Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 May 19;22(1):19.
doi: 10.1186/s12950-025-00444-y.

Exosomes derived from M2 macrophages regulate airway inflammation by modulating epithelial cell proliferation and apoptosis

Affiliations

Exosomes derived from M2 macrophages regulate airway inflammation by modulating epithelial cell proliferation and apoptosis

Yinying Ren et al. J Inflamm (Lond). .

Abstract

Background: Asthma is a chronic inflammatory disease characterized by airway remodeling and immune dysregulation. This study aimed to explore the mechanisms by which M2 macrophage-derived exosomes (M2Φ-Exos) regulate airway inflammation in asthma by modulating epithelial cell proliferation and apoptosis.

Methods: M2Φ-Exos were extracted and characterized by morphology, size, and marker protein expression. In vitro, the effects of M2Φ-Exos on House Dust Mites (HDM)-stimulated mouse lung epithelial cells (MLE-12s) were evaluated using western blotting to analyze Proliferating Cell Nuclear Antigen (PCNA), B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), and cleaved caspase-3 expression. In vivo, M2Φ-Exos were administered to HDM-induced asthmatic mice to assess their impact on airway inflammation, epithelial remodeling, and proliferation-apoptosis balance using immunohistochemistry, immunofluorescence, and western blotting. Cytokine levels in lung tissue and bronchoalveolar lavage fluid (BALF) were measured by qRT-PCR and ELISA.

Results: M2Φ-Exos displayed typical cup-shaped morphology, an average diameter of 115.5 nm, and expressed marker proteins CD9, TSG101, and CD63. MLE-12 cells internalized M2Φ-Exos, leading to reduced abnormal proliferation and apoptosis in HDM-stimulated cells. In asthmatic mice, M2Φ-Exos alleviated airway inflammation and epithelial thickening while reducing PCNA, cleaved caspase-3, and Bax levels and increasing Bcl-2 expression. M2Φ-Exos suppressed pro-inflammatory cytokines (IL-4, IL-5, IL-13) and Transforming growth factor (TGF)-β, while enhancing anti-inflammatory cytokine IFN-γ and IL-10.

Conclusion: These findings demonstrate that M2Φ-Exos regulate the imbalance in epithelial proliferation and apoptosis in asthma, reducing inflammation and mitigating tissue remodeling, and provide new insights into potential therapeutic strategies for asthma management.

Keywords: Apoptosis; Asthma; Exosome; M2 macrophage; Proliferation.

PubMed Disclaimer

Conflict of interest statement

Declarations. Ethics approval and consent to participate: All experimental procedures were reviewed and approved by the Ethics Committee of the Children’s Hospital of Chongqing Medical University (Approval Numbers: 0918001). Consent for publication: Not applicable. Competing interests: The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Identification of M2Φ and M2Φ-Exosomes. A Morphological analysis of M2Φ under a microscope. Scale bar = 100 nm. B-G Gene expression of M2Φ-associated markers (Arg1, Mrc1, and YM1) and M1Φ-associated markers (TNF-α, IL-1β and CD86) quantified by RT-qPCR. H Expression of surface biomarkers F4/80 and CD206 on M2Φ, determined by flow cytometry. I Extraction process of M2Φ-derived exosomes (M2Φ-Exos). J Transmission electron microscopy (TEM) of exosomes isolated from the supernatant of M2Φ. Scale bar = 100 nm. K Size distribution of M2Φ-Exos measured by nanoparticle tracking analysis (NTA). L Western blot analysis of exosome marker proteins CD9, TSG101, and CD63, along with the endoplasmic reticulum marker protein Calnexin. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 2
Fig. 2
M2-Exosomes inhibit the proliferation of MLE-12 cells induced by HDM. A Immunofluorescence staining showing the uptake of PKH-26-labeled M2Φ-Exosomes (red) by MLE-12 cells (DAPI, blue; PKH-26, red; BF, bright field). Control: untreated cells (without PKH26). Scale bar = 10 μm. B Cell viability of MLE-12 cells analyzed by the CCK-8 assay following treatment with varying concentrations of M2Φ-Exosomes. C Migration ability of MLE-12 cells assessed by wound healing assays at 24 h and 48 h post-treatment. D Quantitative analysis of wound healing in MLE-12 cells. E Proliferation of MLE-12 cells analyzed by EdU incorporation assay. Scale bar = 50 μm. F Quantification of EdU-positive cells. G Immunofluorescence staining for PCNA in MLE-12 cells. Scale bar = 10 μm. H Quantification of the region of interest (ROI) mean intensity of PCNA. I Western blot analysis of PCNA expression in the three groups. J Quantification of grayscale values from Western blot analysis. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 3
Fig. 3
M2Φ-Exosomes reduce apoptosis in MLE-12 cells induced by HDM. A Apoptosis of MLE-12 cells was analyzed using Annexin V-FITC/PI staining by flow cytometry. B Flow cytometric analysis of apoptosis in MLE-12 cells. C Representative immunofluorescence images of TUNEL staining in the three groups. Scale bar = 10 μm. D Quantification of the region of interest (ROI) mean intensity of TUNEL staining. E Immunofluorescence staining for Cleaved-caspase3 in MLE-12 cells. Scale bar = 20 μm. F Quantification of ROI mean intensity of Cleaved-caspase3. G Representative western blots of caspase 3 and Cleaved-caspase3 proteins, and H, I quantification of grayscale values. J Representative western blots of Bax and Bcl2 proteins, and K, L quantification of grayscale values. N = 3/group. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant
Fig. 4
Fig. 4
M2Φ-Exosomes inhibit lung injury in HDM-induced asthmatic mice. A Schematic representation of the process used to establish the HDM-induced allergic asthma mouse model. M2Φ-Exosomes were administered intranasally as a reference treatment. B Representative H&E staining of lung tissue sections from the three experimental groups. Scale bars: 50 μm and 20 μm. C Lung inflammation scores in the mice across the different groups. D Collagen fibers in mouse lung tissues visualized by Masson’s trichrome staining. Scale bars: 50 μm and 20 μm. E Quantification of collagen volume fraction. F Total cell count in the bronchoalveolar lavage fluid (BALF). G Eosinophil count in the BALF. H Total serum IgE levels in the mice, as analyzed by ELISA. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 5
Fig. 5
M2Φ-Exosomes Inhibit Aberrant Airway Epithelial Cell Proliferation in Asthmatic Mice. A Intranasal administration of DiR-labeled M2Φ-Exosomes was detected via fluorescence imaging. B Localization of DiR-labeled M2Φ-Exosomes within lung tissues was observed using fluorescence imaging. C Representative Western blots showing PCNA protein levels in lung tissues. D Quantification of grayscale values of PCNA expression. E Immunohistochemical (IHC) staining of PCNA-positive proliferating cells in lung tissues. Scale bar: 50 μm and 20 μm. F Quantification of immunohistochemical staining. *P < 0.05, **P < 0.01
Fig. 6
Fig. 6
M2Φ-Exosomes inhibit apoptosis of airway epithelial cells in asthmatic mice. A Representative images of TUNEL staining in lung tissue from asthmatic mice. Scale bar = 100 μm. B Quantification of the region of interest (ROI) mean intensity of TUNEL staining. C Immunofluorescence staining for Cleaved-caspase3 in lung tissue of mice. Scale bar = 50 μm. D Quantification of ROI mean intensity of Cleaved-caspase3. E Representative western blots showing caspase 3 and Cleaved-caspase3 protein expression. F, G Quantification of grayscale values for caspase 3 and Cleaved-caspase3. H Representative western blots of Bax and Bcl2 protein expression. I, J Quantification of grayscale values for Bax and Bcl2. K IHC staining for Cleaved-caspase3-positive cells in lung tissues. Scale bars = 50 μm and 20 μm. L Quantification of immunohistochemical staining. *P < 0.05, **P < 0.01, ***P < 0.001, ns, not significant
Fig. 7
Fig. 7
M2Φ-Exosomes regulate inflammatory cytokines in asthmatic mice. Quantification of mRNA levels for (A) IL-4, (B) IL-5, (C) IL-13, (D) TGF-β, (E) IL-10 and (F) IFN-γ in lung tissue using qRT-PCR. Expression of (G) IL-4, (H) IL-5, (I) IL-13, (J) TGF-β, (K) IL-10 and (L) IFN-γ in BALF of mice detected by ELISA. *P < 0.05, **P < 0.01, ***P < 0.001

Similar articles

References

    1. Shipp CL, Gergen PJ, Gern JE, Matsui EC, Guilbert TW. Asthma management in children. J Allergy Clin Immunol Pract. 2023;11:9–18. - PubMed
    1. Zhu C, Sun Y, Zhao Y, Hou J, Zhang Q, Wang P. Associations between children’s asthma and allergic symptoms and phthalates in dust in metropolitan Tianjin, China. Chemosphere. 2022;302:134786. - PubMed
    1. Shin YH, Hwang J, Kwon R, Lee SW, Kim MS, Shin JI, Yon DK, GBD 2019 Allergic Disorders Collaborators. Global, regional, and National burden of allergic disorders and their risk factors in 204 countries and territories, from 1990 to 2019: a systematic analysis for the global burden of disease study 2019. Allergy. 2023;78:2232–54. - PMC - PubMed
    1. Zhou X, Zhang P, Tan H, Dong B, Jing Z, Wu H, Luo J, Zhang Y, Zhang J, Sun X. Progress in diagnosis and treatment of difficult-to-treat asthma in children. Ther Adv Respir Dis. 2023;17:17534666231213637. - PMC - PubMed
    1. Russell RJ, Boulet LP, Brightling CE, Pavord ID, Porsbjerg C, Dorscheid D, Sverrild A. The airway epithelium: an orchestrator of inflammation, a key structural barrier and a therapeutic target in severe asthma. Eur Respir J. 2024;63:2301397. - PMC - PubMed

LinkOut - more resources