Next Article in Journal
Development of Native Essential Oils from Forestry Resources in South Korea
Next Article in Special Issue
Impact of Experimental Conditions on Extracellular Vesicles’ Proteome: A Comparative Study
Previous Article in Journal
Low Incidence of Acute Antibody-Mediated Rejection after HLA Desensitization in Living Donor Kidney Transplant Recipients
Previous Article in Special Issue
Extracellular Vesicles in Chronic Demyelinating Diseases: Prospects in Treatment and Diagnosis of Autoimmune Neurological Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease

Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan 602-739, Republic of Korea
*
Author to whom correspondence should be addressed.
Life 2022, 12(12), 1994; https://doi.org/10.3390/life12121994
Submission received: 17 October 2022 / Revised: 23 November 2022 / Accepted: 25 November 2022 / Published: 29 November 2022

Abstract

:
Mesenchymal stem cells (MSCs) have been reported as promising candidates for the treatment of various diseases, especially allergic diseases, as they have the capacity to differentiate into various cells. However, MSCs itself have several limitations such as creating a risk of aneuploidy, difficulty in handling them, immune rejection, and tumorigenicity, so interest in the extracellular vesicles (EVs) released from MSCs are increasing, and many studies have been reported. Previous studies have shown that extracellular vesicles (EVs) produced by MSCs are as effective as the MSCs themselves in suppression of allergic airway inflammation through the suppression of Th2 cytokine production and the induction of regulatory T cells (Treg) expansion. EVs are one of the substances secreted by paracrine induction from MSCs, and because it exerts its effect by delivering contents such as mRNA, microRNA, and proteins to the receptor cell, it can reduce the problems or risks related to stem cell therapy. This article reviews the immunomodulatory properties of MSCs-derived EVs and their therapeutic implications for allergic airway disease.

1. Introduction

Allergic rhinitis and asthma are representative atopic diseases, and 30% of allergic rhinitis patients have asthma and 70% of asthma patients have allergic rhinitis [1,2]. Allergic rhinitis and asthma are caused by various antigens and specific immunoglobulin E (Ig E), and they are immunologically characterized by the excessive activation of type 2 helper T (Th2) cells [3]. Th2 cell-induced inflammation is characterized by a significant increase in interleukin (IL)-4, IL-5, and IL-13 levels, which induce and high serum Ig E levels and airway eosinophilic inflammation and airway hyper responsiveness (AHR) [3]. The Th1 and Th2 cytokines are mutually antagonistic, and the selective suppression of Th2 responses may be crucial for protection against allergic inflammation [1,3]. There is mounting evidence that the insufficient suppression of Tregs as well as the imbalance of Th1/Th2 responses in the pathogenesis of allergic airway disease play an important role in excessive Th2 responses [4].
Stem cells are undifferentiated cells that have the potential to develop into many different cell types. They have been reported to possess the ability to self-renew in an undifferentiated state and to differentiate into many types of cells with specific functions upon receiving appropriate triggers. Stem cells can be divided into two groups, embryonic and adult. Embryonic stem cells are derived during early development at the blastocyst stage, and they are pluripotent, implying they can develop into any cell type. However, adult stem cells are multipotent, meaning they can only differentiate into certain types of tissue [5]. Both embryonic and adult stem cells have been studied as a promising source for therapeutic applications in the repair of damaged tissues and regenerative medicine [6,7,8,9].
Mesenchymal stem cells (MSCs) represent an important stem cell population with multi-potent capabilities, which means that they may have high utility for translational clinical applications. MSCs can be isolated from a number of adult tissues, and they can differentiate into several mesenchymal lineages both in vitro and in vivo, such as adipose tissue, cartilage, bone, muscle, and nerve [10,11,12,13,14].
In addition to their multi-lineage potential, MSCs have been shown to possess the unique ability to suppress an immune response and modulate inflammation. MSCs can inhibit natural killer cell functions, modulate dendritic cell maturation, and suppress the allogeneic T cell response by alternating the cytokine secretion profile of dendritic cells and T cells induced by an allogeneic immune reaction [15,16]. Therefore, MSCs have been reported to have anti-inflammatory and immunomodulatory effects in various chronic inflammatory diseases such as autoimmune encephalomyelitis, graft-versus-host disease, inflammatory bowel disease, and type I diabetes [17,18,19,20,21,22,23].
Several studies have shown that MSCs derived from adipose tissue (ASCs) and other MSCs can improve allergic airway inflammation in allergic rhinitis and asthma [24,25,26,27,28,29,30,31,32]. The immunomodulatory mechanism of MSC is related to the increase of soluble factors such as transforming growth factor-β (TGF-β) and prostaglandin E2 (PGE2) and the upregulation of Treg, but further studies are needed [24,25,26]. Furthermore, MSCs could modulate the antigen recognition of antigen presenting cells that mediate the cellular immune response, including dendritic cells, macrophages, and B cells [29,30]. However, the treatment using MSCs has some clear limitations including tumorigenicity, immune rejection, difficulty in handling them, and a risk of aneuploidy [31].
MSCs secrete a variety of autocrine and paracrine factors including angiogenic factors, chemokines, cytokines, and growth factors that have similar therapeutic effects as systemically administered MSCs do [32]. Previous several studies demonstrated that the intranasal administration of secretome, also known as a conditioned medium, is as secreted molecule from cultured MSCs can convey the same therapeutic actions as the systemic administration of the MSCs themselves [32,33,34]. The MSCs secretome can decrease an allergic airway inflammation by inhibiting Th2 cytokine release and the induction of Treg expansion even without MSCs [32,33,34]. These findings indicate that the main beneficial effects of MSCs are mediated via paracrine actions, although potentially triggered by cell-to-cell contact events. Furthermore, compared with MSCs transplantation, cell-free therapies mediated by the MSC secretome have a lot of advantages including the low possibility of immune rejection, the ease of storage, the ease of handling, and no risk of vascular occlusion or aneuploidy [35]. All of these paracrine factors can be found within the extracellular vesicles (EVs), which are cell-secreted vesicles, they are a key part of the secretome which is receiving lots of attention. Therefore, the main focus of this article is to review the immunomodulatory effects of MSC-derived EVs and to summarize the latest knowledge on clinical applications in allergic airway diseases.

2. Characteristics of EVs

The EV isa spherical bi-layered proteolipid secreted from the cell into the extracellular space, and it is derived by various cells such as hematopoietic cells, endothelial cells, epithelial cells, and cancer cells. The EVs are classified into 30–100 nm exosomes, 100–1000 nm microvesicles, and 1–5 μm apoptotic bodies, according to their origin, biogenesis, shape, composition, and size [36].
Exosomes, which are referred to as the intraluminal vesicles, are heterogenous in the 40–200 nm size range, and they are generated through the integration of the endosomal membrane of multivesicular bodies (MVB) and subsequent exocytosis. Additionally, exosomes are secreted by all cell types such as saliva, urine, plasma, semen, cerebrospinal fluid (CSF), bronchial fluid, synovial fluid, gastric acid, breast milk, serum, amniotic fluid, tears, lymph, and bile [37,38,39,40,41,42,43,44,45,46,47,48]. In particular, they are involved in protein storage, transportation, release, and recycling [49]. The MVB is eventually sent to the lysosome where it is either degraded along with all of its components or fuses with the cell’s plasma membrane to release its contents, including exosomes, into the extracellular space [50]. The MVB and exosome formation and release are regulated through endosomal sorting complexes that are essential for transport (ESCRT) pathways [51].
Because exosome formation and MVB transport are regulated by ESCRT proteins, these proteins are expected to be found in exosomes, and regardless of the cell type from which they originate, these proteins are termed the exosome marker proteins [52]. Although some studies indicate that there is an ESCRT-independent mechanism as another mechanism by which cells release exosomes into the extracellular space, in both of the mechanisms, CD9, CD63, and CD81 are identified as exosome-specific markers [53].
Exosomes were originally known to remove unnecessary or unwanted substances, but they have been reported to be involved in cell-to-cell communication, cell maintenance, and tumors. Several studies have reported that exosomes act as antigen-presenting vesicles to stimulate immune responses, and in the nervous system, exosomes play an important role in tissue repair and regeneration by promoting myelin formation, neurite outgrowth, and neuronal survival [54,55]. In addition, pathogenic proteins such as alpha-synuclein, superoxide dismutase, and beta-amyloid peptide which may aid in disease progression have been found to be exosomes from the central nervous system (CNS) [56,57]. The exact mechanism is not yet fully understood, and it appears that the formation of MVB can be stimulated by growth factors, and the cells modulate exosome production as needed [58].
In clinical applications and uses of exosomes, it is important to study their ability to act as carriers of biomarkers for several diseases. The exosomes in cerebrospinal fluid were found to contain alpha-synuclein, a protein related to Parkinson’s disease [59,60]. In addition, the exosomes isolated from urine were reported to reflect acute kidney injury [61], and the markers for pancreatic cancer and lung cancer were also found in exosomes [62,63]. Because exosomes are found in blood or urine, the EVs can be used to monitor the disease state of a patient or their state after the treatment of a specific disease, and changes in the biomarkers according to the course of the disease can also be evaluated [64]. It has also been reported that it can be used for immunological purposes or vaccine development because it acts as an antigen-presenting vesicle [54,65].
In addition, the exosomes can cross cell membranes and target specific cell types, making them suitable candidates for immunological applications, as well as creating drug delivery systems that move RNA and proteins to specific parts of the body through exosomes [66,67]. There are also several reports that the exosomes themselves can act as therapeutic substances to reduce tissue damage, and the potential applications and uses of exosomes have a broad range as biomarkers, drug delivery devices, and therapeutic tools in the clinical setting [68,69,70].
On the other hand, microvesicles (MVs), unlike exosomes, are relatively heterogeneous in their size, ranging from 50 to 1000 nm, and they are released either directly from the plasma membrane or through budding [71,72]. Specific protein markers that distinguish different types of EVs, MVs, exosomes, and apoptosis have not been identified, but they differ in their protein profiles due to them having different pathways of formation [73,74]. Although the proteomic profile of MVs is dependent on the method of isolation, there is a category of proteins called “marker proteins” that are proteins found in MVs irrespective of their cellular origin as a result of the biogenesis processes [75]. They are known to be clustered on the surface of plasma membrane, mainly as cytoplasmic and plasma membrane-related proteins such as tetraspanin [76]. However, specific markers for differentiating MVs from exosomes are still lacking. MVs, like exosomes, are known to be involved in intercellular communication between adjacent cells and distant cells, such as hormones, growth factors, and cytokines [77].
The apoptotic body is released into the extracellular space by the dying cell, and after the cell contracts, the hydrostatic pressure increases, and the plasma membrane of the cell is separated from the cytoskeleton, and it is formed [78]. The composition of the apoptotic bodies is in contrast to the MVs and exosomes, with there being distinct differences in their proteomic profiles [79].
As mentioned above, the released EVs contain various bioactive transmitters such as proteins, microRNAs, and mRNAs, and they play an important role in cell-to-cell communication and interaction with the surrounding microenvironment [80,81,82]. As the paracrine effect of the MSC secretome released by the MSCs which migrates into the diseased site has been reported to be an important factor in the mechanism of MSCs transplantation, the importance of stem cell-derived EVs is further emphasized in the field of stem cell application.
Gan et al. reported that EVs mediate the pathological communication between dysfunctional adipose tissue and the heart, exacerbating ischemic heart damage in obesity and diabetes. The EVs can be used as a therapeutic tool to prevent ischemic heart damage in obesity and diabetes by inhibiting the production of exosomes from abnormal adipocytes [83]. Certain regulatory microRNAs have been reported to have an immunomodulatory effect even in allergic airway disease. miR-21-5p, miR-142-3p, miR-223-3p, and miR-126-3p induce the maturation of dendritic cells, and miR 1470 modulates the immune response in asthmatics by differentiating CD4+ T cells into Treg cells via P27KIP1 [84,85,86]. In particular, miR-126-3p induces an increase in the endothelial cell function and tight junction protein expression, decreases the VEGF-α and HMGB1 levels, and miR-146a-5p inhibits group 2 innate lymphoid cells [85,87]. In addition, the MSC-EVs-derived microRNAs with biological activity in allergic airway diseases are summarized in Table 1 [88,89,90,91,92,93,94].
The mechanism by which the EVs are secreted and moved to the target cell to deliver therapeutic effects is that the EVs are secreted and contact the plasma membrane of the target cells, and then, they are absorbed through the interaction between the EVs and plasma membrane by direct fusion or endocytosis. In most cases, it moves to the lysosome, where the proteins and lipids in the EVs are degraded to supply metabolites to the target cells [80]. Since all of the EVs are surrounded by a bi-layered lipid membrane, it is possible to target the end organ cell or microenvironment, while protecting the inner factor from degrading enzymes in the blood circulation. The EVs have a biological structure similar to that of liposomes, so they are more stable in vivo than MSCs are, and it is relatively easy to improve and modify their surface properties for their application as a drug delivery system or for enhancing their therapeutic potential [75,76]. Accumulating evidence shows that administration of MSC-derived EVs have a therapeutic efficacy that is equal to or greater than that of MSC in suppressing allergic airway inflammation [32,34].

3. Immunomodulatory Effects of EVs for Allergic Airway Diseases

MSCs could improve the allergic airway disease by inhibiting the proliferation and function of dendritic cells which have an immunomodulatory effect, and which are differentiated into T cells and B cells [95,96,97]. The intravenous injection of MSCs significantly reduced eosinophil infiltration in the nasal mucosa and lung tissue of allergic mouse models and improved the degree of airway hypersensitivity and allergic symptoms [24,25,26]. The MSCs administrated intravenously decreased the number of Th2 cytokines such as IL-4, IL-5, IL-13, and IL-4-positive CD4+ T cells, but they increased the number of Th1 cytokine, IFN- γ and IFN- γ positive CD4+ T cells in the bronchoalveolar lavage (BAL) fluid and lung draining lymph nodes (LLNs) in an AR and asthma mouse model. The MSCs resulted in a significant decrease in the total and ovalbumin (OVA)-specific IgE and IgG1 levels. Tregs, which is characterized by the expression of transcription factor Foxp3, was significantly increased in the LLNs of asthmatic mice after the MSCs administration. Various soluble factors, including TGF-β and PGE2, are secreted by the MSCs that have migrated to the lungs by intravenous or nasal routes of administration, leading to the expansion of Tregs. Anti-inflammatory cytokines (IL-10 and TGF-β) are secreted by Tregs, which ultimately reduce the amount of pulmonary eosinophil infiltration as well as the production of allergy-specific Th2 cytokines and Ig. [24,25,26]. Additionally, in lung histology, eosinophil infiltration and inflammatory cell deposition in the peribronchial and perivascular areas were significantly decreased in the EV group compared to that of the asthma-inducing group [98].
Several studies have reported that the MSC secretomes and MSCs-derived EVs show the same immunomodulatory effects as the stem cells themselves do in allergic airway diseases [32,99,100,101]. Previous studies have shown that bone marrow, umbilical cord, and adipose tissue-derived MSCs and their EVs have the similar immunomodulatory effects in asthmatic mice. Therefore, the efficacy of MSCs-derived EVs does not depend on the MSC source tissue. Furthermore, the systemic and intranasal administration of MSC-derived EVs showed similar immunosuppressive effects in asthmatic mice [99]. De Castro et al. reported that intravenous stem cell culture media and EVs significantly reduced the degree of airway hypersensitivity and eosinophil infiltration of the lung tissues in a mouse model of asthma in the same manner as stem cells did, and the levels of IL-4, IL-5, and IL-6 were significantly reduced. Furthermore, the intravenous administration of human adipose tissue-derived MSCs and EVs reduced the total number of inflammatory cells and the ratio of eosinophils in BAL fluid, IL-5 levels in lung tissues and CD3+ CD4+ T cells in the thymus. However, the number of eosinophils in the lung tissues, the levels of IL-4, IL-13, eotaxin, and CD3+ CD4+ T cells in the BALF, and the pulmonary function showed inconsistent results [100]. Recently, intranasally administrated ASC-derived EVs significantly reduced the degree of allergic airway inflammation and improved AHR through induction of Tregs expansion in asthmatic mice. The intranasal administration of ASC-derived EVs to asthmatic mice resulted in a remarkable reduction of eosinophils and inflammatory cells in the BAL fluid, the serum total and the OVA-specific IgE levels, and the degree of eosinophilic lung inflammation. The level of IL-4 was significantly decreased in the BAL fluid and LLNs, whereas IFN-γ was significantly increased in the BAL fluid. Additionally, CD4+IL-4+ T cells were markedly decreased after an ASC-derived EV treatment, whereas the CD4+CD25+Foxp3+ T cells and CD4+IFN-γ+ T cells were notably increased in the LLNs of asthmatic mice [98].
In an in vitro study, the authors isolated EVs from culture supernatants of murine ASC, which were evaluated the immunomodulatory effects of EVs on Th2-mediated inflammation which was induced by Aspergillus protease antigens in mouse lung epithelial cells and primary lung epithelial cells.
Cho et al. reported that ASC-derived EVs suppressed Th2-mediated inflammation through the upregulation of TGF-β and IL-10 and the downregulation of IL-25 and eotaxin which stimulate the release and recruitment of eosinophils to the sites of inflammation synergistically with IL-5. Furthermore, the ASC-derived EVs induced an anti-inflammatory state in Th2-mediated inflammation through polarization to the M1 and M2 macrophages and dendritic cell maturation for effector T cell induction [101].
The functional enhancement of gene analysis and the microRNA expression pattern analysis, which are methods used to identify a set of overexpressed genes or proteins, have been performed, and studies on the expression pattern and differential expression characteristics of specific genes have been reported.
Kim et al. performed DNA microarray to identify the differentially expressed genes (DEGs) related to the suppression of allergic airway inflammation by ASCs-derived EVs. After the hierarchical clustering of DEGs and after the functional and pathway analysis of the potential DEGs, a total of 249 DEGs were identified, of which 21 were upregulated in the EVs group, resulting in more than 2-fold changes compared to that which was seen in the OVA group. These results suggest that PON1, Bex2, Igfbp6, and Scgb1c1 may be involved in the immunosuppressive mechanism mechanisms of MSCs-derived EVs in allergic airway diseases [102].
Paraoxonase1 (PON1) is a calcium-dependent aryldialkylphosphatase belonging to the paraoxonase (PON) family, and it has antioxidant, anti-adhesive, anti-inflammatory, anti-thrombotic, and anti-apoptotic effects. In addition to asthma, various diseases such as diabetes, rheumatism, arthritis, psoriasis, and systemic lupus erythematosus are also associated with PON1 [103,104,105,106,107,108]. The expression and activity of PON1 in asthmatic patients were significantly lower when they were compared to those of the healthy controls [104,109,110]. Furthermore, PON1 reduced the degree of airway inflammation and airway remodeling and inhibited the lipopolysaccharide (LPS)-induced inflammatory cytokine expression and lung fibroblast proliferation in asthmatic mice, thereby having significant potential effects in allergic airway disease [111]. Bex2 is a protein-coding gene known to be involved in carcinogenesis and it is a regulator of mitochondrial apoptosis and the G1 cell cycle, particularly in breast cancer [112]. Although few reports have been reported on allergic diseases, a recent study reported that Bex2 expression was suppressed by the increased DNA methylation of IL-13 which was induced in allergic airway inflammation [113]. Igfbp6 is a family of insulin-like growth factor (IGF) binding proteins related to the growth inhibitory protein regulating the availability of IGFs. The family of proteins binding to IGFs includes Igfbp1, Igfbp2, Igfbp3, Igfbp4, and Igfbp5 in addition to Igfbp6 [114]. The biological functions of Igfbp can be classified into IGF-independent and IGF-dependent actions. In particular, Igfbp has been reported as a biomarker and a therapeutic target acting on the pathogenesis of various autoimmune diseases, and Igfbp6 was associated with fibroblast proliferation and cell growth in asthmatic patients [115,116]. Fpr1 is a group of G protein-coupled cell surface receptors that have important roles in inflammation and host defenses. Since Fpr1 are expressed across a variety of cell types and interact with structurally diverse chemotactic agents, they either accelerate or inhibit the inflammatory processes upon binding to other ligands [117]. In allergic airway disease, Fpr1 has been reported to stimulate neutrophil chemotaxis and inflammatory cytokine production by phagocytes such as dendritic cells and macrophages [118]. Scgb1c1 is a member of the secretoglobin family of secreted proteins which are found in high concentrations in body fluids of the lungs, lacrimal glands, salivary glands, prostate, uterus, and in other tissues. In the human respiratory mucosa, Scgb1c1 is upregulated by IL-4 and IL-13, and it is downregulated by IFN-γ, and it plays an important role in recognizing and clearance of pathogenic microorganisms in the lung epithelial mucosa [119,120,121]. The important pulmonary genes associated with suppression of allergic airway inflammation by MSC-derived EVs are summarized in Table 2.
Although studies on the mechanisms of MSCs-derived EVs on the immunomodulatory effect of ASCs are still lacking, we may present a hypothesized schematic based on previous studies. The intranasal administration of EVs isolated from the MSC secretome, including the exosomes and microvesicles, increases the expression of Bex2, PON1, Scgb1c1, and Igfbp6 in the lung tissues of asthmatic mice. These pulmonary genes induce the expansion of Tregs. Tregs secrete regulatory cytokines such as and IL-10 and TGF-β, which reduce pulmonary eosinophil infiltration, allergy-specific Th2 cytokines (IL-4, IL-5, and IL-13), allergy specific IgG1 and IgE production, allergic rhinitis symptoms, and AHR (Figure 1).

4. Therapeutic Implications of EVs for Allergic Airway Diseases

The ability to control the immune system in both directions is a specific advantage of EVs. To expand, the EVs have an immunomodulatory effect like dendritic cells (DCs) or regulatory T cells do, and on the other hand, they have the ability to enhance immunity like conventional vaccines do.
There are several reports of using EVs for allergic immunotherapy in allergic airway diseases. Mast cells are closely related to allergic immune responses, and they are mainly located in the intestinal or skin barrier, and they are involved in allergic sensitization and inflammation. It can also modulate the immune responses and transport the MHC molecules, which can induce the functional maturation of DCs and play important roles in the functional maturation of DCs and the efficient antigen presentation of the T cells [122]. Furthermore, it has been reported that anti-IgE treatment is the key to the treatment mechanism in allergy treatments, and mast cell-derived exosomes can neutralize IgE through the FcεRI that is displayed on the surface [123].
There is also a report using a virus-like particle-based vaccine model that can induce high protective allergen-specific IgG titers, while reducing the allergenicity of the allergen [124,125]. Several studies have reported that EVs-based vaccines can increase IgG antibody responses by inducing TH1 responses and inducing IFN-γ and TNF-α T cell responses [126,127,128].
The IgE-antigen immune complex, which is also known as IgE-promoted antigen presentation (IgE-FAP), induces CD4+ T cell and antibody responses by binding IgE and foreign antigens [129,130]. IgE-FAP contains the low-affinity receptor CD23 that is expressed on B cells, which negatively regulates the serum IgE levels by inhibiting the IgE response and downregulating the serum IgE levels [131,132,133,134]. Therefore, several studies have reported that CD23-activated B-cell derived EVs can potentially engineer IgE-promoting antigen presentation to promote T cell and antibody responses, while downregulating the serum IgE levels [135,136].
Studies using EVs as advanced therapeutics for asthma are lacking. As mentioned above, the immunomodulatory effect of the EVs have been confirmed at the pathophysiology level and in the histological findings of the asthma, but additional clinical studies are needed.

5. Limitations and Future Prospects of MSC-Derived EVs

Since various MSC-derived bioactive molecules are contained in MSC-derived EVs, it is necessary to secure the safety and efficacy of MSC-derived EVs by conducting additional preclinical and clinical evaluations of EVs in various disease conditions. Through the analysis of these candidate genes as well as big data-based transcriptome and proteome analyses, we may understand the common characteristics of EVs, as well as compare the unique characteristics and advantages of each of the EVs’ origins. Since the unique characteristics and advantages of each EV origin can be compared, the optimized MSC-derived EVs can be applied to an appropriate target. Nevertheless, in order to use the EVs clinically, the following limitations be considered. Firstly, since the EVs are separated from the MSCs culture medium, the culture conditions such as the number of stem cells, the volume of the medium, and the time to separate the EVs are closely related to the quantity and quality of the culture medium. For this reason, it is difficult to secure large-scale EVs derived from MSCs at high concentrations [137]. However, a recent study reported that when they were used with large capacity media, it reduces the risk of vesicle destruction and aggregation with a yield of up to 80% and high reproducibility [138].
In addition, although various methods have been introduced to isolate MSCs-derived EVs from the culture medium, their standardization and optimization are also needed [139]. In this regard, safety regulatory frameworks with good manufacturing practice standards for exosome characterization have been developed by international societies and European networks [140]. Finally, the clinical evaluation of the safety and efficacy of the MSCs-derived EVs for various diseases should be conducted, and additional studies on the precise mechanism of action are needed.

6. Conclusions

Many studies have been reported for the clinical application of MSCs-derived EVs in various diseases. Although the studies on MSCs-derived EVs in allergic airway diseases are still lacking in the literature, previous studies have shown that EVs have the same immunomodulatory effect as stem cells in allergic airway diseases, and they have potential as a new therapeutic agent. We believe that MSCs-derived EVs have a potential mechanism and could be novel therapeutic candidates for the treatment of allergic rhinitis and asthma in the future.

Author Contributions

Conceptualization: K.-S.C., Writing—original draft preparation: S.-D.K., Literature search and data analysis: S.-D.K., Writing—review and editing: K.-S.C., Supervision: K.-S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

This article does not contain any studies with human or animal subjects performed by any of the authors.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Boulay, M.E.; Boulet, L.-P. The relationships between atopy, rhinitis and asthma: Pathophysiological considerations. Curr. Opin. Allergy Clin. Immunol. 2003, 3, 51–55. [Google Scholar] [CrossRef] [PubMed]
  2. Togias, A. Rhinitis and asthma: Evidence for respiratory system integration. J. Allergy Clin. Immunol. 2003, 111, 1171–1183. [Google Scholar] [CrossRef] [PubMed]
  3. Wilson, M.S.; Taylor, M.D.; Balic, A.; Finney, C.A.; Lamb, J.R.; Maizels, R.M. Suppression of allergic airway inflammation by helminth-induced regulatory T cells. J. Exp. Med. 2005, 202, 1199–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shi, H.-Z.; Qin, X.-J. CD4CD25 regulatory T lymphocytes in allergy and asthma. Allergy 2005, 60, 986–995. [Google Scholar] [CrossRef]
  5. Biehl, J.K.; Russell, B. Introduction to Stem Cell Therapy. J. Cardiovasc. Nurs. 2009, 24, 98–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Law, S.; Chaudhuri, S. Mesenchymal stem cell and regenerative medicine: Regeneration versus immunomodulatory challenges. Am. J. Stem Cells 2013, 2, 22–38. [Google Scholar] [PubMed]
  7. Tuan, R.S.; Boland, G.; Tuli, R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res. Ther. 2003, 5, 32–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Uccelli, A.; Moretta, L.; Pistoia, V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 2008, 8, 726–736. [Google Scholar] [CrossRef] [PubMed]
  9. Cho, K.-S. Application of Mesenchymal Stem Cells in Rhinologic Fields. Korean J. Otorhinolaryngol.-Head Neck Surg. 2014, 57, 207–213. [Google Scholar] [CrossRef]
  10. Scuteri, A.; Miloso, M.; Foudah, D.; Orcianni, M.; Cavaletti, G.; Trdeici, G. Mesenchymal stem cells neuronal differentiation ability: A real perspective for nervous system repair? Curr. Stem Cell Res. Ther. 2011, 6, 82–92. [Google Scholar] [CrossRef]
  11. Titorencu, I.; Jinga, V.; Constantinescu, E.; Gafencu, A.; Ciohodaru, C.; Manolescu, I.; Zaharia, C.; Simionescu, M. Proliferation, differentiation and characterization of osteoblasts from human BM mesenchymal cells. Cytotherapy 2007, 9, 682–696. [Google Scholar] [CrossRef]
  12. Bhagavati, S.; Xu, W. Isolation and enrichment of skeletal muscle progenitor cells from mouse bone marrow. Biochem. Biophys. Res. Commun. 2004, 318, 119–124. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, T.M.; Martina, M.; Hutmacher, D.W.; Hui, J.H.P.; Lee, E.H.; Lim, B. Identification of Common Pathways Mediating Differentiation of Bone Marrow- and Adipose Tissue-Derived Human Mesenchymal Stem Cells into Three Mesenchymal Lineages. Stem Cells 2007, 25, 750–760. [Google Scholar] [CrossRef] [PubMed]
  14. Delorme, B.; Charbord, P. Culture and Characterization of Human Bone Marrow Mesenchymal Stem Cells. Methods Mol. Med. 2007, 140, 67–81. [Google Scholar] [CrossRef] [PubMed]
  15. Ma, S.; Xie, N.; Li, W.; Yuan, B.; Shi, Y.; Wang, Y. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014, 21, 216–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Shi, M.; Liu, Z.W.; Wang, F.S. Imunomodulatory properties and therapeutic application of mesenchymal stem cells. Clin. Exp. Immunol. 2011, 164, 1–8. [Google Scholar] [CrossRef]
  17. Kim, H.; Shin, T.; Lee, B.; Yu, K.; Seo, Y.; Lee, S.; Seo, M.; Hong, I.; Choi, S.W.; Seo, K.; et al. Human Umbilical Cord Blood Mesenchymal Stem Cells Reduce Colitis in Mice by Activating NOD2 Signaling to COX2. Gastroenterology 2013, 145, 1392–1403.e8. [Google Scholar] [CrossRef]
  18. Németh, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E2–dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42–49. [Google Scholar] [CrossRef] [Green Version]
  19. González, M.A.; Gonzalez–Rey, E.; Rico, L.; Büscher, D.; Delgado, M. Adipose-Derived Mesenchymal Stem Cells Alleviate Experimental Colitis by Inhibiting Inflammatory and Autoimmune Responses. Gastroenterology 2009, 136, 978–989. [Google Scholar] [CrossRef]
  20. Augello, A.; Tasso, R.; Negrini, S.M.; Cancedda, R.; Pennesi, G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007, 56, 1175–1186. [Google Scholar] [CrossRef]
  21. Lee, R.H.; Seo, M.J.; Reger, R.L.; Spees, J.L.; Pulin, A.A.; Olson, S.D.; Prockop, D.J. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc. Natl. Acad. Sci. USA 2006, 103, 17438–17443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zappia, E.; Casazza, S.; Pedemonte, E.; Benvenuto, F.; Bonanni, I.; Gerdoni, E.; Giunti, D.; Ceravolo, A.; Cazzanti, F.; Frassoni, F.; et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 2005, 106, 1755–1761. [Google Scholar] [CrossRef] [Green Version]
  23. Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Götherström, C.; Hassan, M.; Uzunel, M.; Ringdén, O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004, 363, 1439–1441. [Google Scholar] [CrossRef] [PubMed]
  24. Cho, K.-S.; Park, M.-K.; Kang, S.-A.; Park, H.-Y.; Hong, S.-L.; Park, H.-K.; Yu, H.-S.; Roh, H.-J. Adipose-Derived Stem Cells Ameliorate Allergic Airway Inflammation by Inducing Regulatory T Cells in a Mouse Model of Asthma. Mediat. Inflamm. 2014, 2014, 1–12. [Google Scholar] [CrossRef]
  25. Cho, K.-S.; Lee, J.-H.; Park, M.-K.; Park, H.-K.; Yu, H.-S.; Roh, H.-J. Prostaglandin E2 and Transforming Growth Factor-β Play a Critical Role in Suppression of Allergic Airway Inflammation by Adipose-Derived Stem Cells. PLoS ONE 2015, 10, e0131813. [Google Scholar] [CrossRef] [Green Version]
  26. Cho, K.-S.; Park, M.-K.; Mun, S.-J.; Park, H.-Y.; Yu, H.-S.; Roh, H.-J. Indoleamine 2,3-Dioxygenase Is Not a Pivotal Regulator Responsible for Suppressing Allergic Airway Inflammation through Adipose-Derived Stem Cells. PLoS ONE 2016, 11, e0165661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Goodwin, M.; Sueblinvong, V.; Eisenhauer, P.; Ziats, N.P.; LeClair, L.; Poynter, M.E.; Steele, C.; Rincon, M.; Weiss, D.J. Bone Marrow-Derived Mesenchymal Stromal Cells Inhibit Th2-Mediated Allergic Airways Inflammation in Mice. Stem Cells 2011, 29, 1137–1148. [Google Scholar] [CrossRef] [Green Version]
  28. Nemeth, K.; Keane-Myers, A.; Brown, J.M.; Metcalfe, D.D.; Gorham, J.D.; Bundoc, V.G.; Hodges, M.G.; Jelinek, I.; Madala, S.; Karpati, S.; et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc. Natl. Acad. Sci. USA 2010, 107, 5652–5657. [Google Scholar] [CrossRef] [Green Version]
  29. Beyth, S.; Borovsky, Z.; Mevorach, D.; Liebergall, M.; Gazit, Z.; Aslan, H.; Galun, E.; Rachmilewitz, J. Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood 2005, 105, 2214–2219. [Google Scholar] [CrossRef] [Green Version]
  30. Desai, M.B.; Gavrilova, T.; Liu, J.; Patel, S.A.; Kartan, S.; Greco, S.J.; Capitle, E.; Rameshwar, P. Pollen-induced antigen presentation by mesenchymal stem cells and T cells from allergic rhinitis. Clin. Transl. Immunol. 2013, 2, e7. [Google Scholar] [CrossRef]
  31. Barkholt, L.; Flory, E.; Jekerle, V.; Lucas-Samuel, S.; Ahnert, P.; Bisset, L.; Büscher, D.; Fibbe, W.; Foussat, A.; Kwa, M.; et al. Risk of tumorigenicity in mesenchymal stromal cell-based therapies bridning scientific observations and regulatory viewpoints. Cytotherapy 2013, 15, 753–759. [Google Scholar] [CrossRef] [PubMed]
  32. Yu, H.S.; Park, M.-K.; Kang, S.A.; Cho, K.-S.; Mun, S.J.; Roh, H.-J. Culture supernatant of adipose stem cells can ameliorate allergic airway inflammation via recruitment of CD4+CD25+Foxp3 T cells. Stem Cell Res. Ther. 2017, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ahmadi, M.; Rahbarghazi, R.; Aslani, M.R.; Shahbazfar, A.-A.; Kazemi, M.; Keyhanmanesh, R. Bone marrow mesenchymal stem cells and their conditioned media could potentially ameliorate ovalbumin-induced asthmatic changes. Biomed. Pharmacother. 2017, 85, 28–40. [Google Scholar] [CrossRef]
  34. Ionescu, L.I.; Alphonse, R.S.; Arizmendi, N.; Morgan, B.; Abel, M.; Eaton, F.; Duszyk, M.; Vliagoftis, H.; Aprahamian, T.R.; Walsh, K.; et al. Airway Delivery of Soluble Factors from Plastic-Adherent Bone Marrow Cells Prevents Murine Asthma. Am. J. Respir. Cell Mol. Biol. 2012, 46, 207–216. [Google Scholar] [CrossRef] [Green Version]
  35. Basu, J.; Ludlow, J.W. Exosomes for repair, regeneration and rejuvenation. Expert. Opin. Biol. Ther. 2016, 16, 489–506. [Google Scholar] [CrossRef]
  36. 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. 2011, 68, 2667–2688. [Google Scholar] [CrossRef] [Green Version]
  37. Hornick, N.I.; Huan, J.; Doron, B.; Goloviznina, N.A.; Lapidus, J.; Chang, B.H.; Kurre, P. Serum Exosome MicroRNA as a Minimally-Invasive Early Biomarker of AML. Sci. Rep. 2015, 5, 11295. [Google Scholar] [CrossRef] [Green Version]
  38. Akers, J.C.; Ramakrishnan, V.; Kim, R.; Skog, J.; Nakano, I.; Pingle, S.; Kalinina, J.; Hua, W.; Kesari, S.; Mao, Y.; et al. miR-21 in the extracellular vesicles (EVs) of Cerebrospinal Fluid (CSF): A Platform for Glioblastoma Biomarker Development. PLoS ONE 2013, 8, e78115. [Google Scholar] [CrossRef]
  39. Shi, R.; Wang, P.-Y.; Li, X.-Y.; Chen, J.-X.; Li, Y.; Zhang, X.-Z.; Zhang, C.-G.; Jiang, T.; Li, W.-B.; Ding, W.; et al. Exosomal levels of miRNA-21 from cerebrospinal uids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 2015, 6, 26971. [Google Scholar] [CrossRef] [Green Version]
  40. Goto, T.; Fujiya, M.; Konishi, H.; Sasajima, J.; Fujibayashi, S.; Hayashi, A.; Utsumi, T.; Sato, H.; Iwama, T.; Ijiri, M.; et al. An elevated expression of serum exosomal microRNA-191, -21, -451a of pancreatic neoplasm is considered to be efficient diagnostic marker. BMC Cancer 2018, 18, 116. [Google Scholar] [CrossRef]
  41. Zlotogorski-Hurvitz, A.; Dayan, D.; Chaushu, G.; Korvala, J.; Salo, T.; Sormunen, R.; Vered, M. Human saliva-derived exosomes: Comparing methods of isolation. J. Histochem. Cytochem. 2015, 63, 181–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yuan, Z.; Bedi, B.; Sadikot, R.T. Bronchoalveolar Lavage Exosomes in Lipopolysaccharide-induced Septic Lung Injury. J. Vis. Exp. 2018, 135, e57737. [Google Scholar] [CrossRef] [Green Version]
  43. Dixon, C.L.; Sheller-Miller, S.; Saade, G.R.; Fortunato, S.J.; Lai, A.; Palma, C.; Guanzon, D.; Salomon, C.; Menon, R. Amniotic Fluid Exosome Proteomic Profile Exhibits Unique Pathways of Term and Preterm Labor. Endocrinology 2018, 159, 2229–2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Li, Z.; Wang, Y.; Xiao, K.; Xiang, S.; Li, Z.; Weng, X. Emerging Role of Exosomes in the Joint Diseases. Cell. Physiol. Biochem. 2018, 47, 2008–2017. [Google Scholar] [CrossRef] [PubMed]
  45. Grigor’Eva, A.E.; Tamkovich, S.N.; Eremina, A.V.; Tupikin, A.E.; Kabilov, M.R.; Chernykh, V.V.; Vlassov, V.V.; Laktionov, P.P.; Ryabchikova, E.I.; Kabilov, M. Exosomes in tears of healthy individuals: Isolation, identification, and characterization. Biochem. Suppl. Ser. B Biomed. Chem. 2016, 10, 165–172. [Google Scholar] [CrossRef]
  46. Milasan, A.; Tessandier, N.; Tan, S.; Brisson, A.; Boilard, E.; Martel, C. Extracellular vesicles are present in mouse lymph and their level di_ers in atherosclerosis. J. Extracell. Vesicles 2016, 5, 76. [Google Scholar] [CrossRef] [Green Version]
  47. Yoon, S.B.; Chang, J.H. Extracellular vesicles in bile: A game changer in the diagnosis of indeterminate biliary stenoses? HepatoBiliary Surg. Nutr. 2017, 6, 408–410. [Google Scholar] [CrossRef] [Green Version]
  48. Yoshida, Y.; Yamamoto, H.; Morita, R.; Oikawa, R.; Matsuo, Y.; Maehata, T.; Nosho, K.; Watanabe, Y.; Yasuda, H.; Itoh, F. Detection of DNA methylation of gastric juice-derived exosomes in gastric cancer. Integr. Mol. Med. 2014, 1, 17–21. [Google Scholar]
  49. Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  50. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
  51. Wollert, T.; Hurley, J.H. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature 2010, 464, 864–869. [Google Scholar] [CrossRef] [PubMed]
  52. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
  53. Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bobrie, A.; Colombo, M.; Raposo, G.; Théry, C. Exosome Secretion: Molecular Mechanisms and Rolesin Immune Responses. Traffic 2011, 12, 1659–1668. [Google Scholar] [CrossRef]
  55. Chaput, N.; Théry, C. Exosomes: Immune properties and potential clinical implementations. Semin. Immunopathol. 2011, 33, 419–440. [Google Scholar] [CrossRef]
  56. Gomes, C.; Keller, S.; Altevogt, P.; Costa, J. Evidence for secretion of Cu, Zn superoxide dismutase via exosomes from a cell model of amyotrophic lateral sclerosis. Neurosci. Lett. 2007, 428, 43–46. [Google Scholar] [CrossRef]
  57. Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Ntzouni, M.; Stefanis, L.; Vekrellis, K.; Garbis, S.D.; Margaritis, L.H. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851. [Google Scholar] [CrossRef] [Green Version]
  58. Borges, F.T.; Melo, S.A.; Özdemir, B.C.; Kato, N.; Revuelta, L.; Miller, C.A.; Gattone, V.H.; LeBleu, V.S.; Kalluri, R. TGF-beta1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J. Am. Soc. Nephrol. 2013, 24, 385–392. [Google Scholar] [CrossRef] [Green Version]
  59. Al-Nedawi, K.; Meehan, B.; Rak, J. Microvesicles: Messengers and mediators of tumor progression. Cell Cycle 2009, 8, 2014–2018. [Google Scholar] [CrossRef]
  60. Simpson, R.J.; Lim, J.W.; Moritz, R.L.; Mathivanan, S. Exosomes: Proteomic insights and diagnostic potential. Expert Rev. Proteom. 2009, 6, 267–283. [Google Scholar] [CrossRef]
  61. Zhou, H.; Pisitkun, T.; Aponte, A.; Yuen, P.; Hoffert, J.; Yasuda, H.; Hu, X.; Chawla, L.; Shen, R.-F.; Knepper, M.; et al. Exosomal Fetuin-A identified by proteomics: A novel urinary biomarker for detecting acute kidney injury. Kidney Int. 2006, 70, 1847–1857. [Google Scholar] [CrossRef] [PubMed]
  62. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sandfeld-Paulsen, B.; Aggerholm-Pedersen, N.; Baek, R.; Jakobsen, K.R.; Meldgaard, P.; Folkersen, B.H.; Rasmussen, T.R.; Varming, K.; Jørgensen, M.M.; Sorensen, B.S. Exosomal proteins as prognostic biomarkers in non-small cell lung cancer. Mol. Oncol. 2016, 10, 1595–1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sonoda, H.; Yokota-Ikeda, N.; Oshikawa, S.; Kanno, Y.; Yoshinaga, K.; Uchida, K.; Ueda, Y.; Kimiya, K.; Uezono, S.; Ueda, A.; et al. Decreased abundance of urinary exosomal aquaporin-1 in renal ischemia-reperfusion injury. Am. J. Physiol. Ren. Physiol. 2009, 297, F1006–F1016. [Google Scholar] [CrossRef] [Green Version]
  65. Pitt, J.M.; Charrier, M.; Viaud, S.; André, F.; Besse, B.; Chaput, N.; Zitvogel, L. Dendritic Cell–Derived Exosomes as Immunotherapies in the Fight against Cancer. J. Immunol. 2014, 193, 1006–1011. [Google Scholar] [CrossRef] [Green Version]
  66. Lai, R.C.; Yeo, R.W.Y.; Tan, K.H.; Lim, S.K. Exosomes for drug delivery—A novel application for the mesenchymal stem cell. Biotechnol. Adv. 2013, 31, 543–551. [Google Scholar] [CrossRef]
  67. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brainby systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
  68. Reis, L.A.; Borges, F.T.; Simões, M.J.; Borges, A.A.; Sinigaglia-Coimbra, R.; Schor, N. Bone Marrow-Derived Mesenchymal Stem Cells Repaired but Did Not Prevent Gentamicin-Induced Acute Kidney Injury through Paracrine E_ects in Rats. PLoS ONE 2012, 7, e44092. [Google Scholar] [CrossRef] [Green Version]
  69. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles Derived from Mesenchymal Stem Cells Enhance Survival in a Lethal Model of Acute Kidney Injury. PLoS ONE 2012, 7, e33115. [Google Scholar] [CrossRef]
  70. Akao, Y.; Iio, A.; Itoh, T.; Noguchi, S.; Itoh, Y.; Ohtsuki, Y.; Naoe, T. Microvesicle-mediated RNA Molecule Delivery System Using Monocytes/Macrophages. Mol. Ther. 2011, 19, 395–399. [Google Scholar] [CrossRef] [Green Version]
  71. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.-C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. J. Clin. Investig. 2018, 3, e99263. [Google Scholar] [CrossRef] [PubMed]
  72. Zhu, X.; Badawi, M.; Pomeroy, S.; Sutaria, D.S.; Xie, Z.; Baek, A.; Jiang, J.; Elgamal, O.A.; Mo, X.; Perle, K.; et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. J. Extracell. Vesicles 2017, 6, 1324730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ingato, D.; Lee, J.U.; Sim, S.J.; Kwon, Y.J. Good things come in small packages: Overcoming challenges to harness extracellular vesicles for therapeutic delivery. J. Control. Release 2016, 241, 174–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kooijmans, S.A.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R. Modulation of tissue tropism and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer treatment. Pharmacol. Res. 2016, 111, 487–500. [Google Scholar] [CrossRef] [PubMed]
  75. Østergaard, O.; Nielsen, C.T.; Iversen, L.V.; Jacobsen, S.; Tanassi, J.T.; Heegaard, N.H.H. Quantitative Proteome Profiling of Normal Human Circulating Microparticles. J. Proteome Res. 2012, 11, 2154–2163. [Google Scholar] [CrossRef]
  76. Zöller, M. Tetraspanins: Push and pull in suppressing and promoting metastasis. Nat. Rev. Cancer 2009, 9, 40–55. [Google Scholar] [CrossRef]
  77. Harding, C.V.; Heuser, J.E.; Stahl, P.D. Exosomes: Looking back three decades and into the future. J. Cell Biol. 2013, 200, 367–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Wickman, G.; Julian, L.; Olson, M.F.; Olson, M. How apoptotic cells aid in the removal of their own cold dead bodies. Cell Death Differ. 2012, 19, 735–742. [Google Scholar] [CrossRef] [Green Version]
  79. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [Green Version]
  80. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  81. Mathivanan, S.; Ji, H.; Simpson, R.J. Exosomes: Extracellular organelles important in intercellular communication. J. Proteom. 2010, 73, 1907–1920. [Google Scholar] [CrossRef] [PubMed]
  82. Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowskawieczorek, A.; Ratajczak, M.Z. Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  83. Gan, L.; Xie, D.; Liu, J.; Lau, W.B.; Christopher, T.A.; Lopez, B.; Zhang, L.; Gao, E.; Koch, W.; Ma, X.-L.; et al. Small Extracellular Microvesicles Mediated Pathological Communications Between Dysfunctional Adipocytes and Cardiomyocytes as a Novel Mechanism Exacerbating Ischemia/Reperfusion Injury in Diabetic Mice. Circulation 2020, 141, 968–983. [Google Scholar] [CrossRef]
  84. Wang, Q.; Sun, B.; Wang, D.; Ji, Y.; Kong, Q.; Wang, G.; Wang, J.; Zhao, W.; Jin, L.; Li, H. Murine bone marrow mesenchymal stem cells cause mature dendtiric cells to promote T-cell tolerance. Scand. J. Immunol. 2008, 68, 607–615. [Google Scholar] [CrossRef] [PubMed]
  85. Wu, X.; Liu, Z.; Hu, L.; Gu, W.; Zhu, L. Exosomes derived from endothelial progenitor cells ameliorate acute lung injury by transferring miR-126. Exp. Cell Res. 2018, 370, 13–23. [Google Scholar] [CrossRef]
  86. Zhuansun, Y.; Du, Y.; Huang, F.; Lin, L.; Chen, R.; Jiang, S.; Li, J. MSCs exosomal miR-1470 promotes the differentiation of CD4(+)CD25(+)FOXP3(+) Tregs in asthmatic patients by inducing the expression of P27KIP1. Int. Immunopharmacol. 2019, 77, 105981. [Google Scholar] [CrossRef]
  87. Fang, S.B.; Zhang, H.Y.; Wang, C.; He, B.X.; Liu, X.Q.; Meng, X.C.; Peng, Y.Q.; Xu, Z.B.; Fan, X.L.; Wu, Z.J.; et al. Small extracellular vesicles derived from human mesenchymal stromal cells prevent group 2 innate lymphoid cell-dominant allergic airway inflammation through delivery of mir-146a-5p. J. Extracell Vesicles 2020, 9, 1723260. [Google Scholar] [CrossRef] [Green Version]
  88. 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-1b-primed mesenchymal stem cells against sepsis. Stem Cells 2017, 35, 1208–1221. [Google Scholar] [CrossRef] [Green Version]
  89. Hao, Q.; Gudapati, V.; Monsel, A.; Park, J.H.; Ho, S. Mesenchymal stem cell-derived extracellular vesciles decrese lung injury in Mice. J. Immunol. 2019, 203, 1961–1972. [Google Scholar] [CrossRef]
  90. 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]
  91. Yi, X.; Wei, X.; Lv, H.; An, Y.; Li, L.; Lu, P.; Yang, Y.; Zhang, Q.; Yi, H.; Chen, G. Exosomes derived from microRNA-30b-3p-overexpressing mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting SAA3. Exp. Cell Res. 2019, 383, 111454. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, W.-X.; Zhou, J.; Zhou, S.-S.; Zhang, Y.-D.; Ji, T.-Y.; Zhang, X.-L.; Wang, S.-M.; Du, T.; Ding, D.-G. Microvesicles derived from human Wharton’s jelly mesenchymal stem cells enhance autophagy and ameliorate acute lung injury via delivery of miR-100. Stem Cell Res. Ther. 2020, 11, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wang, J.; Huang, R.; Xu, Q.; Zheng, G.; Qiu, G.; Ge, M.; Shu, Q.; Xu, J. Mesenchymal Stem Cell–Derived Extracellular Vesicles Alleviate Acute Lung Injury Via Transfer of miR-27a-3p. Crit. Care Med. 2020, 48, e599–e610. [Google Scholar] [CrossRef]
  94. Zhang, C.; Wang, P.; Mohammed, A.; Zhou, Z.; Zhang, S.; Ni, S.; Tang, Z. Function of Adiopse-derived mesenchymal stem cells in monocrotalineinduced pulmonary arterial hypertension through miR-191 via regulation of BMPR2. BioMed Res. Int. 2020, 2019, 2858750. [Google Scholar]
  95. Ge, X.; Bai, C.; Yang, J.; Lou, G.; Li, Q.; Chen, R. Effect of mesenchymal stem cells on inhibiting airway remodeling and airway inflammation in chronic asthma. J. Cell. Biochem. 2013, 114, 1595–1605. [Google Scholar] [CrossRef] [PubMed]
  96. Ge, X.; Bai, C.; Yang, J.; Lou, G.; Li, Q.; Chen, R. Intratracheal transplantation of bone marrow-derived mesenchymal stem cells reduced airway inflammation and up-regulated CD4+CD25+regulatory T cells in asthmatic mouse. Cell Biol. Int. 2013, 37, 675–686. [Google Scholar] [CrossRef]
  97. Fu, Q.L.; Chow, Y.Y.; Sun, S.J.; Zeng, Q.X.; Li, H.B.; Shi, J.B.; Sun, Y.-Q.; Wen, W.; Tse, H.F.; Lian, Q.; et al. Mesenchymal stem cells derived from human induced pluripotent stem cells modulate T-cell phenotypes in allergic rhinitis. Allergy 2012, 67, 1215–1222. [Google Scholar] [CrossRef]
  98. Mun, S.J.; Kang, S.A.; Park, H.-K.; Yu, H.S.; Cho, K.-S.; Roh, H.-J. Intranasally Administered Extracellular Vesicles from Adipose Stem Cells Have Immunomodulatory Effects in a Mouse Model of Asthma. Stem Cells Int. 2021, 2021, 1–11. [Google Scholar] [CrossRef]
  99. Cruz, F.F.; Borg, Z.D.; Goodwin, M.; Sokocevic, D.; Wagner, D.E.; Coffey, A.; Antunes, M.; Robinson, K.L.; Mitsialis, S.A.; Kourembanas, S.; et al. Systemic Administration of Human Bone Marrow-Derived Mesenchymal Stromal Cell Extracellular Vesicles Ameliorates Aspergillus Hyphal Extract-Induced Allergic Airway Inflammation in Immunocompetent Mice. Stem Cells Transl. Med. 2015, 4, 1302–1316. [Google Scholar] [CrossRef] [Green Version]
  100. de Castro, L.L.; Xisto, D.G.; Kitoko, J.Z.; Cruz, F.F.; Olsen, P.C.; Redondo, P.A.G.; Ferreira, T.P.T.; Weiss, D.J.; Martins, M.A.; Morales, M.M.; et al. Human adipose tissue mesenchymal stromal cells and their extracellular vesicles act differentially on lung mechanics and inflammation in experimental allergic asthma. Stem Cell Res. Ther. 2017, 8, 151. [Google Scholar] [CrossRef] [Green Version]
  101. Cho, K.-S.; Kang, S.A.; Kim, S.-D.; Mun, S.-J.; Yu, H.S.; Roh, H.-J. Dendritic cells and M2 macrophage play an important role in suppression of Th2-mediated inflammation by adipose stem cells-derived extracellular vesicles. Stem Cell Res. 2019, 39, 101500. [Google Scholar] [CrossRef] [PubMed]
  102. Kim, S.-D.; Kang, S.A.; Kim, Y.-W.; Yu, H.S.; Cho, K.-S.; Roh, H.-J. Screening and Functional Pathway Analysis of Pulmonary Genes Associated with Suppression of Allergic Airway Inflammation by Adipose Stem Cell-Derived Extracellular Vesicles. Stem Cells Int. 2020, 27, 5684250. [Google Scholar] [CrossRef] [PubMed]
  103. Bahrehmand, F.; Vaisi-Raygani, A.; Rahimi, Z.; Ahmadi, R.; Kiani, A.; Tavilani, H.; Pourmotabbed, T. Synergistic effects of BuChE non-UU phenotype and paraoxonase (PON1) 55 M allele on the risk of systemic lupus erythematosus: Influence on lipid and lipoprotein metabolism and oxidative stress, preliminary report. Lupus 2014, 23, 263–272. [Google Scholar] [CrossRef] [PubMed]
  104. Sarioglu, N.; Hismiogullari, A.A.; Erel, F.; Demir, D.; Gencer, N. Paraoxonase 1 phenotype and paraoxonase activity in asthmatic patients. Iran. J. Allergy Asthma Immunol. 2015, 14, 60–66. [Google Scholar]
  105. Tanimoto, N.; Kumon, Y.; Suehiro, T.; Ohkubo, S.; Ikeda, Y.; Nishiya, K.; Hashimoto, K. Serum paraoxonase activity decreases in rheumatoid arthritis. Life Sci. 2003, 72, 2877–2885. [Google Scholar] [CrossRef]
  106. Asefi, M.; Vaisi-Raygani, A.; Bahrehmand, F.; Kiani, A.; Rahimi, Z.; Nomani, H.; Ebrahimi, A.; Tavilani, H.; Pourmotabbed, T. Paraoxonase 1 (PON1) 55 polymorphism, lipid profiles and psoriasis. Br. J. Dermatol. 2012, 167, 1279–1286. [Google Scholar] [CrossRef]
  107. Berg, S.W.V.D.; Jansen, E.H.J.; Kruijshoop, M.; Beekhof, P.K.; Blaak, E.; Van Der Kallen, C.J.; Van Greevenbroek, M.M.; Feskens, E.J.M. Paraoxonase 1 phenotype distribution and activity differs in subjects with newly diagnosed Type 2 diabetes (the CODAM Study). Diabet. Med. 2008, 25, 186–193. [Google Scholar] [CrossRef]
  108. Isik, A.; Koca, S.S.; Ustundag, B.; Celik, H.; Yildirim, A. Paraoxonase and arylesterase levels in rheumatoid arthritis. Clin. Rheumatol. 2007, 26, 342–348. [Google Scholar] [CrossRef]
  109. Tölgyesi, G.; Molnár, V.; Semsei, Á.F.; Kiszel, P.; Ungvári, I.; Pócza, P.; Wiener, Z.; Komlósi, Z.I.; Kunos, L.; Gálffy, G.; et al. Gene expression profiling of experimental asthma reveals a possible role of paraoxonase-1 in the disease. Int. Immunol. 2009, 21, 967–975. [Google Scholar] [CrossRef] [Green Version]
  110. Emin, O.; Hasan, A.; Rusen, D. Plasma paraoxonase, oxidative status level, and their relationship with asthma control test in children with asthma. Allergol. Immunopathol. 2015, 43, 346–352. [Google Scholar] [CrossRef]
  111. Chen, W.; Xie, Z.; Wang, X.; Zhao, J.; Hu, Q.; Chen, Y.; Gao, W.; Liu, Y. Influences of PON1 on airway inflammation and remodeling in bronchial asthma. J. Cell. Biochem. 2018, 119, 793–805. [Google Scholar] [CrossRef]
  112. Naderi, A.; Liu, J.; Bennett, I.C. BEX2 regulates mitochondrial apoptosis and G1 cell cycle in breast cancer. Int. J. Cancer 2010, 126, 1596–1610. [Google Scholar] [CrossRef]
  113. Ooi, A.T.; Ram, S.; Kuo, A.; Gilbert, J.L.; Yan, W.; Pellegrini, M.; Nickerson, D.W.; Chatila, T.; Gomperts, B.N. Identification of an interleukin 13-induced epigenetic signature in allergic airway inflammation. Am. J. Transl. Res. 2012, 4, 219–228. [Google Scholar] [PubMed]
  114. Ding, H.; Wu, T. Insulin-Like Growth Factor Binding Proteins in Autoimmune Diseases. Front. Endocrinol. 2018, 9, 499. [Google Scholar] [CrossRef] [Green Version]
  115. Vaillancourt, V.T.; Bordeleau, M.; Laviolette, M.; Laprise, C. From expression pattern to genetic association in asthma and asthma-related phenotypes. BMC Res. Notes 2012, 5, 630. [Google Scholar] [CrossRef] [Green Version]
  116. Kostecka, Y.; Blahovec, J. Insulin-like growth factor binding proteins and their biological functions (mini review). Endocr. Regul. 1999, 33, 90–94. [Google Scholar] [PubMed]
  117. Filina, Y.; Tikhonova, I.; Gabdoulkhakova, A.; Rizvanov, A.; Safronova, V. Mechanisms of ERK phosphorylation triggered via mouse formyl peptide receptor 2. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119356. [Google Scholar] [CrossRef] [PubMed]
  118. Cardini, S.; Dalli, J.; Fineschi, S.; Perretti, M.; Lungarella, G.; Lucattelli, M. Genetic Ablation of the Fpr1 Gene Confers Protection from Smoking-Induced Lung Emphysema in Mice. Am. J. Respir. Cell Mol. Biol. 2012, 47, 332–339. [Google Scholar] [CrossRef]
  119. Jackson, B.C.; Thompson, D.C.; Wright, M.; McAndrews, M.; Bernard, A.; Nebert, D.W.; Vasiliou, V. Update of the human secretoglobin (SCGB) gene superfamily and an example of ‘evolutionary bloom’ of androgen-binding protein genes within the mouse Scgb gene superfamily. Hum. Genom. 2011, 5, 691–702. [Google Scholar] [CrossRef]
  120. Orysiak, J.; Malczewska-Lenczowska, J.; Bik-Multanowski, M. Expression of SCGB1C1 gene as a potential marker of susceptibility to upper respiratory tract infections in elite athletes—A pilot study. Biol. Sport 2016, 33, 107–110. [Google Scholar] [CrossRef] [Green Version]
  121. Sjödin, A.; Guo, D.; Sørhaug, S.; Bjermer, L.; Henriksson, R.; Hedman, H. Dysregulated secretoglobin expression in human lung cancers. Lung Cancer 2003, 41, 49–56. [Google Scholar] [CrossRef] [PubMed]
  122. Skokos, D.; Botros, H.G.; Demeure, C.; Morin, J.; Peronet, R.; Birkenmeier, G.; Boudaly, S.; Mécheri, S. Mast Cell-Derived Exosomes Induce Phenotypic and Functional Maturation of Dendritic Cells and Elicit Specific Immune Responses In Vivo. J. Immunol. 2003, 170, 3037–3045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. 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]
  124. Engeroff, P.; Caviezel, F.; Storni, F.; Thoms, F.; Vogel, M.; Bachmann, M.F. Allergens displayed on virus-like particles are highly immunogenic but fail to activate human mast cells. Allergy 2018, 73, 341–349. [Google Scholar] [CrossRef] [PubMed]
  125. Storni, F.; Zeltins, A.; Balke, I.; Heath, M.D.; Kramer, M.F.; Skinner, M.A.; Zha, L.; Roesti, E.; Engeroff, P.; Muri, L.; et al. Vaccine against peanut allergy based on engineered virus-like particles displaying single major peanut allergens. J. Allergy Clin. Immunol. 2020, 145, 1240–1253.e3. [Google Scholar] [CrossRef] [PubMed]
  126. Jesus, S.; Soares, E.; Cruz, M.T.; Borges, O. Exosomes as adjuvants for the recombinant hepatitis B antigen: First report. Eur. J. Pharm. Biopharm. 2018, 133, 1–11. [Google Scholar] [CrossRef]
  127. Wahlund, C.; Güclüler, G.; Hiltbrunner, S.; Veerman, R.E.; Näslund, T.I.; Gabrielsson, S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
  128. Colino, J.; Snapper, C.M. Exosomes from bone marrow dendritic cells pulsed with diphtheria toxoid preferentially induce type 1 antigen-specific IgG responses in naive recipients in the absence of free antigen. J. Immunol. 2006, 177, 3757–3762. [Google Scholar] [CrossRef] [Green Version]
  129. Hjelm, F.; Karlsson, M.C.I.; Heyman, B. A Novel B Cell-Mediated Transport of IgE-Immune Complexes to the Follicle of the Spleen. J. Immunol. 2008, 180, 6604–6610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Getahun, A.; Hjelm, F.; Heyman, B. IgE Enhances Antibody and T Cell Responses In Vivo via CD23+ B Cells. J. Immunol. 2005, 175, 1473–1482. [Google Scholar] [CrossRef] [Green Version]
  131. Yu, P.; Kosco-Vilbois, M.; Richards, M.; Köhler, G.; Lamers, M.C. Negative feedback regulation of IgE synthesis by murine CD23. Nature 1994, 369, 753–756. [Google Scholar] [CrossRef] [PubMed]
  132. Engeroff, P.; Plattner, K.; Storni, F.; Thoms, F.; Boligan, K.F.; Muerner, L.; Eggel, A.; von Gunten, S.; Bachmann, M.F.; Vogel, M. Glycan-specific IgG anti-IgE autoantibodies are protective against allergic anaphylaxis in a murine model. J. Allergy Clin. Immunol. 2021, 147, 1430–1441. [Google Scholar] [CrossRef] [PubMed]
  133. Engeroff, P.; Caviezel, F.; Mueller, D.; Thoms, F.; Bachmann, M.F.; Vogel, M. CD23 provides a noninflammatory pathway for IgE-allergen complexes. J. Allergy Clin. Immunol. 2020, 145, 301–311.e4. [Google Scholar] [CrossRef] [PubMed]
  134. Fellmann, M.; Buschor, P.; Röthlisberger, S.; Zellweger, F.; Vogel, M. High affinity targeting of CD23 inhibits IgE synthesis in human B cells. Immun. Inflamm. Dis. 2015, 3, 339–349. [Google Scholar] [CrossRef] [Green Version]
  135. Martin, R.K.; Brooks, K.B.; Henningsson, F.; Heyman, B.; Conrad, D.H. Antigen Transfer from Exosomes to Dendritic Cells as an Explanation for the Immune Enhancement Seen by IgE Immune Complexes. PLoS ONE 2014, 9, e110609. [Google Scholar] [CrossRef]
  136. Engeroff, P.; Vogel, M. The role of CD23 in the regulation of allergic responses. Allergy 2021, 76, 1981–1989. [Google Scholar] [CrossRef]
  137. Eirin, A.; Riester, S.M.; Zhu, X.Y.; Tang, H.; Evans, J.M.; O’Brien, D.; van Wijnen, A.J.; Lerman, L.O. MicroRNA and mRNA cargo of extracellular vesicule from porcrine adipose tissue-derived mesenchymal stem cells. Gene 2014, 551, 55–64. [Google Scholar] [CrossRef] [Green Version]
  138. Corso, G.; Mäger, I.; Lee, Y.; Görgens, A.; Bultema, J.; Giebel, B.; Wood, M.J.A.; Nordin, J.Z.; El Andaloussi, S. Reproducible and scalable purification of extracellular vesicles using combined bind-elute and size exclusion chromatography. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef]
  139. Tomasoni, S.; Longaretti, L.; Rota, C.; Morigi, M.; Conti, S.; Gotti, E.; Capelli, C.; Introna, M.; Remuzzi, G.; Benigni, A. Transfer of growh factor receptor mRNA via exosomes unravels the regenerative effect of mesenchymal stem cells. Stem Cells Dev. 2013, 22, 772–780. [Google Scholar] [CrossRef] [Green Version]
  140. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic presentation of plausible mechanisms by which MSC-derived EVs regulate the allergic airway diseases. MSC-derived EVs carry microRNAs such as miR-146a-5p, miR-1470, and miR-126-3p and deliver them into lung tissues. Intranasal administration of MSC-derived EVs increases the expression of PON1, Bex2, Igfbp6, and Scgb1c1 in lung tissues of asthmatic mice. These microRNAs and pulmonary genes by MSC-derived EVs induce the expansion of Tregs. Tregs secrete IL-10 and TGF-β, which lead to decrease of allergy-specific Th2 cytokines, lung eosinophil infiltration, and allergy-specific IgG1 and IgE production.
Figure 1. Schematic presentation of plausible mechanisms by which MSC-derived EVs regulate the allergic airway diseases. MSC-derived EVs carry microRNAs such as miR-146a-5p, miR-1470, and miR-126-3p and deliver them into lung tissues. Intranasal administration of MSC-derived EVs increases the expression of PON1, Bex2, Igfbp6, and Scgb1c1 in lung tissues of asthmatic mice. These microRNAs and pulmonary genes by MSC-derived EVs induce the expansion of Tregs. Tregs secrete IL-10 and TGF-β, which lead to decrease of allergy-specific Th2 cytokines, lung eosinophil infiltration, and allergy-specific IgG1 and IgE production.
Life 12 01994 g001
Table 1. Biological activity of microRNAs associated with suppression of allergic airway inflammation by MSC-derived EVs.
Table 1. Biological activity of microRNAs associated with suppression of allergic airway inflammation by MSC-derived EVs.
microRNAsBiological Activity
miR-146a [88]Decreased lung inflammation;
Polarization of macrophages to M2.
miR-126-3p [85]Increased endothelial cell function;
Decreased VEGF-α and HMGB1 levels;
Increased tight junction protein expression.
miR-145 [89]Increased IL-4 production and bacterial clearance;
Decreased multidrug resistance-associated protein-1 expression.
miR-21-5p [90]Decreased lung edema; M1 polarization.
miR-30b-3p [91]Increased proliferation of alveolar epithelial cells;
Decreased apoptosis of alveolar epithelial cells.
miR-1470 [86]Anti-inflammatory properties by increasing FOXP3+ T cell.
miR-100 [92]Decreased lung inflammation and apoptosis;
Downregulation of mammalian target of rapamycin signaling.
miR-146a-5p [87]Inhibition of group 2 innate lymphoid cells.
miR-27a-3p [93]Decreased lung inflammation and alveolar septum thickness;
Polarization of macrophages to M2 anti-inflammatory phenotype.
miR-191 [94]Inhibition of bone marrow morphogenetic protein receptor 2.
VEGF, vascular endothelial growth factor; HMGB, high mobility group box; IL, interleukin.
Table 2. Pulmonary genes associated with suppression of allergic airway inflammation by MSC-derived EVs.
Table 2. Pulmonary genes associated with suppression of allergic airway inflammation by MSC-derived EVs.
GenesDescriptionGeneral CharacteristicsPotential Effects in Allergic Airway Disease
PON1Family of PON related to calcium-dependent aryldialkylphosphataseAntioxidant, anti-adhesive, anti-inflammatory, anti-thrombotic, and anti-apoptotic effects [103,104,105,106,107,108]Reduced airway inflammation and airway remodeling and inhibited LPS-induced inflammatory cytokine expression and lung fibroblast proliferation in asthmatic mice [109,110,111]
Bex2Family of brain expressed X-linked gene and protein-coding gene, highly expressed in brain, pancreas, and testisCarcinogenesis, regulator of mitochondrial apoptosis and the G1 cell cycle in breast cancer [112]Associated with inhibition of IL-13 induced in allergic airway inflammation [113]
Igfbp6Family of IGFBP related to growth inhibitory protein that regulate the availability of insulin-like growth factorsBiomarker and therapeutic target acting on the pathogenesis of various autoimmune diseases [116]Associated with fibroblast proliferation and cell growth in asthma [115]
Fpr1Family of FPR, group of G protein-coupled cell surface receptors of mammalian phagocytic cellsImportant roles in host defense as well as inflammatory responses including cell adhesion, directed migration, granule release, and superoxide production [117]Associated with stimulation of neutrophil chemotaxis and inflammatory cytokine production by phagocytes such as dendritic cells and macrophages [118]
Scgb1c1Family of secretoglobin secreted proteins found in high concentrations in body fluids of the lungs, lacrimal glands, salivary glands, prostate, uterus, and other tissuesLocalized to Bowman’s glands in the olfactory mucosa [120]Upregulated by IL-4, IL-13 and downregulated by IFN-γ, and it plays an important role in recognizing and clearance of pathogenic microorganisms in the lung epithelial mucosa [119,120,121]
MSC, Mesenchymal stem cells; EVs, extracellular vesicles; PON1, paraoxonase1; LPS, lipopolysaccharide; Bex2, brain expressed X-linked 2; Igfbp6, insulin-like growth factor binding protein 6; Fpr1, formyl peptide receptor 1; Scgb1c1, secretoglobin family 1C member 1; IL, interleukin; IFN-γ, interferon gamma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, S.-D.; Cho, K.-S. Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease. Life 2022, 12, 1994. https://doi.org/10.3390/life12121994

AMA Style

Kim S-D, Cho K-S. Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease. Life. 2022; 12(12):1994. https://doi.org/10.3390/life12121994

Chicago/Turabian Style

Kim, Sung-Dong, and Kyu-Sup Cho. 2022. "Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease" Life 12, no. 12: 1994. https://doi.org/10.3390/life12121994

APA Style

Kim, S.-D., & Cho, K.-S. (2022). Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease. Life, 12(12), 1994. https://doi.org/10.3390/life12121994

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop