic perspective. Eur. Respir. J. 2020, 55, 2000607.
  7. Zhang, W.; Du, R.-H.; Li, B.; Zheng, X.-S.; Yang, X.-L.; Hu, B.; Wang, Y.-Y.; Xiao, G.-F.; Yan, B.; Shi, Z.-L.; et al. Molecular and serological investigation of 2019-nCoV infected patients: Implication of multiple shedding routes. Emerg. Microbes Infect. 2020, 9, 386–389.
  8. Markus, H.; Hannah, K.W.; Nadine, K.; Marcel, M.; Christian, D.; Stefan, P. The novel coronavirus 2019 (2019-nCoV) uses the SARS-coronavirus receptor ACE2 and the cellular protease TMPRSS2 for entry into target cells. bioRxiv 2020.
  9. Tortorici, M.A.; Veesler, D. Structural Insights into Coronavirus Entry; Elsevier Inc.: Amsterdam, The Netherlands, 2019; Volume 105, pp. 93–116.
  10. Wu, Z.; McGoogan, J.M. Characteristics of and important lessons from the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a report of 72314 cases from the chinese center for disease control and prevention. J. Am. Med. Assoc. 2020, 323, 1239–1242.
  11. Whelan, D.S.; Caplice, N.M.; Clover, A.J.P. Mesenchymal stromal cell derived CCL2 is required for accelerated wound healing. Sci. Rep. 2020, 10, 1–12.
  12. Gazdic, M.; Volarevic, V.; Arsenijevic, N.; Stojkovic, M. Mesenchymal Stem Cells: A Friend or Foe in Immune-Mediated Diseases. Stem Cell Rev. Rep. 2015, 11, 280–287.
  13. Matthay, M.A.; Calfe, C.S.; Zhuo, H.; Thompson, B.T.; Wilson, J.G.; Levitt, J.E.; Rogers, A.J.; Gotts, J.E.; Wiener-Kronish, J.P.; Bajwa, E.K.; et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): A randomized phase 2a safety trial. Lancet Respir. Med. 2019, 7, 154–162.
  14. Xu, A.L.; Rodriguez, L.A.; Walker, K.P.; Mohammadipoor, A.; Kamucheka, R.M.; Cancio, L.C.; Batchinsky, A.I.; Antebi, B. Mesenchymal stem cells reconditioned in their own serum exhibit augmented therapeutic properties in the setting of acute respiratory distress syndrome. Stem Cells Transl. Med. 2019, 8, 1092–1106.
  15. Morrison, T.J.; Jackson, M.V.; Cunningham, E.K.; Kissenpfennig, A.; McAuley, D.F.; O’Kane, C.M.; Krasnodembskaya, A.D. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am. J. Respir. Crit. Care Med. 2017, 196, 1275–1286.
  16. Abraham, A.; Krasnodembskaya, A. Mesenchymal stem cell-derived extracellular vesicles for the treatment of acute respiratory distress syndrome. Stem Cells Transl. Med. 2020, 9, 28–38.
  17. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1–43.
  18. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  19. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 1–18.
  20. Fujita, Y.; Kadota, T.; Araya, J.; Ochiya, T.; Kuwano, K. Clinical application of mesenchymal stem cell-derived extracellular vesicle-based therapeutics for inflammatory lung diseases. J. Clin. Med. 2018, 7, 355.
  21. 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.
  22. Ramos, T.L.; Sánchez-Abarca, L.; Muntión, S.; Preciado, S.; Puig, N.; López-Ruano, G.; Hernández-Hernánde, Á.; Redondo, A.; Ortega, R.; Rodríguez, C.; et al. MSC surface markers (CD44, CD73 and CD90) can identify human MSC-derived extracellular vesicles by conventional flow cytometry. Cell Commun. Signal. 2016, 14, 1–14.
  23. Kim, H.-S.; Choi, D.-Y.; Yun, S.J.; Choi, S.-M.; Kang, J.W.; Jung, J.W.; Hwang, D.; Kim, K.P.; Kim, D.-W. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J. Proteome Res. 2012, 11, 839–849.
  24. Chen, T.S.; Lai, R.C.; Lee, M.M.; Choo, A.B.; Lee, C.N.; Lim, S.K. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2010, 38, 215–224.
  25. Lai, R.C.; Tan, S.S.; Teh, B.J.; Sze, S.K.; Arslan, F.; De Kleijn, D.P.; Choo, A.; Lim, S.K. Proteolytic Potential of the MSC Exosome proteome: Implications for an exosome-mediated delivery of therapeutic proteasome. Int. J. Proteom. 2012, 2012, 1–14.
  26. Taghavi-Farahabadi, M.; Mahmoudi, M.; Soudi, S.; Hashemi, S.M. Hypothesis for the management and treatment of the COVID-19-induced acute respiratory distress syndrome and lung injury using mesenchymal stem cell-derived exosomes. Med. Hypotheses 2020, 144, 109865.
  27. Khatri, M.; Richardson, L.A.; Meulia, T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res. Ther. 2018, 9, 17.
  28. Chang, C.-L.; Chen, H.-H.; Chen, K.-H.; Chiang, J.Y.; Li, Y.-C.; Lin, H.-S.; Sung, P.-H.; Yip, H.-K. Adipose-derived mesenchymal stem cell-derived exosomes markedly protected the brain against sepsis syndrome induced injury in rat. Am. J. Transl. Res. 2019, 11, 3955–3971.
  29. Song, Y.; Dou, H.; Li, X.; Zhao, X.; Li, Y.; Liu, D.; Ji, J.; Liu, F.; Ding, L.; Ni, Y.; et al. Exosomal miR-146a contributes to the enhanced therapeutic efficacy of interleukin-1β-primed mesenchymal stem cells against sepsis. Stem Cells 2017, 35, 1208–1221.
  30. Li, X.; Geng, M.; Peng, Y.; Meng, L.; Lu, S. Molecular immune pathogenesis and diagnosis of COVID-19. J. Pharm. Anal. 2020, 19, 1–7.
  31. Devaney, J.; Horie, S.; Masterson, C.; Elliman, S.; Barry, F.; O’Brien, T.; Curley, G.F.; O’Toole, D.; Laffey, J.G. Human mesenchymal stromal cells decrease the severity of acute lung injury induced by E. coli in the rat. Thorax 2015, 70, 625–635.
  32. Lee, J.W.; Fang, X.; Gupta, N.; Serikov, V.; Matthay, M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc. Natl. Acad. Sci. USA 2009, 106, 16357–16362.
  33. Bao, L.; Deng, W.; Huang, B.; Gao, H.; Liu, J.; Ren, L.; Wei, Q.; Yu, P.; Xu, Y.; Qi, F.; et al. The Pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 2020, 583, 830–833.
  34. Mahmoudi, M.; Taghavi, F.M.; Hashemi, S.M. Exosomes: Mediators of immune regulation. Immunoregulation 2019, 2, 3–8.
  35. El Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357.
  36. Monsel, A.; Zhu, Y.G.; Gennai, S. Therapeutic effects of human mesenchymal stem cell-derived microvesicles in severe pneumonia in mice. Am. J. Respir. Crit. Care Med. 2015, 192, 324–336.
  37. Seraphin, K.; Jindrich, C.; Hans, W.D.; Uberala, K. Exosomal vaccines containing the S protein of the SARS coronavirus inducehigh levels of neutralizing antibodies. Virology 2007, 362, 26–37.
  38. A Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus Pneumonia. Available online: (accessed on 3 March 2021).
  39. 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.
  40. Available online: (accessed on 3 March 2021).
  41. COVID-19 Specific T Cell Derived Exosomes (CSTC-Exo). Available online: (accessed on 3 March 2021).
  42. Golchin, A.; Seyedjafari, E.; Ardeshirylajimi, A. mesenchymal stem cell therapy for COVID-19: Present or future. Stem Cell Rev. Rep. 2020, 16, 427–433.
  43. Rajarshi, K.; Chatterjee, A.; Ray, S. Combating COVID-19 with mesenchymal stem cell therapy. Biotechnol. Rep. 2020, 26, e00467.
  44. Metcalfe, S.M. Mesenchymal stem cells and management of COVID-19 pneumonia. Med. Drug Discov. 2020, 5, 100019.
  45. Leng, Z.; Zhu, R.; Hou, W.; Feng, Y.; Yang, Y.; Han, Q.; Shan, G.; Meng, F.; Du, D.; Wang, S.; et al. Transplantation of ACE2- mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020, 11, 216–228.
  46. Liang, B.; Chen, J.; Li, T.; Wu, H.; Yang, W.; Li, Y.; Li, J.; Yu, C.; Nie, F.; Ma, Z.; et al. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells. Medicine 2020, 99, 1–6.
  47. Zhang, B.; Yin, Y.; Lai, R.C.; Tan, S.S.; Choo, A.B.H.; Lim, S.K. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014, 23, 1233–1244.
  48. Cosenza, S.; Toupet, K.; Maumus, M.; Luz-Crawford, P.; Blanc-Brude, O.; Jorgensen, C.; Noël, D. Mesenchymal stem cells-derived exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics 2018, 8, 1399–1410.
  49. Huang, L.; Ma, W.; Ma, Y.; Feng, D.; Chen, H.; Cai, B. Exosomes in mesenchymal stem cells, a new therapeutic strategy for cardiovascular diseases? Int. J. Biol. Sci. 2015, 11, 238–245.
  50. Cosenza, S.; Ruiz, M.; Toupet, K.; Jorgensen, C.; Noël, D. Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci. Rep. 2017, 7, 1–12.
  51. Stella, C.; Maxime, R.; Karine, T.; Christian, J.; Danièle, N. Mesenchymal stem cell-derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti-inflammation. Cell. Physiol. Biochem. 2015, 37, 2415–2424.
  52. Yu, B.; Zhang, X.; Li, X. Exosomes derived from mesenchymal stem cells. Int. J. Mol. Sci. 2014, 15, 4142–4157.
  53. Maumus, M.; Jorgensen, C.; Noël, D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie 2013, 95, 2229–2234.
  54. Domenis, R.; Cifù, A.; Quaglia, S.; Pistis, C.; Moretti, M.; Vicario, A.; Parodi, P.C.; Fabris, M.; Niazi, K.R.; Soon-Shiong, P.; et al. Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes. Sci. Rep. 2018, 8, 1–11.
  55. Liu, H.; Li, R.; Liu, T.; Yang, L.; Yin, G.; Xie, Q. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunol. Res. 2016, 64, 831–840.
  56. Chowdhury, R.; Webber, J.P.; Gurney, M.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2014, 6, 715–731.
  57. Gong, M.; Yu, B.; Wang, J.; Wang, Y.; Liu, M.; Paul, C.; Millard, R.W.; Xiao, D.-S.; Ashraf, M.; Xu, M. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget 2017, 8, 45200–45212.
  58. Asgarpour, K.; Shojaei, Z.; Amiri, F.; Ai, J.; Mahjoubin-Tehran, M.; Ghasemi, F.; Mirzaei, H.; ArefNezhad, R.; Hamblin, M.R. Exosomal microRNAs derived from mesenchymal stem cells: Cell-to-cell messages. Cell Commun. Signal. 2020, 18, 149.
  59. Lee, J.K.; Park, S.R.; Jung, B.K.; Jeon, Y.K.; Lee, Y.S.; Kim, M.K.; Kim, Y.G.; Jang, J.Y.; Kim, C.W. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS ONE 2013, 8, e84256.
  60. Whiteside, T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin. Immunol. 2018, 35, 69–79.
  61. Vrijsen, K.R.; Maring, J.A.; Chamuleau, S.A.J.; Verhage, V.; Mol, E.A.; Deddens, J.C.; Metz, C.H.G.; Lodder, K.; Van Eeuwijk, E.C.M.; Van Dommelen, S.M.; et al. Exosomes from cardiomyocyte progenitor cells and mesenchymal stem cells stimulate angiogenesis via EMMPRIN. Adv. Healthc. Mater. 2016, 5, 2555–2565.
  62. Helia, N.; Mohit, I.; Sarah, R.; Azam, S.; Ensiyeh, H.S.; Nasser, A.; Hossein, B. Exosomes secreted by hypoxic cardiosphere-derived cells enhance tube formation and increase pro-angiogenic miRNA. J. Cell. Biochem. 2018, 119, 4150–4160.
  63. Shabbir, A.; Cox, A.; Rodriguez-Menocal, L.; Salgado, M.; Van Badiavas, E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis In Vitro. Stem Cells Dev. 2015, 24, 1635–1647.
  64. Hu, G.-W.; Li, Q.; Niu, X.; Hu, B.; Liu, J.; Zhou, S.-M.; Guo, S.-C.; Lang, H.-L.; Zhang, C.-Q.; Wang, Y.; et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res. Ther. 2015, 6, 1–15.
  65. Han, Y.; Ren, J.; Bai, Y.; Pei, X.; Han, Y. Exosomes from hypoxia-treated human adipose-derived mesenchymal stem cells enhance angiogenesis through VEGF/VEGF-R. Int. J. Biochem. Cell Biol. 2019, 109, 59–68.
  66. Wang, B.; Yao, K.; Huuskes, B.M.; Shen, H.H.; Zhuang, J.; Godson, C.; Brennan, E.P.; Wilkinson-Berka, J.L.; Wise, A.F.; Ricardo, S.D. Mesenchymal stem cells deliver exogenous MicroRNA-let7c via exosomes to attenuate renal fibrosis. Mol. Ther. 2016, 24, 1290–1301.
  67. Qu, Y.; Zhang, Q.; Cai, X.; Li, F.; Ma, Z.; Xu, M.; Lu, L. Exosomes derived from miR-181-5p-modified adipose-derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. J. Cell. Mol. Med. 2017, 21, 2491–2502.
  68. Tseliou, E.; Fouad, J.; Reich, H.; Slipczuk, L.; De Couto, G.; Aminzadeh, M.; Middleton, R.; Valle, J.; Weixin, L.; Marbán, E. Fibroblasts rendered antifibrotic, antiapoptotic, and angiogenic by priming with cardiosphere-derived extracellular membrane vesicles. J. Am. Coll. Cardiol. 2015, 66, 599–611.
  69. Maria, A.T.J.; Toupet, K.; Bony, C.; Pirot, N.; Vozenin, M.-C.; Petit, B.; Roger, P.; Batteux, F.; Le Quellec, A.; Jorgensen, C.; et al. Antifibrotic, antioxidant, and immunomodulatory effects of mesenchymal stem cells in HOCL-induced systemic sclerosis. Arthritis Rheumatol. 2016, 68, 1013–1025.
  70. Rong, X.; Liu, J.; Yao, X.; Jiang, T.; Wang, Y.; Xie, F. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/β-catenin pathway. Stem Cell Res. Ther. 2019, 10, 1–11.
  71. Rong, X.; Liu, J.; Yao, X.; Jiang, T.; Wang, Y.; Xie, F. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front. Immunol. 2014, 5, 1–9.
  72. Willis, G.R.; Fernandez-Gonzalez, A.; Anastas, J.; Vitali, S.H.; Liu, X.; Ericsson, M.; Kwong, A.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am. J. Respir. Crit. Care Med. 2018, 197, 104–116.
  73. Wen, D.; Peng, Y.; Liu, D.; Weizmann, Y.; Mahato, R.I. Mesenchymal stem cell and derived exosome as small RNA carrier and Immunomodulator to improve islet transplantation. J. Control. Release 2016, 238, 166–175.
  74. Casado, J.G.; Blázquez, R.; Vela, F.J.; Álvarez, V.; Tarazona, R.; Sánchez-Margallo, F.M. Mesenchymal stem cell-derived exosomes: Immunomodulatory evaluation in an antigen-induced synovitis porcine model. Front. Veter. Sci. 2017, 4, 39.
  75. Nojehdehi, S.; Soudi, S.; Hesampour, A.; Rasouli, S.; Soleimani, M.; Hashemi, S.M. Immunomodulatory effects of mesenchymal stem cell–derived exosomes on experimental type-1 autoimmune diabetes. J. Cell. Biochem. 2018, 119, 9433–9443.
  76. Liu, M.; Wang, J.; Liu, M.; Hu, X.; Xu, J. Study of immunomodulatory function of exosomes derived from human umbilical cord mesenchymal stem cells. Zhonghua Yi Xue Za Zhi 2015, 95, 2630–2633.
  77. Showalter, M.R.; Wancewicz, B.; Fiehn, O.; Archard, J.A.; Clayton, S.; Wagner, J.; Deng, P.; Halmai, J.; Fink, K.D.; Bauer, G.; et al. Primed mesenchymal stem cells package exosomes with metabolites associated with immunomodulation. Biochem. Biophys. Res. Commun. 2019, 512, 729–735.
  78. Börger, V.; Bremer, M.; Ferrer-Tur, R.; Gockeln, L.; Stambouli, O.; Becic, A.; Giebel, B. Mesenchymal stem/stromal cell-derived extracellular vesicles and their potential as novel immunomodulatory therapeutic agents. Int. J. Mol. Sci. 2017, 18, 1450.
  79. Bruno, S.; Deregibus, M.C.; Camussi, G. The secretome of mesenchymal stromal cells: Role of extracellular vesicles in immunomodulation. Immunol. Lett. 2015, 168, 154–158.
  80. Hancock, A.S.; Stairiker, C.J.; Boesteanu, A.C.; Monzón-Casanova, E.; Lukasiak, S.; Mueller, Y.M.; Stubbs, A.P.; García-Sastre, A.; Turner, M.; Katsikis, P.D. Transcriptome analysis of infected and bystander Type 2 alveolar epithelial cells during influenza A virus infection reveals in vivo Wntpathway downregulation. J. Virol. 2018, 92, e01325-18.
  81. Mossel, E.C.; Wang, J.; Jeffers, S.; Edeen, K.E.; Wang, S.; Cosgrove, G.P.; Funk, C.J.; Manzer, R.; Miura, T.A.; Pearson, L.D.; et al. SARS-CoV replicates in primary human alveolar type II cell cultures but not in type I-like cells. Virology 2008, 372, 127–135.
  82. Viola, K.W.; Becher, A.; Tönnies, M.; Gudrun, H.; Jessica, K.; Torsten, T.B.; Paul, S.; Jens, N.; Jens, C.R.; Kolja, S.; et al. Influenza A virus’s target type II pneumocytes in the human lung. J. Infect. Dis. 2012, 206, 1685–1694.
  83. Qian, Z.; Travanty, E.A.; Oko, L.; Edeen, K.; Berglund, A.; Wang, J.; Ito, Y.; Holmes, K.V.; Mason, R.J. Innate immune response of human alveolar type II cells infected with severe acute respiratory syndrome–coronavirus. Am. J. Respir. Cell Mol. Biol. 2013, 48, 742–748.
  84. 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.
  85. Nikolaos, M.N.; John, G.N.; Lori, B.P.; Jason, C.G.; Vasuki, U.; Hoaxing, W.; Atsushi, S.; Kara, E.L.; Human, L.; Mitchell, R.W.; et al. Mitogenic stimulation accelerates influenza induced mortality by increasing susceptibility of alveolar type II cells to infection. Proc. Natl. Acad. Sci. USA 2017, 114, E6613–E6622.
  86. de Wilde, A.H.; Snijder, E.J.; Kikkert, M. Roles of Host Gene and Non-Coding RNA Expression in Virus Infection; Springer International Publishing: Berlin/Heidelberg, Germany, 2018; pp. 1–42.
  87. Sawicki, S.G.; Sawicki, D.L. Coronavirus transcription: A perspective. Curr. Top. Microbiol. Immunol. 2005, 287, 31–55.
More
Video Production Service