Open Access

The role of mesenchymal stem cell‑derived exosomes in asthma (Review)

  • Authors:
    • Kaiying Lv
    • Jiawei Gao
    • Liuxin Yang
    • Xingxing Yuan
  • View Affiliations

  • Published online on: April 11, 2025     https://doi.org/10.3892/mmr.2025.13531
  • Article Number: 166
  • Copyright: © Lv et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Asthma is a chronic respiratory disorder characterized by persistent inflammation, airway hyper‑responsiveness and remodeling, leading to notable morbidity and decreased quality of life for patients. Mesenchymal stem cells (MSCs) have potential in regenerative medicine due to their potent immunomodulatory properties and anti‑inflammatory effects. The therapeutic benefits of MSCs are largely mediated by secreted exosomes that facilitate intercellular communication by transferring bioactive molecules, including proteins, lipids and microRNAs. The present review explores the therapeutic potential of MSC‑derived exosomes in asthma, highlighting their ability to modulate key pathological mechanisms underlying the disease. 

Introduction

Asthma is a chronic respiratory disease characterized by recurrent episodes of airway inflammation, bronchoconstriction and hyperresponsiveness. It affects >300 million individuals globally and contributes to increasing morbidity and healthcare costs (1,2). The etiopathogenesis of asthma involves crosstalk between genetic predispositions and environmental factors, including allergens, infections and pollutants (3). Pathologically, asthma is marked by chronic inflammation of the airway, leading to structural changes and functional impairment (4). Patients with asthma present with intermittent and variable symptoms such as wheezing, shortness of breath, chest tightness and coughing, which result primarily from reversible airway obstruction and vary in intensity and frequency (5,6). Severity of symptoms ranges from mild to life-threatening exacerbations that may require emergency medical intervention (7). Factors such as respiratory infection, allergens and irritants can trigger these exacerbations, highlighting the need for effective long-term management strategies (8).

Current therapeutic strategies for asthma aim to relieve symptoms, improve lung function and decrease the frequency and severity of exacerbations (9). Primary therapeutic approaches include inhaled corticosteroids, long-acting β-agonists, leukotriene modifiers and monoclonal antibodies that target specific immune pathways (10,11). However, these treatments do not fully address the underlying mechanisms of airway remodeling and chronic inflammation, leading to persistent disease progression in patients with poor asthma control and frequent exacerbations (12,13). Therefore, novel therapeutic strategies are needed.

Mesenchymal stem cells (MSCs) are multipotent stromal cells that differentiate into various cell types, including osteocytes, chondrocytes and adipocytes (14). Extracellular vesicles (EVs) are a broad class of vesicles that are secreted by cells into the extracellular space. They can be classified into three subtypes, exosomes, microvesicles and apoptotic bodies, based on their size and mode of release. Exosomes are the smallest subtype of EV, typically 30–150 nm in diameter, and serve key roles in intercellular communication and disease mechanisms (15). MSC-derived exosomes mediate therapeutic effects of MSCs, and have a key role in cell-to-cell communication by transferring bioactive molecules to recipient cells, affecting their behavior and function (16,17). In asthma, MSC-derived exosomes exert immunomodulatory effects that maintain immune homeostasis and suppress excessive immune responses (18,19). Additionally, they exhibit anti-inflammatory effects by inhibiting inflammation-associated signaling pathways and reducing the levels of pro-inflammatory cytokines, which are beneficial in treating asthma (20,21). Compared with drugs targeting IL-4 and IL-5, such as dupilumab and mepolizumab, MSC-derived exosomes offer a potential advantage by providing a more natural and less invasive approach to modulating immune responses (22). They exert long-term effects because of their ability to simultaneously influence multiple cellular pathways (8). While drugs are designed to target specific cytokines, MSC-derived exosomes serve as a multi-target therapy, potentially exerting more comprehensive effects on asthma (23).

The present review discusses the pathogenesis of asthma and the basics of MSC-derived exosomes and emphasizes their potential therapeutic effect, as well as the prospects and challenges of translating MSC-derived exosome therapy from bench to bedside.

Pathological mechanisms of asthma

Asthma is characterized by chronic airway inflammation involving immune cells such as eosinophils, T lymphocytes, mast cells and dendritic cells. These cells release cytokines and chemokines, which perpetuate the inflammatory response and facilitate airway hyperresponsiveness and tissue damage (24,25). These immune cells serve a key role in airway inflammation during disease progression. For example, eosinophils secrete pro-inflammatory cytokines, chemokines, growth factors and lipid mediators that disrupt pulmonary homeostasis and induce inflammatory airway damage (26). They regulate immune responses by modulating T helper 2 (Th2) cell activation, M2 macrophage polarization and group 2 innate lymphoid cell (ILC2) differentiation, as well as upregulating the levels of IL-5 and IL-13, which primarily induce inflammatory tissue damage (27,28). Mast cells, in combination with IgE, are key for initiating inflammation in allergic asthma (29). They release histamine and other inflammatory mediators that contribute to bronchoconstriction and further inflammation (30). Neutrophils, particularly in severe asthma, release nuclear proteins and serine proteases that form neutrophil extracellular traps, which activate inflammasomes in monocytes or macrophages, leading to increased expression of inflammatory cytokines (31).

Chronic inflammation in asthma leads to structural airway alterations, collectively termed airway remodeling (1). This includes epithelial damage, mucous gland hyperplasia, subepithelial collagen deposition and increased proliferation of airway smooth muscle cells (ASMCs) (3234). These changes cause persistent airflow limitation and airway hyperresponsiveness (35). Airway remodeling is driven by numerous factors, including transforming growth factor (TGF)-β, primarily secreted by bronchial epithelial cells and eosinophils (36). TGF-β promotes extracellular matrix deposition by stimulating fibroblasts and myofibroblasts to produce collagen and fibronectin, contributing to airway hyperresponsiveness (37). However, TGF-β exerts both pro-inflammatory and immunosuppressive effects in asthma during airway inflammation depending on cellular context. Elevated TGF-β1 expression in asthmatic airways aggravate eosinophilic inflammation by activating CD8+ T cells while inhibiting Th1 and Th2 cell differentiation (38). It also modulates regulatory T cell (Treg) differentiation and limits the pathogenicity of CD4+ T cells, thereby alleviating airway inflammation (39).

Numerous signaling pathways contribute to asthma pathogenesis. The NF-κB pathway regulates airway inflammation (40). Various stimuli, including pro-inflammatory factors, viruses, drugs and cigarette smoke, activate NF-κB (41). Once activated, NF-κB translocates to the nucleus and upregulates inflammatory gene expression (42). Another key signaling cascade in asthma is the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88) pathway (43). Lipopolysaccharide (LPS) binding to TLR4 activates MyD88, leading to NF-κB nuclear translocation and amplification of the inflammatory response (44). Moreover, the phosphatidylinositol 3-kinase (PI3K)/Akt and Wnt/β-catenin signaling pathways are implicated in airway remodeling and hyperresponsiveness (45,46). These pathways contribute to ASMC proliferation and migration and epithelial-mesenchymal transition (EMT), enhancing airway remodeling (47,48).

Basics of MSC-derived exosomes

MSCs were first identified by Friedenstein et al (49) in the 1960s when they isolated fibroblast colony-forming units from bone marrow. MSCs are present in virtually all postnatal organs and tissues, including the bone marrow, adipose tissue, umbilical cord and placenta (50,51). MSCs express specific surface markers such as CD73, CD90 and CD105 but lack hematopoietic markers such as CD34 and CD45 (52). MSCs can differentiate into osteoblasts, adipocytes and chondroblasts, making them promising candidates for therapeutic application due to their plasticity (53,54).

Exosomes are small EVs ranging from 30 to 150 nm in diameter, released by various cell types into the extracellular environment (55). They originate within the endosomal compartment of the cell and are secreted when multivesicular bodies fuse with the plasma membrane (56) (Fig. 1). Exosomes carry proteins, lipids and nucleic acids, reflecting the content and physiological state of the originating cells (57). This cargo includes mRNA, microRNAs (miRNAs or miRs) and other non-coding RNAs (5860). The biological properties of exosomes make them key mediators of intercellular communication (61). They influence target cells by transferring their cargo, thereby modulating cellular processes, such as proliferation, differentiation and immune responses (62).

Numerous methods have been developed for exosome isolation and characterization, which are key for studying their biological functions and clinical application. Ultracentrifugation is the most commonly used technique, and is considered the gold standard for exosome isolation. It involves sequential centrifugation steps to remove cells, apoptotic bodies and other EVs, followed by high-speed ultracentrifugation to pellet exosomes (63). By using sucrose or iodixanol gradients to separate exosomes based on density, density gradient centrifugation refines ultracentrifugation and improves purity by separating exosomes from contaminants (64). Size-exclusion chromatography separates vesicles based on size using porous columns, allowing exosomes to be eluted while smaller proteins and particles are retained, which maintains exosome integrity and biological activity (65). Additionally, polymer-based precipitation uses polymers such as polyethylene glycol to precipitate exosomes from biofluids, followed by low-speed centrifugation to collect the pellet, which is suitable for processing large sample volumes (66). For small sample volumes, microfluidic platforms use nanoscale filtration, immunoaffinity capture or acoustic forces to isolate exosomes with high precision (67). Serving as a highly specific isolation method for exosome subpopulations, immunoaffinity-based isolation uses antibodies targeting exosome surface markers conjugated to magnetic beads or chromatography matrices (68). Standard characterization techniques of exosomes include morphological analysis, size determination, marker profiling and functional validation. Nanoparticle tracking analysis measures the size distribution and concentration of exosomes in a liquid suspension based on Brownian motion (69). Transmission electron microscopy provides high-resolution images of exosomes, allowing visualization of their characteristic cup-shaped morphology (70). By analyzing scattered light fluctuations, dynamic light scattering determines the hydrodynamic diameter of exosomes (71). In addition, western blotting detects exosome-specific protein markers, whereas flow cytometry allows high-throughput analysis using fluorescently labeled antibodies (72). Thus, the combination of multiple techniques can ensure accurate identification and functional validation. Future research should focus on developing standardized protocols and scalable isolation techniques to enhance reproducibility and facilitate clinical translation.

MSC-derived exosomes are key for mediating the therapeutic effects of MSCs. They promote tissue repair and regeneration by transferring growth factors and miRNAs that enhance cell proliferation and differentiation (73). In addition, they modulate immune responses by altering the phenotype and function of immune cells such as T and B cells and macrophages (74). MSC-derived exosomes suppress pro-inflammatory cytokine production and induce anti-inflammatory cytokine release (75). By inhibiting inflammation-associated signaling pathways such as NF-κB, they decrease pro-inflammatory cytokines levels and exert potent anti-inflammatory effects (76). These properties make MSC-derived exosomes promising therapeutic tools for inflammatory and degenerative disease. Their ability to mimic MSC functions while avoiding the risks associated with direct cell transplantation makes them attractive candidates for clinical application.

Effects of MSC-derived exosomes on asthma

Immunomodulation

MSC-derived exosomes exhibit immunomodulatory effects, which are key in the context of asthma, a disease characterized by immune dysregulation (77). These exosomes modulate the activity of immune cells, such as T cells and macrophages, suppressing asthma exacerbation. For example, in mice with severe refractory asthma and neutrophil-mediated allergic airway inflammation, systemic administration of exosomes from human bone marrow-derived MSCs ameliorates airway hyperreactivity and pulmonary inflammation by reducing Th2 and Th17 phenotypes (78). Suppressing Th2 and Th17 cell proliferation is important to prevent immune system overactivation during disease progression (79,80). Mechanistically, exosomal miR-146a-5p inhibits Th2 cell differentiation by downregulating expression of plasminogen activator inhibitor-2 (81). Th17 cells serve a key role in the pathogenesis of steroid-resistant asthma, characterized by neutrophilic airway inflammation (82). As an inactivator of Th17 cells, exosomes derived from human MSCs decrease IL-17A levels in bronchoalveolar lavage fluid (BALF) and Th17 cell numbers in lung tissue by suppressing the Janus kinase 2/signal transduction and activator of transcription 3 (STAT3) signaling pathway. This further decreases the infiltration of inflammatory cells, particularly neutrophils (83). Similarly, the increased Treg population can produce anti-inflammatory cytokines such as IL-10 and TGF-β, leading to the maintenance of immune homeostasis in asthma (39,84). Du et al (85) demonstrated that MSC-derived exosomes elevate the proliferation and immunosuppressive capacity of Tregs in patients with asthma by upregulating the expression of IL-10 and TGF-β1 in peripheral blood mononuclear cells. miR-1470 within MSC-derived exosomes facilitates the differentiation of CD4+CD25+FOXP3+ Tregs in patients with asthma by upregulating p27kip1, a cyclin-dependent kinase regulator that controls cell differentiation (86,87). Therefore, exosomes from MSCs modulate T cell activation and proliferation, decrease overactivated immune responses and further mitigate asthma severity. MSC-derived exosomes also mediate the polarization of macrophages towards the M2 phenotype, which is associated with anti-inflammatory and tissue repair function (88). Exosomes from MSCs from human umbilical cord inhibit M1 macrophage polarization and induce M2 polarization by targeting tumor necrosis factor receptor-associated factor 1 (89). They suppresses NF-κB and PI3K/Akt signaling, and reduce airway hyperresponsiveness and damage, highlighting the therapeutic function of MSC-derived exosomes in moderating immune responses through macrophage polarization (89).

In summary, MSC-derived exosomes exert immunomodulatory effects on asthma by modulating immune cell populations, including Th17 cells, Tregs and macrophages, through distinct signaling pathways and miRNA-mediated mechanisms. Their therapeutic potential in asthma management offers a novel approach for treating severe and refractory forms of the disease. Future studies should focus on elucidating the precise mechanisms of action to validate these findings in preclinical settings

Airway inflammation

Anti-inflammatory properties of MSC-derived exosomes are attributed to their ability to decrease the infiltration of inflammatory cells and generation of pro-inflammatory cytokines in the airway (85,90). For example, MSC-derived EVs decrease inflammatory cell counts within the lung tissue and alleviate cellularity and eosinophilia in BALF, which is associated with lower levels of eotaxin-2 in a murine model of allergic airway inflammation (91). Ovalbumin-loaded MSC-derived exosome formulation decreases levels of inflammatory cells and eosinophilia in the lung tissues. This effect is accompanied by reduced IgE and IL-4 levels but elevated interferon (IFN)-γ and TGF-β production (90). Moreover, exosomes carry anti-inflammatory molecules, such as miRNAs, that modulate the infiltration of inflammatory cells and expression of cytokines (88,92). MSC-derived exosomal miR-146a-5p hinders the function of ILC2s and decreases inflammatory cell infiltration, pulmonary mucus production and Th2 cytokine secretion, thus alleviating airway hyperresponsiveness in a mouse model of ILC2-dominant asthma (83). By inhibiting the Jumonji AT Rich interacting domain 2/Wnt/β-catenin axis, miR-188 from MSC-derived exosomes decreases inflammation infiltration, mucus production and collagen deposition in lung tissue of asthmatic mice (93). Additionally, in LPS-stimulated alveolar macrophages, MSC-derived conditioned medium can modulate the release of TNF-α, IL-6, and TGF-β1. This indicates the therapeutic potential of MSC-derived exosomes in lung inflammatory disorder (94). Similarly, exosomes from MSCs have been shown to downregulate the transcription and protein expression of pro-inflammatory cytokines such as IL-1β, IL-8 and IL-6 by upregulating the expression of peroxisome proliferator-activated receptor γ, which serves as a transcription factor controlling anti-inflammatory and antioxidant mechanisms via NF-κB and heme oxygenase-1, thereby attenuating inflammation and oxidative stress in asthma (95). By preventing the activation of STAT3, exosomal miR-301a-3p from adipose-derived MSCs decreases the generation of inflammatory factors, including TNF-α, IL-1β and IL-6, in ASMCs treated with platelet-derived growth factor. This is accompanied by suppressed proliferation and migration of ASMCs, thus alleviating airway inflammation and remodeling in an ovalbumin-induced asthma mouse model (96). The aforementioned studies highlight the potential of MSC-derived exosomes in modulating airway inflammation in asthma.

Altogether, MSC-derived exosomes impede inflammatory cell infiltration and pro-inflammatory cytokine production by modulating key inflammatory signaling pathways, providing new avenues for developing effective and targeted therapies for asthma. Further research is needed to identify the precise molecular mechanisms by which MSC-derived exosomes exert anti-inflammatory effects in asthma.

Airway remodeling

As a hallmark of chronic asthma, airway remodeling is characterized by structural changes, such as subepithelial fibrosis, increased smooth muscle mass and mucus gland hyperplasia, which can be mitigated by MSC-derived exosomes (97,98). For example, exosomes from MSCs inhibit the deposition of collagen fibers in the airway and lung parenchyma and reduce eosinophil counts in lung tissue and BALF, thereby decreasing subepithelial fibrosis. This effect is mediated by the downregulation of fibrogenic factors, such as TGF-β, and the inhibition of myofibroblast differentiation (99). Furthermore, MSC-derived exosomes decrease the number of immune cells in BALF, proliferation of goblet cells and deposition of collagens by inactivating the Wnt/β-catenin signaling pathway, which ameliorates EMT and airway remodeling in asthmatic rats (100). Furthermore, MSC-derived exosomal miRNAs decrease allergic airway inflammation and further alleviate airway remodeling by modulating signaling pathways involved in ASMC proliferation and migration (101,102). This helps prevent thickening of the airway wall and maintain airway patency (97). Exosomal miR-221-3p derived from human bone marrow MSCs can restrict ASMC proliferation, migration and extracellular matrix deposition by targeting fibroblast growth factor 2, thus inhibiting the ERK1/2 signaling pathway, which lessens airway hyperresponsiveness and histopathological damage in an ovalbumin-induced asthmatic mouse model (103). Similarly, exosomal miR-146a-5p derived from human umbilical cord MSCs cultured under hypoxic conditions is capable of decreasing the expression of profibrogenic markers such as α-smooth muscle actin and collagen-1. It is associated with the suppression of TGF-β1/Smad2/3 signaling pathway, thus ameliorating airway remodeling in mice with chronic asthma (104). Li and Yang (105) revealed that miR-223-3p, which is highly expressed in bone marrow MSC-derived exosomes, alleviates airway inflammation and remodeling by targeting the nucleotide-binding domain, leucine-rich–containing family, pyrin domain-containing-3-induced apoptosis-associated speck-like protein/caspase-1/gasdermin D signaling pathway (105), which accelerates the restoration of the epithelial barrier function and decreases mucus hypersecretion. The aforementioned findings suggest therapeutic potential of MSC-derived exosomes in addressing airway remodeling in asthma.

MSC-derived exosomes ameliorate airway remodeling through various mechanisms, including the inhibition of key signaling pathways associated with ASMC proliferation and migration, modulation of immune cell function and suppression of pro-inflammatory cytokines. Future research should focus on the precise molecular mechanisms underlying the severity of asthma. MSC-derived exosomes may represent a promising frontier in the development of innovative therapy for asthma.

In conclusion, MSC-derived exosomes alleviate asthma progression by modulating immune responses and mitigating airway inflammation and remodeling (Fig. 2). They modulate multiple molecular pathways, including NF-κB, TGF-β/Smad and JAK/STAT, which are central to inflammation and tissue remodeling in asthma. Therefore, MSC-derived exosomes have a potential as treatments for asthma. Future investigations should focus on identifying the mechanisms by which these exosomes suppress pathological processes and alleviate disease severity. Exosomes from MSCs transfer proteins such as ovalbumin that are decorated with a dendritic cell-specific aptamer, which have been shown to improve the efficacy of specific immunotherapy in asthma model (106). Furthermore, MSC-derived exosomal miRNAs inhibit disease progression (81,83,93,96,103105) (Table I). Targeting exosomal miRNAs presents a novel therapeutic approach for asthma treatment. Strategies to control exosomal miRNA activity include the use of miRNA mimics to restore the function of downregulated miRNAs and miRNA inhibitors to block the activity of upregulated miRNAs (107,108).

Table I.

Exosomal miRs derived from MSCs that inhibit asthma progression.

Table I.

Exosomal miRs derived from MSCs that inhibit asthma progression.

First author, yearExosomal miRsDownstream targetOutcomeExperimental model(Refs.)
Zhou et al, 2021miR-146a-5pSERPINB2Inhibits the differentiation of Th2 cells and allergic inflammationPatients with allergic rhinitis(81)
Fang et al, 2020miR-146a-5pUnknownDecreases ILC2 levels, inflammatory cell infiltration and mucus production in the lung, as well as Th2 cytokines and airway hyperresponsivenessIL-33-stimulated ILC2, IL-33-treated mice(83)
Zhuansun et al, 2019miR-1470 c-Jun/p27kip1Upregulates the proportion of CD4+CD25+FOXP3+ Tregs and inhibits asthma progressionIL-2-stimulated CD4+ T cells(86)
Shan et al, 2022miR-188 JARID2/Wnt/β-cateninAttenuates the proliferation and migration of ASMCs, as well as decreasing inflammatory cell infiltration, mucus production and collagen deposition in lung tissueTGF-β1-stimulated ASMCs, OVA-sensitized mice(93)
Feng et al, 2022miR-301a-3pSTAT3Suppresses the proliferation and migration of ASMCs, as well as decreasing the secretion of inflam-PDGF-treated ASMCs, OVA-sensitized mice(96)
matory factors and lung injuryTGF-β1-treated ASMCs, OVA-(103)
Liu et al, 2022miR-221-3pFGF2/ERK1/2Attenuates the proliferation, migration and ECM deposition in ASMCs and alleviates airway hyperresponsiveness and asthma progressionsensitized mice
Dong et al, 2021miR-146a-5pTGF-β1/Smad2/3Decreases levels of IL-4, IL-13 and eosinophils, as well expression of α-SMA and collagen-1TGF-β1-stimulated fibroblasts, OVA-sensitized mice(104)
Li and Yang, 2023miR-223-3p NLRP3/ASC/caspase-1/GSDMDMitigates airway inflammation and remodelingOVA-sensitized mice(105)

[i] ASMC, airway smooth muscle cell; α-SMA, α-smooth muscle actin; FGF2, fibroblast growth factor 2; GSDMD, gasdermin D; ILC2, group 2 innate lymphoid cell; JARID2, Jumonji, AT-rich interaction domain containing 2; MSC, mesenchymal stem cell; miR, microRNA; NLRP3, NOD-like receptor family pyrin domain-containing 3; SERPINB2, serpin family B member 2; TGF, transforming growth factor; Th, T helper; Treg, regulatory T cell; OVA, ovalbumin; PDGF, platelet-derived growth factor; ASC, apoptosis-associated speck-like protein.

Future directions

MSC-derived exosomes offer a novel therapeutic approach for asthma with the potential to modulate immune responses, decrease inflammation and repair airway damage (83,86). Their ability to deliver bioactive molecules such as proteins, lipids and nucleic acids to target cells makes them a versatile tool for therapeutic intervention. The immunomodulatory and anti-inflammatory properties of MSC-derived exosomes have been demonstrated in various preclinical models (103105), demonstrating promise for decreasing asthma symptoms and improving lung function.

Despite their therapeutic potential, several challenges must be addressed before MSC-derived exosomes can be translated into clinical application. First, standardizing exosome isolation and characterization methods is essential to ensure reproducibility and consistency. Different techniques used to isolate exosomes, such as ultracentrifugation, size exclusion chromatography and immunoaffinity capture, yield exosomes with varying purity and characteristics, potentially affecting their therapeutic efficacy. Secondly, understanding the pharmacokinetics and biodistribution of MSC-derived exosomes is key for optimizing their therapeutic use. The mechanisms underlying their uptake, distribution and clearance in vivo need to be investigated to improve targeted delivery to the lungs and minimize off-target effects. Additionally, the potential immunogenicity and long-term safety of MSC-derived exosomes require evaluation in both preclinical and clinical studies.

Further research is essential to elucidate the therapeutic potential of MSC-derived exosomes. Future studies should focus on defining their mechanisms of action, determining optimal dosing regimens and exploring combination therapies to enhance their efficacy. Advances in exosome engineering may enable the customization of exosome content and surface modification to improve their targeting and therapeutic effects. Moreover, the development of advanced delivery systems to enhance targeted lung delivery is key for maximizing the therapeutic potential of MSC-derived exosomes.

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 82205007).

Availability of data and materials

Not applicable.

Authors' contributions

KL, JG and LY wrote and revised the manuscript. XY edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ASMC

airway smooth muscle cell

BALF

bronchoalveolar lavage fluid

EV

extracellular vesicle

MSC

mesenchymal stem cell

MyD88

myeloid differentiation primary response 88

PI3K

phosphatidylinositol 3-kinase

TGF-β

transforming growth factor-β

TLR4

toll-like receptor 4

Treg

regulatory T cell

EMT

epithelial-mesenchymal transition

ILC2

group 2 innate lymphoid cell

Th2

T helper 2

TNF

tumor necrosis factor

LPS

lipopolysaccharide

References

1 

Varricchi G, Ferri S, Pepys J, Poto R, Spadaro G, Nappi E, Paoletti G, Virchow JC, Heffler E and Canonica WG: Biologics and airway remodeling in severe asthma. Allergy. 77:3538–3552. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Cheng Q, Pan J, Zhou ZL, Yin F, Xie HY, Chen PP, Li JY, Zheng PQ, Zhou L, Zhang W, et al: Caspase-11/4 and gasdermin D-mediated pyroptosis contributes to podocyte injury in mouse diabetic nephropathy. Acta Pharmacol Sin. 42:954–963. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Stern J, Pier J and Litonjua AA: Asthma epidemiology and risk factors. Semin Immunopathol. 42:5–15. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Ntontsi P, Photiades A, Zervas E, Xanthou G and Samitas K: Genetics and epigenetics in asthma. Int J Mol Sci. 22:24122021. View Article : Google Scholar : PubMed/NCBI

5 

Chupp GL, Kaur R and Mainardi A: New therapies for emerging endotypes of asthma. Annu Rev Med. 71:289–302. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Ray A, Das J and Wenzel SE: Determining asthma endotypes and outcomes: Complementing existing clinical practice with modern machine learning. Cell Rep Med. 3:1008572022. View Article : Google Scholar : PubMed/NCBI

7 

Liu T, Prescott WG and Zhou X: Advances in non-type 2 asthma in the severe cases: From molecular insights to novel treatment strategies. Eur Respir J. 64:23008262024. View Article : Google Scholar : PubMed/NCBI

8 

von Mutius E and Smits HH: Primary prevention of asthma: From risk and protective factors to targeted strategies for prevention. Lancet. 396:854–866. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Lommatzsch M, Brusselle GG, Levy ML, Canonica GW, Pavord ID, Schatz M and Virchow JC: A2BCD: A concise guide for asthma management. Lancet Respir Med. 11:573–576. 2023. View Article : Google Scholar : PubMed/NCBI

10 

McDowell PJ, McDowell R, Busby J, Eastwood MC, Patel PH, Jackson DJ, Mansur A, Patel M, Burhan H, Doe S, et al: Clinical remission with biologic therapies in severe asthma: A matter of definition. Eur Respir J. 63:24001602024. View Article : Google Scholar : PubMed/NCBI

11 

Wenzel SE: Severe adult asthmas: Integrating clinical features, biology, and therapeutics to improve outcomes. Am J Respir Crit Care Med. 203:809–821. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Thomas D, McDonald VM, Pavord ID and Gibson PG: Asthma remission: What is it and how can it be achieved? Eur Respir J. 60:21025832022. View Article : Google Scholar : PubMed/NCBI

13 

Coleman C, Khaleva E, Rattu A, Frankemölle B, Nielsen H, Roberts G and Williams C: Narrative review to capture patients' perceptions and opinions about Non-response and response to biological therapy for severe asthma. Eur Respir J. 61:22008372023. View Article : Google Scholar : PubMed/NCBI

14 

Zhou T, Yuan Z, Weng J, Pei D, Du X, He C and Lai P: Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 14:242021. View Article : Google Scholar : PubMed/NCBI

15 

Mathieu M, Martin-Jaular L, Lavieu G and Thery C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 21:9–17. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Fang J, Liu B, Shao C and Shi Y: Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell. 29:1515–1530. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Li P, Gong Z, Shultz LD and Ren G: Mesenchymal stem cells: From regeneration to cancer. Pharmacol Ther. 200:42–54. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Varderidou-Minasian S and Lorenowicz MJ: Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics. 10:5979–5997. 2020. View Article : Google Scholar : PubMed/NCBI

19 

van Griensven M and Balmayor ER: Extracellular vesicles are key players in mesenchymal stem cells' dual potential to regenerate and modulate the immune system. Adv Drug Deliv Rev. 207:1152032024. View Article : Google Scholar : PubMed/NCBI

20 

Zhu X, Ma D, Yang B, An Q, Zhao J, Gao X and Zhang L: Research progress of engineered mesenchymal stem cells and their derived exosomes and their application in autoimmune/inflammatory diseases. Stem Cell Res Ther. 14:712023. View Article : Google Scholar : PubMed/NCBI

21 

Wang S, Lei B, Zhang E, Gong P, Gu J, He L, Han L and Yuan Z: Targeted therapy for inflammatory diseases with mesenchymal stem cells and their derived exosomes: From basic to clinics. Int J Nanomedicine. 17:1757–1781. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Brusselle GG and Koppelman GH: Biologic therapies for severe asthma. N Engl J Med. 386:157–171. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Ma Y, Liu X, Long Y and Chen Y: Emerging therapeutic potential of mesenchymal stem Cell-derived extracellular vesicles in chronic respiratory diseases: An overview of recent progress. Front Bioeng Biotechnol. 10:8450422022. View Article : Google Scholar : PubMed/NCBI

24 

Bao Y and Zhu X: Role of chemokines and inflammatory cells in respiratory allergy. J Asthma Allergy. 15:1805–1822. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Kim YM, Kim H, Lee S, Kim S, Lee JU, Choi Y, Park HW, You G, Kang H, Lee S, et al: Airway G-CSF identifies neutrophilic inflammation and contributes to asthma progression. Eur Respir J. 55:19008272020. View Article : Google Scholar : PubMed/NCBI

26 

Al-Shaikhly T, Murphy RC, Parker A, Lai Y, Altman MC, Larmore M, Altemeier WA, Frevert CW, Debley JS, Piliponsky AM, et al: Location of eosinophils in the airway wall is critical for specific features of airway hyperresponsiveness and T2 inflammation in asthma. Eur Respir J. 60:21018652022. View Article : Google Scholar : PubMed/NCBI

27 

Nagakumar P, Puttur F, Gregory LG, Denney L, Fleming L, Bush A, Lloyd CM and Saglani S: Pulmonary type-2 innate lymphoid cells in paediatric severe asthma: Phenotype and response to steroids. Eur Respir J. 54:18018092019. View Article : Google Scholar : PubMed/NCBI

28 

Wang W, Xu Y, Wang L, Zhu Z, Aodeng S, Chen H, Cai M, Huang Z, Han J, Wang L, et al: Single-cell profiling identifies mechanisms of inflammatory heterogeneity in chronic rhinosinusitis. Nat Immunol. 23:1484–1494. 2022. View Article : Google Scholar : PubMed/NCBI

29 

Tiotiu A, Badi Y, Kermani NZ, Sanak M, Kolmert J, Wheelock CE, Hansbro PM, Dahlén SE, Sterk PJ, Djukanovic R, et al: Association of differential mast cell activation with granulocytic inflammation in severe asthma. Am J Respir Crit Care Med. 205:397–411. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Dwyer DF, Ordovas-Montanes J, Allon SJ, Buchheit KM, Vukovic M, Derakhshan T, Feng C, Lai J, Hughes TK, Nyquist SK, et al: Human airway mast cells proliferate and acquire distinct inflammation-driven phenotypes during type 2 inflammation. Sci Immunol. 6:eabb72212021. View Article : Google Scholar : PubMed/NCBI

31 

Chacón P, Vega-Rioja A, Doukkali B, Del Valle Rodriguez A, Fernández-Delgado L, Domínguez-Cereijo L, Segura C, Pérez-Machuca BM, Perkins JR, El Bekay R, et al: Human neutrophils couple nitric oxide production and extracellular traps formation in allergic asthma. Am J Respir Crit Care Med. 210:593–606. 2024. View Article : Google Scholar : PubMed/NCBI

32 

Jesenak M, Durdik P, Oppova D, Franova S, Diamant Z, Golebski K, Banovcin P, Vojtkova J and Novakova E: Dysfunctional mucociliary clearance in asthma and airway remodeling-new insights into an old topic. Respir Med. 218:1073722023. View Article : Google Scholar : PubMed/NCBI

33 

Li Z, Cheng T, Guo Y, Gao R, Ma X, Mao X and Han X: Cd147 induces asthmatic airway remodeling and activation of circulating fibrocytes in a mouse model of asthma. Respir Res. 25:62024. View Article : Google Scholar : PubMed/NCBI

34 

Defnet AE, Shah SD, Huang W, Shapiro P, Deshpande DA and Kane MA: Dysregulated retinoic acid signaling in airway smooth muscle cells in asthma. FASEB J. 35:e220162021. View Article : Google Scholar : PubMed/NCBI

35 

Shimizu K, Tanabe N, Oguma A, Kimura H, Suzuki M, Yokota I, Makita H, Sato S, Hirai T, Nishimura M and Konno S: Parenchymal destruction in asthma: Fixed airflow obstruction and lung function trajectory. J Allergy Clin Immunol. 149:934–942.e8. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Halwani R, Al-Muhsen S, Al-Jahdali H and Hamid Q: Role of transforming growth factor-β in airway remodeling in asthma. Am J Respir Cell Mol Biol. 44:127–133. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Kraik K, Tota M, Laska J, Lacwik J, Pazdzierz L, Sedek L and Gomulka K: The Role of Transforming Growth Factor-β (TGF-β) in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Cells. 13:12712024. View Article : Google Scholar : PubMed/NCBI

38 

Musiol S, Alessandrini F, Jakwerth CA, Chaker AM, Schneider E, Guerth F, Schnautz B, Grosch J, Ghiordanescu I, Ullmann JT, et al: TGF-β1 drives inflammatory th cell but not treg cell compartment upon allergen exposure. Front Immunol. 12:7632432021. View Article : Google Scholar : PubMed/NCBI

39 

Whitehead GS, Thomas SY, Nakano K, Royer DJ, Burke CG, Nakano H and Cook DN: A neutrophil/TGF-β axis limits the pathogenicity of allergen-specific CD4+ T cells. JCI Insight. 7:e1502512022. View Article : Google Scholar : PubMed/NCBI

40 

Lim JO, Kim WI, Pak SW, Lee SJ, Park SH, Shin IS and Kim JC: Toll-like receptor 4 is a key regulator of asthma exacerbation caused by aluminum oxide nanoparticles via regulation of NF-κB phosphorylation. J Hazard Mater. 448:1308842023. View Article : Google Scholar : PubMed/NCBI

41 

Athari SS: Targeting cell signaling in allergic asthma. Signal Transduct Target Ther. 4:452019. View Article : Google Scholar : PubMed/NCBI

42 

de Jesús TJ, Centore JT and Ramakrishnan P: Differential regulation of basal expression of inflammatory genes by NF-κB family subunits. Cell Mol Immunol. 16:720–723. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Shang L, Wang L, Shi X, Wang N, Zhao L, Wang J and Liu C: HMGB1 was negatively regulated by HSF1 and mediated the TLR4/MyD88/NF-κB signal pathway in asthma. Life Sci. 241:1171202020. View Article : Google Scholar : PubMed/NCBI

44 

Kuzmich NN, Sivak KV, Chubarev VN, Porozov YB, Savateeva-Lyubimova TN and Peri F: TLR4 signaling pathway modulators as potential therapeutics in inflammation and sepsis. Vaccines (Basel). 5:342017. View Article : Google Scholar : PubMed/NCBI

45 

Liu JH, Li C, Zhang CH and Zhang ZH: LncRNA-CASC7 enhances corticosteroid sensitivity via inhibiting the PI3K/AKT signaling pathway by targeting miR-21 in severe asthma. Pulmonology. 26:18–26. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Kwak HJ, Park DW, Seo JY, Moon JY, Kim TH, Sohn JW, Shin DH, Yoon HJ, Park SS and Kim SH: The wnt/β-catenin signaling pathway regulates the development of airway remodeling in patients with asthma. Exp Mol Med. 47:e1982015. View Article : Google Scholar : PubMed/NCBI

47 

Wang H, Zhong B, Geng Y, Hao J, Jin Q, Zhang Y, Dong L, Gao D, Li J and Hou W: TIPE2 inhibits PDGF-BB-induced phenotype switching in airway smooth muscle cells through the PI3K/Akt signaling pathway. Respir Res. 22:2382021. View Article : Google Scholar : PubMed/NCBI

48 

Dai Y, Li Y, Cheng R, Gao J, Li Y and Lou C: TRIM37 inhibits PDGF-BB-induced proliferation and migration of airway smooth muscle cells. Biomed Pharmacother. 101:24–29. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Friedenstein AJ, Piatetzky S II and Petrakova KV: Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 16:381–390. 1966.PubMed/NCBI

50 

Pærregaard SI, Wulff L, Schussek S, Niss K, Mörbe U, Jendholm J, Wendland K, Andrusaite AT, Brulois KF, Nibbs RJB, et al: The small and large intestine contain related mesenchymal subsets that derive from embryonic gli1+ precursors. Nat Commun. 14:23072023. View Article : Google Scholar

51 

Miclau K, Hambright WS, Huard J, Stoddart MJ and Bahney CS: Cellular expansion of mscs: Shifting the regenerative potential. Aging Cell. 22:e137592023. View Article : Google Scholar : PubMed/NCBI

52 

Zhang P, Dong J, Fan X, Yong J, Yang M, Liu Y, Zhang X, Lv L, Wen L, Qiao J, et al: Characterization of mesenchymal stem cells in human fetal bone marrow by single-cell transcriptomic and functional analysis. Signal Transduct Target Ther. 8:1262023. View Article : Google Scholar : PubMed/NCBI

53 

Yu H, Huang Y and Yang L: Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev. 80:1016842022. View Article : Google Scholar : PubMed/NCBI

54 

Huang CW, Lu SY, Huang TC, Huang BM, Sun HS, Yang SH, Chuang JI, Hsueh YY, Wu YT and Wu CC: FGF9 induces functional differentiation to Schwann cells from human adipose derived stem cells. Theranostics. 10:2817–2831. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Wang J, Yue BL, Huang YZ, Lan XY, Liu WJ and Chen H: Exosomal RNAs: Novel potential biomarkers for diseases-A review. Int J Mol Sci. 23:24612022. View Article : Google Scholar : PubMed/NCBI

56 

Krylova SV and Feng D: The machinery of exosomes: Biogenesis, release, and uptake. Int J Mol Sci. 24:13372023. View Article : Google Scholar : PubMed/NCBI

57 

Gurung S, Perocheau D, Touramanidou L and Baruteau J: The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun Signal. 19:472021. View Article : Google Scholar : PubMed/NCBI

58 

Hessvik NP and Llorente A: Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 75:193–208. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Colombo M, Raposo G and Thery C: Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 30:255–289. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Feng Y, Guo K, Jiang J and Lin S: Mesenchymal stem cell-derived exosomes as delivery vehicles for non-coding RNAs in lung diseases. Biomed Pharmacother. 170:1160082024. View Article : Google Scholar : PubMed/NCBI

61 

Niazi V, Parseh B, Ahani M, Karami F, Gilanchi S, Atarodi K, Soufi M, Soleimani M, Ghafouri-Fard S, Taheri M and Zali H: Communication between stromal and hematopoietic stem cell by exosomes in normal and malignant bone marrow niche. Biomed Pharmacother. 132:1108542020. View Article : Google Scholar : PubMed/NCBI

62 

Yaghoubi Y, Movassaghpour A, Zamani M, Talebi M, Mehdizadeh A and Yousefi M: Human umbilical cord mesenchymal stem cells Derived-exosomes in diseases treatment. Life Sci. 233:1167332019. View Article : Google Scholar : PubMed/NCBI

63 

Dilsiz N: A comprehensive review on recent advances in exosome isolation and characterization: Toward clinical applications. Transl Oncol. 50:1021212024. View Article : Google Scholar : PubMed/NCBI

64 

Li K, Wong DK, Hong KY and Raffai RL: Cushioned-density gradient ultracentrifugation (C-DGUC): A refined and high performance method for the isolation, characterization, and use of exosomes. Methods Mol Biol. 1740:69–83. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Boing AN, van der Pol E, Grootemaat AE, Coumans FA, Sturk A and Nieuwland R: Single-step isolation of extracellular vesicles by Size-exclusion chromatography. J Extracell Vesicles. 32014.doi: 10.3402/jev.v3.23430.

66 

Rahmatinejad F, Kharat Z, Jalili H, Renani MK and Mobasheri H: Comparison of morphology, protein concentration, and size distribution of bone marrow and Wharton's jelly-derived mesenchymal stem cells exosomes isolated by ultracentrifugation and Polymer-based precipitation techniques. Tissue Cell. 88:1024272024. View Article : Google Scholar : PubMed/NCBI

67 

Mousavi SM, Amin Mahdian SM, Ebrahimi MS, Taghizadieh M, Vosough M, Sadri Nahand J, Hosseindoost S, Vousooghi N, Javar HA, Larijani B, et al: Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. Mol Ther Nucleic Acids. 28:758–791. 2022. View Article : Google Scholar : PubMed/NCBI

68 

Mondal SK and Whiteside TL: Immunoaffinity-Based isolation of melanoma Cell-Derived and T Cell-derived exosomes from plasma of melanoma patients. Methods Mol Biol. 2265:305–321. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Sharma V, Nikolajeff F and Kumar S: Employing nanoparticle tracking analysis of salivary neuronal exosomes for early detection of neurodegenerative diseases. Transl Neurodegener. 12:72023. View Article : Google Scholar : PubMed/NCBI

70 

Wen J, Zhang Z, Feng G, Zhang Y, Li H, Lambert C, Mallouk N and Li G: Transmission electron microscopy assessment of a novel method for isolating pure exosomes from serum. Biotech Histochem. 98:391–395. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Lyu TS, Ahn Y, Im YJ, Kim SS, Lee KH, Kim J, Choi Y, Lee D, Kang E, Jin G, et al: The characterization of exosomes from fibrosarcoma cell and the useful usage of Dynamic Light Scattering (DLS) for their evaluation. PLoS One. 16:e02319942021. View Article : Google Scholar : PubMed/NCBI

72 

Miron RJ and Zhang Y: Understanding exosomes: Part 1-Characterization, quantification and isolation techniques. Periodontol. 94:231–256. 2024. View Article : Google Scholar : PubMed/NCBI

73 

Su N, Hao Y, Wang F, Hou W, Chen H and Luo Y: Mesenchymal stromal Exosome-functionalized scaffolds induce innate and adaptive immunomodulatory responses toward tissue repair. Sci Adv. 7:eabf72072021. View Article : Google Scholar : PubMed/NCBI

74 

Tan F, Li X, Wang Z, Li J, Shahzad K and Zheng J: Clinical applications of Stem cell-derived exosomes. Signal Transduct Target Ther. 9:172024. View Article : Google Scholar : PubMed/NCBI

75 

Planat-Benard V, Varin A and Casteilla L: MSCs and inflammatory cells crosstalk in regenerative medicine: Concerted actions for optimized resolution driven by energy metabolism. Front Immunol. 12:6267552021. View Article : Google Scholar : PubMed/NCBI

76 

Li W, Liu Q, Shi J, Xu X and Xu J: The role of TNF-α in the fate regulation and functional reprogramming of mesenchymal stem cells in an inflammatory microenvironment. Front Immunol. 14:10748632023. View Article : Google Scholar : PubMed/NCBI

77 

Li H, Tian Y, Xie L, Liu X, Huang Z and Su W: Mesenchymal stem cells in allergic diseases: Current status. Allergol Int. 69:35–45. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Cruz FF, Borg ZD, Goodwin M, Sokocevic D, Wagner DE, Coffey A, Antunes M, Robinson KL, Mitsialis SA, 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. 4:1302–1316. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Saikumar Jayalatha AK, Ketelaar ME, Hesse L, Badi YE, Zounemat-Kermani N, Brouwer S, Dijk NF, van den Berge M, Guryev V, Sayers I, et al: IL-33 induced gene expression in activated Th2 effector cells is dependent on IL-1RL1 haplotype and asthma status. Eur Respir J. 63:24000052024. View Article : Google Scholar : PubMed/NCBI

80 

Stark JM, Liu J, Tibbitt CA, Christian M, Ma J, Wintersand A, Dunst J, Kreslavsky T, Murrell B, Adner M, et al: Recombinant multimeric dog allergen prevents airway hyperresponsiveness in a model of asthma marked by vigorous TH 2 and TH 17 cell responses. Allergy. 77:2987–3001. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Zhou J, Lu Y, Wu W and Feng Y: HMSC-Derived Exosome Inhibited Th2 Cell Differentiation via Regulating miR-146a-5p/SERPINB2 Pathway. J Immunol Res. 2021:66965252021. View Article : Google Scholar : PubMed/NCBI

82 

Xie Y, Abel PW, Casale TB and Tu Y: TTH17 cells and corticosteroid insensitivity in severe asthma. J Allergy Clin Immunol. 149:467–479. 2022. View Article : Google Scholar : PubMed/NCBI

83 

Fang SB, Zhang HY, Wang C, He BX, Liu XQ, Meng XC, Peng YQ, Xu ZB, Fan XL, Wu ZJ, 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. 9:17232602020. View Article : Google Scholar : PubMed/NCBI

84 

Zhou JY, Alvarez CA and Cobb BA: Integration of IL-2 and IL-4 signals coordinates divergent regulatory T cell responses and drives therapeutic efficacy. Elife. 10:e574172021. View Article : Google Scholar : PubMed/NCBI

85 

Du YM, Zhuansun YX, Chen R, Lin L, Lin Y and Li JG: Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp Cell Res. 363:114–120. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Zhuansun Y, Du Y, Huang F, Lin L, Chen R, Jiang S and 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. 77:1059812019. View Article : Google Scholar : PubMed/NCBI

87 

Bencivenga D, Stampone E, Roberti D, Della Ragione F and Borriello A: p27Kip1, an intrinsically unstructured protein with scaffold properties. Cells. 10:22542021. View Article : Google Scholar : PubMed/NCBI

88 

Li C, Deng C, Zhou T, Hu J, Dai B, Yi F, Tian N, Jiang L, Dong X, Zhu Q, et al: MicroRNA-370 carried by M2 macrophage-derived exosomes alleviates asthma progression through inhibiting the FGF1/MAPK/STAT1 axis. Int J Biol Sci. 17:1795–1807. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Dong B, Wang C, Zhang J, Zhang J, Gu Y, Guo X, Zuo X, Pan H, Hsu AC, Wang G and Wang F: Exosomes from human umbilical cord mesenchymal stem cells attenuate the inflammation of severe Steroid-resistant asthma by reshaping macrophage polarization. Stem Cell Res Ther. 12:2042021. View Article : Google Scholar : PubMed/NCBI

90 

Dehnavi S, Khodadadi A, Asadirad A and Ghadiri AA: Immune response modulation by allergen loaded into mesenchymal stem Cell-derived exosomes as an effective carrier through sublingual immunotherapy. Immunobiology. 228:1523612023. View Article : Google Scholar : PubMed/NCBI

91 

Bandeira E, Jang SC, Lässer C, Johansson K, Rådinger M and Park KS: Effects of mesenchymal stem Cell-derived nanovesicles in experimental allergic airway inflammation. Respir Res. 24:32023. View Article : Google Scholar : PubMed/NCBI

92 

Li X, Yang N, Cheng Q, Zhang H, Liu F and Shang Y: MiR-21-5p in Macrophage-derived exosomes targets Smad7 to promote epithelial mesenchymal transition of airway epithelial cells. J Asthma Allergy. 14:513–524. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Shan L, Liu S, Zhang Q, Zhou Q and Shang Y: Human bone marrow-mesenchymal stem cell-derived exosomal microRNA-188 reduces bronchial smooth muscle cell proliferation in asthma through suppressing the JARID2/Wnt/β-catenin axis. Cell Cycle. 21:352–367. 2022. View Article : Google Scholar : PubMed/NCBI

94 

Zucca E, Corsini E, Galbiati V, Lange-Consiglio A and Ferrucci F: Evaluation of amniotic mesenchymal cell derivatives on cytokine production in equine alveolar macrophages: An in vitro approach to lung inflammation. Stem Cell Res Ther. 7:1372016. View Article : Google Scholar : PubMed/NCBI

95 

Zulueta A, Colombo M, Peli V, Falleni M, Tosi D, Ricciardi M, Baisi A, Bulfamante G, Chiaramonte R and Caretti A: Lung mesenchymal stem Cells-derived extracellular vesicles attenuate the inflammatory profile of cystic fibrosis epithelial cells. Cell Signal. 51:110–118. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Feng CY, Bai SY, Li ML, Zhao JY, Sun JM, Bao HJ, Ren Y and Su XM: Adipose-derived mesenchymal stem cell-derived exosomal miR-301a-3p regulates airway smooth muscle cells during asthma by targeting STAT3. J Asthma Allergy. 15:99–110. 2022. View Article : Google Scholar : PubMed/NCBI

97 

Abbaszadeh H, Ghorbani F, Abbaspour-Aghdam S, Kamrani A, Valizadeh H, Nadiri M, Sadeghi A, Shamsasenjan K, Jadidi-Niaragh F, Roshangar L and Ahmadi M: Chronic obstructive pulmonary disease and asthma: Mesenchymal stem cells and their extracellular vesicles as potential therapeutic tools. Stem Cell Res Ther. 13:2622022. View Article : Google Scholar : PubMed/NCBI

98 

Liu H, Chen Y, Yin G and Xie Q: Therapeutic prospects of MicroRNAs carried by mesenchymal stem Cells-derived extracellular vesicles in autoimmune diseases. Life Sci. 277:1194582021. View Article : Google Scholar : PubMed/NCBI

99 

de Castro LL, Xisto DG, Kitoko JZ, Cruz FF, Olsen PC, Redondo PAG, Ferreira TPT, Weiss DJ, Martins MA, Morales MM and Rocco PRM: 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. 8:1512017. View Article : Google Scholar : PubMed/NCBI

100 

Song J, Zhu XM and Wei QY: MSCs reduce airway remodeling in the lungs of asthmatic rats through the Wnt/β-catenin signaling pathway. Eur Rev Med Pharmacol Sci. 24:11199–11211. 2020.PubMed/NCBI

101 

Peng YQ, Deng XH, Xu ZB, Wu ZC and Fu QL: Mesenchymal stromal cells and their small extracellular vesicles in allergic diseases: From immunomodulation to therapy. Eur J Immunol. 53:e21495102023. View Article : Google Scholar : PubMed/NCBI

102 

Yuan JY, Wang XY, Tong ZY, Dong YC, Zhao JY, Zhang Y and Shang Y: Promising therapeutic functions of bone marrow mesenchymal stem cells Derived-exosome in asthma. Can Respir J. 2022:14857192022.PubMed/NCBI

103 

Liu W, Lin H, Nie W, Wan J, Jiang Q and Zhang A: Exosomal miR-221-3p derived from bone marrow mesenchymal stem cells alleviates asthma progression by targeting FGF2 and inhibiting the ERK1/2 signaling pathway. Evid Based Complement Alternat Med. 2022:59108742022.PubMed/NCBI

104 

Dong L, Wang Y, Zheng T, Pu Y, Ma Y, Qi X, Zhang W, Xue F, Shan Z, Liu J, et al: Hypoxic hUCMSC-derived extracellular vesicles attenuate allergic airway inflammation and airway remodeling in chronic asthma mice. Stem Cell Res Ther. 12:42021. View Article : Google Scholar : PubMed/NCBI

105 

Li X and Yang N: Exosome miR-223-3p in the bone marrow-derived mesenchymal stem cells alleviates the inflammation and airway remodeling through NLRP3-induced ASC/Caspase-1/GSDMD signaling pathway. Int Immunopharmacol. 123:1107462023. View Article : Google Scholar : PubMed/NCBI

106 

Sadeghi M, Dehnavi S, Khodadadi A, Ghadiri AA, Ganji A, Sharifat M and Asadirad A: Immunomodulatory features of MSC-derived exosomes decorated with DC-specific aptamer for improving sublingual immunotherapy in allergic mouse model. Stem Cell Res Ther. 15:4812024. View Article : Google Scholar : PubMed/NCBI

107 

Xu W, Wang Y, Ma Y and Yang J: MiR-223 plays a protecting role in neutrophilic asthmatic mice through the inhibition of NLRP3 inflammasome. Respir Res. 21:1162020. View Article : Google Scholar : PubMed/NCBI

108 

Kim JY, Stevens P, Karpurapu M, Lee H, Englert JA, Yan P, Lee TJ, Pabla N, Pietrzak M, Park GY, et al: Targeting ETosis by miR-155 inhibition mitigates mixed granulocytic asthmatic lung inflammation. Front Immunol. 13:9435542022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2025
Volume 31 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lv K, Gao J, Yang L and Yuan X: The role of mesenchymal stem cell‑derived exosomes in asthma (Review). Mol Med Rep 31: 166, 2025.
APA
Lv, K., Gao, J., Yang, L., & Yuan, X. (2025). The role of mesenchymal stem cell‑derived exosomes in asthma (Review). Molecular Medicine Reports, 31, 166. https://doi.org/10.3892/mmr.2025.13531
MLA
Lv, K., Gao, J., Yang, L., Yuan, X."The role of mesenchymal stem cell‑derived exosomes in asthma (Review)". Molecular Medicine Reports 31.6 (2025): 166.
Chicago
Lv, K., Gao, J., Yang, L., Yuan, X."The role of mesenchymal stem cell‑derived exosomes in asthma (Review)". Molecular Medicine Reports 31, no. 6 (2025): 166. https://doi.org/10.3892/mmr.2025.13531