Skip to main content

Fisetin reduces ovalbumin-triggered airway remodeling by preventing phenotypic switching of airway smooth muscle cells

Abstract

Background

The transformation of airway smooth muscle cells (ASMCs) from a quiescent phenotype to a hypersecretory and hypercontractile phenotype is a defining feature of asthmatic airway remodeling. Fisetin, a flavonoid compound, possesses anti-inflammatory characteristics in asthma; yet, its impact on airway remodeling and ASMCs phenotype transition has not been investigated.

Objectives

This research seeked to assess the impact of fisetin on ovalbumin (OVA) induced asthmatic airway remodeling and ASMCs phenotype transition, and clarify the mechanisms through network pharmacology predictions as well as in vivo and in vitro validation.

Methods

First, a fisetin-asthma-ASMCs network was constructed to identify potential targets. Subsequently, cellular and animal studies were carried out to examine the inhibitory effects of fisetin on airway remodeling in asthmatic mice, and to detemine how fisetin impacts the phenotypic transition of ASMCs.

Results

Network analysis indicated that fisetin might affect asthma via mediating the phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (AKT) pathway. Intraperitoneal administration of fisetin in vivo reduced airway inflammation and remodeling, as shown by reduced inflammatory cells, decreased T helper type 2 (Th2) cytokine release, diminished collagen accumulation, mitigated airway smooth muscle thickening, and decreased expression of osteopontin (OPN), collagen-I and α-smooth muscle actin (α-SMA). Moreover, fisetin suppressed the PI3K/AKT pathway in asthmatic lung tissue. According to the in vitro data, fisetin downregulated the expression of the synthetic phenotypic proteins OPN and collagen-I, contractile protein α-SMA, and inhibited cellular migration, potentially through the PI3K/AKT pathway.

Conclusion

These results suggest that fisetin inhibits airway remodeling in asthma by regulating ASMCs phenotypic shift, emphasizing that fisetin is a promising candidate for the treatment of airway smooth muscle remodeling.

Background

Asthma is a long-lasting, multifaceted disease with the pathological features such as airway inflammation and remodeling. Airway remodeling involves subepithelial fibrosis, increased smooth muscle bulk, and extracellular matrix aggregation [1]. Airway structural remodeling causes airway stenosis, restricted airflow, and decreased lung function, and severely affects the quality of life [2]. Glucocorticoids are widely used as first-line drugs for asthma, but are associated with a range of adverse drug reactions, including psychiatric symptoms, growth retardation, abnormalities of the teeth and rashes [3]. Thus, novel and affective anti-asthma agents are urgently needed.

Impaired airway smooth muscle cells (ASMCs) play important roles in airway structural remodeling. Extracellular matrix proteins and transforming growth factors, promote the shift of ASMCs from a dormant to a synthetic stage in vitro [4, 5]. ASMCs from asthmatic patients exhibit a preference for a synthetic phenotype compared to those from non-asthmatic donors [6]. ASMCs with a synthetic phenotype display features such as hypertrophy, migration, proliferation and parasecretion, leading to increased airway tension, worsened mechanics and exacerbated airway hyperresponsiveness [7]. Additionally, ASMCs contribute to airway stenosis and airway hyperresponsiveness by modulating cell contractility [8]. The expression of the contractile protein α-smooth muscle actin (α-SMA) in the airway is related to airway function in asthma [9]. Therefore, it is essential to develop treatments targeting ASMCs dysfunction.

The compound 3,3’,4’,7-tetrahydroxyflavone (fisetin) is a natural bioactive flavonoid widely derived from fruit and vegetable sources [10]. Fisetin has numerous biological functions, involving anti-aging, anti-cancer, anti-inflammatory, and anti-fibrotic properties [11,12,13,14]. Pervious research revealed that fisetin suppresses airway inflammation in an asthma model [15, 16]. Fisetin is known to have potent effects on a range of airway cell types. Fisetin inhibits lipopolysaccharide-induced neutrophil and macrophage infiltration and attenuates acute lung injury [17]. Fisetin significantly inhibits tumour necrosis factor alpha-induced Interleukin (IL)-8 levels in human airway epithelial cells, suggesting that fisetin is a potential agent for the treatment of inflammatory lung diseases [18]. Fisetin has the potential ability to inhibit cigarette smoke extract-induced epithelial-mesenchymal transition in airway epithelial cells [19]. However, its role in regulating airway remodeling and ASMCs function is still unknown.

Herein, our study focused on the impacts of fisetin in a murine model of asthma. The phenotypic switching and biological functions of ASMCs were examined to elucidate the potential mechanisms.

Materials and methods

Reagents

Fisetin was obtained from Beyotime (Shanghai, China). Ovalbumin (OVA) was acquired from Sigma-Aldrich (Taufkirchen, Germany). Antibodies against β-actin, GAPDH, Phosphoinositide 3-kinase (PI3K), p-PI3K, and the PI3K inhibitor LY294002 were obtained from Beyotime (Shanghai, China). Antibodies against Protein Kinase B (AKT), p-AKT, collagen-I, osteopontin (OPN) and α-SMA were acquired from Cell Signaling Technology (Beverly, USA).

Animals

Female BALB/c mice (6-weeks-old, weighing 18–20 g) were provided by Gem Pharmatech Company (Nanjing, China) and kept in standard laboratory conditions (22–24 °C, 49–51% humidity). Water and food were freely available to all animals. All animal studies were conducted following National Institutes of Health (NIH) guidelines and authorized according to the Ethics Committee of the Second Affiliated Hospital of Shandong First Medical University.

Network pharmacology

The simplified molecular input line entry system (SMILES) structure for fisetin was retrieved from PubChem website. The structure date file (SDF) format were submitted into the Swiss Target prediction database (http://www.swisstargetprediction.ch/) to facilitate further targeting prediction [20]. Targets related to asthma and ASMC were gathered from the GeneCards (https://www.genecards.org/) and Online Mendelian Inheritance in Man (OMIM) databases (https://omim.org/) [21, 22]. The overlapping genes related to fisetin, asthma, and ASMC were considered potentially valuable for asthma treatment. Metascape was implemented to perform gene functional enrichment analysis (https://metascape.org/) [23]. Protein-protein interaction (PPI) networks were created and further analyzed to classify hub genes via the Search Tool for the Retrieval of Interaction Genes/Proteins (STRING) (https://string-db.org/) [24]. Topological analysis and visualization were conducted using the Cytoscape plug-in NetworkAnalyzer.

Asthmatic model establishment and fisetin treatment

Mice were randomly assigned to the following five groups (n = 10 per group): (1) control group, (2) OVA group, OVA-sensitized/OVA-exposed; (3) OVA + FL group, OVA-sensitized/OVA-exposed + low dose of fisetin (2.5 mg/kg); (4) OVA + FH group, OVA-sensitized/OVA-exposed + high dose of fisetin (5 mg/kg); (5) OVA + DEX group, OVA-sensitized/OVA-exposed + 1 mg/kg dexamethasone. The mice were administered 100 µL of OVA mixed with 2 mg of aluminum hydroxide intraperitoneally on days 1 and 8. During days 15–43, the animals were nebulized using 2.5% OVA for 30 min/day, and were then administered three times weekly for 4 weeks using an ultrasonic nebulizer. The drug-treated groups received fisetin or dexamethasone 30 min before each OVA challenge. Dexamethasone was used as a positive control because it is an established first-line treatment of asthma. Figure 1 reveals the experimental strategies.

Fig. 1
figure 1

Experimental strategies and schedule

Examination of bronchoalveolar lavage fluid (BALF)

The animals were intratracheally intubated, and 750 µL of saline was administered three times after euthanasia. The BALF was spun at 2000 rpm and 4 ℃ for 15 min. The pellet was reconstituted and counted with a hemocytometer. Smears were Giemsa-stained and screened under light microscopy. The BALF supernatants were used for cytokine analysis. IL-4, IL-5, and IL-13 levels in BALF were measured with enzyme-linked immunosorbent assay (ELISA) panels. Five mice from each group were randomly selected for cell counts and cytokine analysis.

Lung histopathology

Lung samples were paraffin-embedded and split to 5-µm slices. Hematoxylin and eosin (H&E) staining was employed for assessing pulmonary pathological changes, whereas Masson staining was employed to analyze peribronchial collagen deposition. Periodic acid-Schiff (PAS) staining was used to examine goblet cell hyperplasia. Image-Pro Plus software was used to quantify the bronchial smooth muscle area (WAm), Masson stained area, and perimeter of the bronchial basement membrane (Pbm). Analyses were performed to determine the bronchial smooth muscle thickness (WAm/Pbm), Masson stained area/Pbm and PAS stained area/Pbm. Images were photographed under light microscopy at 400× magnification. Five mice from each group were randomly selected for histological examination, and five bronchioles per mouse were quantified.

Immunohistochemistry

Paraffin samples were dewaxed, rehydrated, subjected to antigen retrieval, and blocked with serum. Afterward, the sections were treated with primary antibodies targeting collagen-I (1:200), OPN (1:100), and α-SMA (1:200) overnight at 4 ℃, and then exposed to secondary antibodies for 1 h. Freshly prepared DAB was applied for chromogenic detection. Photographs of the sections were obtained using a light microscope at 400× magnification. Five mice from each group were randomly selected for immunohistochemical analysis, and five bronchioles per mouse were quantified.

Cell cultivation

Mouse airway smooth muscle cells (MASMCs) were obtained from Otwo Biotech (Shenzhen, China). MASMCs were cultivated in Dulbeccos modified Eagle medium (DMEM) containing 10% fetal bovine serum, and 1% penicillin/streptomycin in 5% CO2 environment.

Cell cytotoxicity assays

Fisetin at 0, 12.5, 25, 50, 100, and 200 µM was applied to the MASMCs for 24 h. Subsequently, a cell counting kit-8 (CCK-8) test was used to evaluate cellular cytotoxicity. This procedure was performed as follows: MASMCs were placed in 96-well dishes (3000 cells per well) overnight and exposed to various doses of fisetin. After the supernatant was discarded, CCK-8 solution was applied to stain the cells for 30 min. Absorbance was analyzed via a spectrophotometer (CANY, Shanghai, China). The cell cytotoxicity assays were repeated thrice.

Wound healing assays

The MASMCs were placed in 6-well dishes with a seeding density of 5 × 105 cells/well. Upon reaching 90–100% confluence, a wound was simulated by creating a straight scratch with a 200 µL micropipette tip. Cell debris from the scratches was rinsed with PBS, and the cells were exposed to serum-free DMEM. Images at 0 and 24 h were observed using an inverted microscope. The scratch area was measured using ImageJ software. The wound healing assay was performed in triplicate.

Western blotting

Lung tissues and MASMCs protein were homogenized and treated with a protease inhibitor cocktail in RIPA. After 5 min spinning at 12,000 rpm, the protein supernates were gathered. Protein samples (20 µg) were resolved through sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently electroblotted on polyvinylidene fluoride (PVDF) membranes. Following a 2-hour blocking step, the PVDF membranes were then exposed to primary antibodies overnight with PI3K (1:1000), p-PI3K (1:1000), AKT (1:1000), p-AKT (1:1000), collagen-I (1:1000), OPN (1:1000), α-SMA (1:1000), β-actin (1:1000) and GAPDH (1:1000). Following incubation with secondary antibodies, the protein bands were revealed with Tanon High-sig ECL Western blotting assay reagent (Tanon, Shanghai, China). Three mice from each group were randomly selected for Western blotting analysis, and the cell Western blotting experiments were performed in triplicate.

Immunofluorescence

MASMCs were cultured on slides and treated according to the established experimental protocol. After being subjected to cellular fixation, membrane breaking, and serum closure, cell slides were exposed to collagen-I (1:200), OPN (1:100), and α-SMA (1:300) at 4 °C. Thereafter, the slides were subjected to fluorescent-labeled secondary antibodies for 2 h in a dark chamber. Finally, the nuclei were stained using DAPI, and samples were sealed with an antifade medium. Images were taken under fluorescent light and processed using ImageJ software. The cell immunofluorescence experiments were performed in triplicate.

Statistical analysis

All experiments were performed with three to five biological replicates and three experimental replicates. GraphPad Prism 8 was utilized to construct graphs and perform data analysis. Results are displayed as mean ± SEM. One-way ANOVA followed by Tukey’s test was implemented to compare groups. P < 0.05 was deemed significantly different.

Results

Network pharmacology identified prospective target genes for fisetin in treating asthma

100 targets of fisetin were obtained from Swiss Target Prediction. Following the elimination of redundant targets, 2551 asthma-associated targets and 5153 ASMC- associated targets were collected from the GeneCards (relevance score > 1) and OMIM databases. Then, 34 intersecting targets were identified through a Venn diagram (Fig. 2A). These overlapping targets are considered as possible targets for the application of fisetin in asthma therapy. Next, enrichment analysis was conducted using Metascape. The primary biological activities focused on biological activities such as controlling inflammation and cell growth, IL-4 and IL-13 signaling, and white blood cell movement, all of which are strongly associated with asthma. Additionally, the data showed significant enrichment in the PI3K/AKT signaling pathway. The top 20 significant genes are shown in Fig. 2B and C.

Analysis of the network structure of 34 common targets indicated that fisetin could potentially targets AKT1, matrix metalloprotein 9 (MMP9), epidermal growth factor receptor (EGFR), and other genes for asthma treatment (Fig. 2D). In the interaction network constructed using Cytoscape software, AKT1, which exhibited the highest maximal clique centrality (MCC), betweenness centrality, and degree values, is considered the predominant node (Fig. 2E). These findings suggest that the PI3K/AKT pathway appears to be enriched and may be involved in the mechanism of fisetin therapy for asthma.

Fig. 2
figure 2

Performing enrichment analysis, constructing PPI networks, and analyzing modules. (A) Venn diagram of fisetin targets, asthma-related genes, and ASMC-related genes. (B) Metascape bar graph of enriched terms showing the top 20 targets. The enhanced terms were prioritized according to p-values, with more vibrant colors representing smaller p-values. (C) Metascape visualization of the networks of the top 20 clusters, with each cluster distinguished by a unique color based on its cluster ID. Nodes with the same cluster ID exhibit stronger interconnections. (D) Construction of a PPI network for genes with increased expression. (E) The top 10 targets were ranked according to MCC, betweenness centrality, and degree, as performed using the plug-in of Cytoscape

Fisetin alleviated airway inflammation in asthmatic mice

To ascertain the protective role of fisetin in asthmatic mice, we examined its influence on airway inflammation. Airway inflammation is driven by aberrant Th2 cells that generate cytokines, including IL-4, IL-5, and IL-13 [25]. Our results displayed that mice exposed to OVA exhibited higher levels of aforementioned cytokines. In comparison to mice challenged by OVA, administration of fisetin or dexamethasone diminished these cytokines levels (Fig. 3A-C). The findings are aligned with the network analysis results.

The analysis of pulmonary inflammation was conducted using BALF cell countsas well as peribronchial and perivascular inflammatory cell infiltration. Our results showed that BALF from saline control mice contained no inflammatory cells. OVA-treated mice had higher BALF counts of neutrophils, eosinophils, and total cells. Fisetin or dexamethasone reduced the inflammatory cell counts compared to asthmatic mice (Fig. 3D-F).

Fig. 3
figure 3

Fisetin attenuated OVA-challenged airway inflammation. IL-4 (A), IL-5 (B), and IL-13 (C) in BALF were quantified using ELISA (n = 5). Total cells (D), eosinophils (E), and neutrophils (F) in BALF were counted (n = 5). Results are displayed as mean ± SEM. ***P < 0.001 vs. control, #P < 0.05, ##P < 0.01, and ###P < 0.001 vs. OVA

Fisetin alleviated airway remodeling in asthmatic mice

H&E staining revealed more peribronchial and perivascular inflammatory lesions in asthmatic mice than in control mice. Fisetin or dexamethasone decreased these inflammatory cell infiltration in the lungs (Fig. 4A). Similarly, fisetin prevented airway structural remodeling in OVA-treated mice. Airway remodeling was assessed using WAm/Pbm, Masson stained area/Pbm and PAS stained area/Pbm. As shown by H&E staining, OVA challenge caused a notable rise in peribronchial smooth muscle thickness compared with that of the controls, and the levels of thickness were mitigated by treatment with fisetin or dexamethasone (Fig. 4A and B). Masson staining showed that the increased peribronchial collagen deposition induced by OVA was inhibited by fisetin or dexamethasone (Fig. 4C and D). PAS staining showed that OVA challenges resulted in marked goblet cell hyperplasia and mucus plugging, which were inhibited by fisetin or dexamethasone (Fig. 4E and F).

Fig. 4
figure 4

Fisetin alleviated OVA-challenged airway remodeling. (A) Histopathological analysis was performed using H&E staining. (B) Quantification of WAm/Pbm (n = 5). (C) Collagen accumulation was detected through Masson staining. (D) Quantification of Masson stained area/Pbm (n = 5). (E) Goblet cell hyperplasia was detected using PAS staining. (F) Quantification of PAS stained area/Pbm (n = 5). Results are represented as the mean ± SEM. ***P < 0.001 vs. control, #P < 0.05, and ##P < 0.01 vs. OVA

Fisetin alleviated OPN, collagen-I and α-SMA levels in asthmatic mice

Asthma biomarkers such as OPN, collagen-I and α-SMA which are also considered novel therapeutic targets, are related to asthma severity and airflow limitations [26, 27]. Immunohistochemical analyses were employed to evaluate OPN expression. Bronchiolar and pulmonary OPN expression was significantly increased in OVA-treated mice, whereas fisetin or dexamethasone treatment decreased OPN expression (Fig. 5A and B). Immunohistochemistry assays also showed an increase in collagen-I and α-SMA staining surrounding the bronchioles in OVA-challenged mice compared with the controls. Fisetin or dexamethasone treatment reversed these increases (Fig. 5C − 5 F).

Western blotting analysis yielded consistent results (Fig. 5G). OPN, collagen-I and α-SMA levels of OVA-challenged mice were much higher than those of the controls. Fisetin or dexamethasone reduced OPN, collagen-I and α-SMA expression compared to OVA-challenged mice (Fig. 5H − 5 J).

Fig. 5
figure 5

Fisetin suppressed OPN, collagen-I and α-SMA levels in asthmatic mice. (A, C, E) Immunohistochemical staining was performed on lung sections to detect OPN, collagen-I and α-SMA expression. (B, D, F) Integrated optical density (IOD) detection of OPN, collagen-I and α-SMA immunohistochemical staining (n = 5). (G) Western blotting images of OPN, collagen-I and α-SMA. (H, I, J) Quantification of protein expression (n = 3). Results are represented as mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. control; #P < 0.05, and ##P < 0.01 vs. OVA

Fisetin inactivated the PI3K/AKT pathway in asthmatic mice

Western blotting was employed to analyze the PI3K/AKT pathway in the lungs. Our findings exhibited that PI3K and AKT phosphorylation levels were greatly raised in OVA-induced mice, whereas fisetin or dexamethasone notably prevented their phosphorylation. There was no discernible variation in total PI3K and AKT expression between the groups (Fig. 6).

Fig. 6
figure 6

Fisetin inactivated the PI3K/AKT pathway in asthmatic mice. (A) Western blotting detection of p-PI3K, PI3K, p-AKT, AKT, and GAPDH. (B, C) Quantification of protein expression (n = 3). Results are represented as mean ± SEM. *P < 0.05 vs. control; #P < 0.05, and ##P < 0.01 vs. OVA

Effect of fisetin on MASMCs cytotoxicity

The cytotoxic effects of fisetin on MASMCs were evaluated in vitro using the CCK-8 assay. The findings indicated that fisetin did not display any noticeable toxicity at low doses ranging from 0 µM to 100 µM. In contrast, 200 µM fisetin inhibited cell viability after 24 h (Fig. 7). Thus, we selected fisetin concentrations below 100 µM for subsequent experiments.

Fig. 7
figure 7

Effects of fisetin on MASMCs cytotoxicity (n = 3). Results are represented as mean ± SEM. *P < 0.05 vs. control

Effects of fisetin on MASMCs phenotypic switching and migration

Increasing evidence suggests that ASMCs in individuals with asthma tend to display a synthetic and contractile phenotype [28]. The increased expression of OPN and collagen-I which are extracellular matrix components, indicates ASMC transformation to a synthetic phenotype. To ascertain whether fisetin regulates synthetic phenotype switching, we investigated its impact on the expression of OPN and collagen-I. According to our findings, fisetin treatment for 24 h displayed a dose-dependent decline in OPN and collagen-I levels (Fig. 8A-C). These findings indicate that fisetin prevents MASMCs from transitioning to a synthetic phenotype. As a marker of ASMCs, α-SMA plays a key role in cell proliferation, migration and contraction [29, 30]. In our study, we investigated the effect of fisetin on α-SMA. We found that fisetin treatment for 24 h reduced α-SMA levels in a dose-dependent manner (Fig. 8A and D).

Prior research has indicated that the change of ASMCs to a synthetic type can enhance cell migration [31, 32]. Subsequently, a wound closure test was performed to evaluate migratory capacity. Our findings indicated that fisetin treatment for 24 h inhibited MASMCs migration in a dosage-dependent way (Fig. 8E and F).

Fig. 8
figure 8

Effects of fisetin on MASMCs phenotypic switching and migration. (A) Western blotting bands of OPN, collagen-I, α-SMA and GAPDH. (B, C, D) Quantification of protein expression (n = 3). (E) Wound healing assay. (F) Relative wound area (n = 3). Results are represented as mean ± SEM. *P < 0.05, and **P < 0.01 vs. control

Effects of fisetin on the PI3K/AKT pathway in MASMCs

The PI3K/AKT pathway regulates many cellular processes, including phenotypic switching and migration [33, 34]. Therefore, we sought to determine whether fisetin modulates the PI3K/AKT pathway. After 1 h of fisetin treatment, our investigation indicated a dosage-dependent decline in p-PI3K and p-AKT levels in MASMCs. However, the PI3K and AKT levels remained unchanged (Fig. 9).

Fig. 9
figure 9

Effect of fisetin on the PI3K/AKT pathway in MASMCs. (A) Western blotting bands of p-PI3K, PI3K, p-AKT, AKT and GAPDH. (B, C) Quantification of expression (n = 3). Results are represented as mean ± SEM.*P < 0.05, **P < 0.01, and ***P < 0.001 vs. control

Fisetin altered the MASMCs phenotype via the PI3K/AKT pathway

To elucidate the impact of the PI3K/AKT pathway on fisetin-induced phenotypic switching, a PI3K inhibitor (LY294002) was selected to disrupt the PI3K/AKT pathway [35]. After 30 min of preincubation with 5 µM LY294002, MASMCs were cultured with or without 100 µM fisetin for 24 h. Subsequently, OPN, collagen-I and α-SMA expression were assessed via Western blotting and immunofluorescence. As displayed in Fig. 10, Western blotting assays showed that the levels of OPN, collagen-I and α-SMA were reduced by fisetin or LY294002. Compared to LY294002 treatment, cells treated with both LY294002 and fisetin showed further reductions in OPN, collagen-I and α-SMA levels. Similarly, immunofluorescence staining showed that fisetin or LY294002 decreased levels of OPN, collagen-I and α-SMA compared to the controls. Combined treatment with fisetin and LY294002 further downregulated the expression of OPN, collagen-I and α-SMA compared with LY294002 treatment alone. These results imply that fisetin-regulated phenotypic switching in MASMCs may be mediated by the PI3K/AKT pathway.

Fig. 10
figure 10

Fisetin altered the MASMC phenotype by regulating the PI3K/AKT pathway. (A) Western blotting bands of OPN, collagen-I, α-SMA and GAPDH. (B, C, D) Quantification of expression (n = 3). (E, G, I) Representative immunofluorescence of OPN, collagen-I and α-SMA (magnification ×200). (F, H, J) Relative immunofluorescence intensity of OPN, collagen-I and α-SMA (n = 3). Results are represented as mean ± SEM.*P < 0.05 and **P < 0.01 vs. control; #P < 0.05, and ##P < 0.01 vs. LY294002

Discussion

Airway remodeling involves the abnormal restructuring of cells and molecules in asthmatic airways, and is characterized by goblet cell hyperplasia, subepithelial fibrosis, hyperproliferation, and hypertrophy of ASMCs [36]. ASMCs from asthmatic donors tend to shift towards a hypersecretory and hypercontractile phenotype compared to those from normal donors [6, 28]. The phenotypic switching of ASMCs can lead to airflow limitation and airway remodeling [37]. Thus, regulating ASMC phenotypic switching may be a novel approach for controlling airway remodeling.

Fisetin is a bioactive polyphenol flavonoid derived from various vegetables, fruits, and nuts [12]. It possesses diverse pharmacological characteristics, involving anti-cancer, anti-aging, and anti-inflammatory actions [38, 39]. Huang W and Wu S at el. revealed the advantages of fisetin in reducing airway inflammation in asthmatic mice [15, 16]. Nevertheless, the contributions of fisetin to airway remodeling are not well understood. Besides, fisetin has been demonstrated the capability to relax ASMCs and prevent airway smooth muscle constriction [40]. Whether fisetin regulates the synthetic ability of ASMCs is unknown. Given its widespread role as a senotherapeutic drug, discovering the effects of fisetin on asthma could expand its therapeutic potential.

Our experiments evaluated the impact of fisetin on asthmatic mice, focusing on the phenotypic switching of MASMCs. In this study, we identified 2551 asthma-related targets, 5153 ASMC-related targets, and 100 fisetin-related targets, yielding 34 common targets. A bar graph summary was generated using Metascape Gene List Analysis. Functional enrichment analysis discovered that these targets primarily participate in controlling cell movement and tissue remodeling, which were partly confirmed in experiments using mouse models of asthma and MASMCs. We found that treatment with fisetin significantly suppressed airway inflammation and remodeling. Severe inflammation, characterized by increased Th2 cytokine levels and elevated inflammatory cell recruitment, was observed in mice challenged with OVA. Fisetin markedly attenuated the resulting airway inflammation. Additionally, fisetin significantly alleviated the pathological characteristics caused by OVA, such as airway smooth muscle thickening, subepithelial collagen accumulation and goblet cell hyperplasia. Given the significant role of enhanced airway smooth muscle mass in airway remodeling [28], we investigated how fisetin impacts MASMCs. Our results showed that fisetin inhibited the phenotypic switching and migration of MASMCs. This discovery offers new perspectives on the ability of fisetin to prevent remodeling, particularly its effects on ASMC phenotypic switching.

Smooth muscle cells transitioning to a synthetic phenotype generate more extracellular matrix components, which causes airway remodeling in asthma. OPN, also termed secreted phosphoprotein 1, functions as an extracellular matrix ingredient as well as a pro-inflammatory cytokine [41]. Asthmatic individuals exhibit elevated OPN levels in their lungs, particularly in ASMCs, epithelial cells, and myofibroblasts [42]. In addition, OPN subepithelial expression levels are related to disease severity [42]. Consistent with these findings, OPN levels are elevated in house dust mites and OVA-challenged asthmatic mice. Furthermore, OPN contributes to eosinophil recruitment by binding to eosinophil-recruiting chemokines [43]. OPN also promotes airway remodeling by enhancing the proliferation, migration, and collagen secretion of pulmonary fibroblasts [44]. Thus, we investigated this novel asthma biomarker in our study. We found that OPN levels were elevated in mice treated with OVA, but significantly decreased after administration of fisetin or DEX. Consistent results were obtained in vitro, showing that fisetin inhibited OPN expression in a dose-dependent manner. Collagen-I is not only a key marker of airway remodeling, but also an indicator of the synthetic phenotype of ASMCs [45]. In this study, fisetin inhibited the upregulation of collagen-I expression in both cell and animal experiments. Our findings suggest that fisetin may inhibit the transformation of synthetic phenotypes.

As one of the main contractile proteins, α-SMA is considered an important indicator of airway remodeling. Exorbitant α-SMA expression serves to accelerate cell proliferation, migration and contraction, resulting in airway remodeling and airway.

hyperresponsiveness [9, 29, 30]. In our study, the results demonstrated that α-SMA levels were elevated in mice treated with OVA, but significantly decreased after administration of fisetin or DEX. Further in vitro studies also revealed that fiestin inhibited α-SMA expression. These results indicate that fisetin may regulate the contractile phenotype of ASMC.

We further examined the effect of fisetin on the migration ability of MASMCs, and the results revealed that fisetin impeded MASMCs migration in a dose-dependent manner. Previous studies have shown that MASMCs characterised by the synthetic phenotype exhibit enhanced cell migration [31, 32]. Owing to the regulation effect of fisetin on the MASMC phenotype, we speculate that the inhibitory effects of fisetin on migration are related to its ability to regulate phenotypic switching.

Based on this, the molecular mechanisms of fisetin in asthma therapy are worthy of in-depth exploration. Network pharmacology analysis revealed that the enriched pathways were also linked to the PI3K/AKT pathway. According to the values obtained from Cytoscape software, 10 core targets were identified. Among them, AKT1 has the highest degree and median centrality values, prompting us to investigate the PI3K/AKT pathway.

The PI3K/AKT pathway is a critical signaling axis that coordinates cellular growth, mobility, and differentiation [46,47,48]. Multiple lines of evidence support that fisetin could suppress the PI3K/AKT pathway in different cells [49,50,51]. Our conclusions are consistent with previous studies. We found that fisetin dosage-dependently blocked PI3K and AKT phosphorylation in ASMCs. In addition, it was noted that the combination of fisetin and LY294002 suppressed OPN, collagen-I and α-SMA expression more effectively than either fisetin or LY294002 individually. These findings suggest that the ability of fisetin to inhibit the phenotypic switching of ASMCs may be mediated by the PI3K/AKT pathway. Research indicates that inhibiting PI3K/AKT signal transduction can attenuate airway remodeling and airway inflammation in asthma [52, 53]. We found that fisetin downregulated p-PI3K and p-AKT levels compared with those in asthmatic mice, indicating that the PI3K/AKT pathway is indispensable for the protective effect of fisetin on asthmatic airway remodeling.

Conclusion

Overall, our research demonstrated that fisetin regulates the phenotype switching of MASMCs and alleviates asthmatic airway remodeling by targeting the PI3K/AKT signaling pathway. Our study indicates that fisetin might be utilized as a treatment for asthma.

Data availability

The data supporting the current study’s findings are available from the corresponding author upon reasonable request.

Abbreviations

ASMCs:

Airway smooth muscle cells

OVA:

Ovalbumin

PI3K:

Phosphoinositide 3-kinase

AKT:

Protein Kinase B

Th2:

T helper type 2

OPN:

Osteopontin

α-SMA:

α-smooth muscle actin

NIH:

National Institutes of Health

BALF:

Bronchoalveolar lavage fluid

DEX:

Dexamethasone

IL:

Interleukin

ELISA:

Enzyme linked immunosorbent assay

H&E:

Hematoxylin and eosin

WAm:

Bronchial smooth muscle area

Pbm:

Bronchial basement membrane

MASMCs:

Mouse airway smooth muscle cells

DMEM:

Dulbeccos modified Eagle medium

CCK-8:

Cell counting kit-8

SDS-PAGE:

Sodium dodecyl sulfate polyacrylamide gel electrophoresis

PVDF:

Polyvinylidene fluoride

SMILES:

Simplified molecular input line entry system

SDF:

Structure data file

OMIM:

Online Mendelian Inheritance in Man

PPI:

Protein-protein interaction

STRING:

Search Tool for the Retrieval of Interaction Gene/Proteins

MMP9:

Matrix metalloprotein 9

EGFR:

Epidermal growth factor receptor

MCC:

Maximal clique centrality

References

  1. Savin IA, Zenkova MA, Sen’kova AV. Bronchial asthma, Airway Remodeling and Lung Fibrosis as Successive steps of one process. Int J Mol Sci 2023; 24:16042.

  2. Winkler T, Frey U. Airway remodeling: shifting the trigger point for exacerbations in asthma. J Allergy Clin Immunol. 2021;148:710–2.

    Article  PubMed  Google Scholar 

  3. de Vries TW, de Langen-Wouterse JJ, van Puijenbroek E, Duiverman EJ. De Jong-Van den Berg LT: reported adverse drug reactions during the use of inhaled steroids in children with asthma in the Netherlands. Eur J Clin Pharmacol. 2006;62:343–6.

    Article  CAS  PubMed  Google Scholar 

  4. Wright DB, Trian T, Siddiqui S, Pascoe CD, Johnson JR, Dekkers BG, Dakshinamurti S, Bagchi R, Burgess JK, Kanabar V, Ojo OO. Phenotype modulation of airway smooth muscle in asthma. Pulm Pharmacol Ther. 2013;26:42–9.

    Article  CAS  PubMed  Google Scholar 

  5. Dekkers BG, Schaafsma D, Nelemans SA, Zaagsma J, Meurs H. Extracellular matrix proteins differentially regulate airway smooth muscle phenotype and function. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1405–1413.

    Article  CAS  PubMed  Google Scholar 

  6. Johnson PR, Burgess JK, Underwood PA, Au W, Poniris MH, Tamm M, Ge Q, Roth M, Black JL. Extracellular matrix proteins modulate asthmatic airway smooth muscle cell proliferation via an autocrine mechanism. J Allergy Clin Immunol. 2004;113:690–6.

    Article  CAS  PubMed  Google Scholar 

  7. Chetty A, Nielsen HC. Targeting Airway smooth muscle hypertrophy in Asthma: an Approach whose time has come. J Asthma Allergy. 2021;14:539–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Khalfaoui L, Pabelick CM. Airway smooth muscle in contractility and remodeling of asthma: potential drug target mechanisms. Expert Opin Ther Targets. 2023;27:19–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Slats AM, Janssen K, van Schadewijk A, van der Plas DT, Schot R, van den Aardweg JG, de Jongste JC, Hiemstra PS, Mauad T, Rabe KF, Sterk PJ. Expression of smooth muscle and extracellular matrix proteins in relation to airway function in asthma. J Allergy Clin Immunol. 2008;121:1196–202.

    Article  CAS  PubMed  Google Scholar 

  10. Khan N, Syed DN, Ahmad N, Mukhtar H. Fisetin: a dietary antioxidant for health promotion. Antioxid Redox Signal. 2013;19:151–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shen J, Shan J, Zhong L, Liang B, Zhang D, Li M, Tang H. Dietary phytochemicals that can extend longevity by regulation of metabolism. Plant Foods Hum Nutr. 2022;77:12–9.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kubina R, Krzykawski K, Kabala-Dzik A, Wojtyczka RD, Chodurek E, Dziedzic A. Fisetin, a potent Anticancer Flavonol exhibiting cytotoxic activity against neoplastic malignant cells and cancerous conditions: A Scoping. Comprehensive Review. 2022;14:2604.

  13. Nabizadeh Z, Nasrollahzadeh M, Shabani AA, Mirmohammadkhani M, Nasrabadi D. Evaluation of the anti-inflammatory activity of fisetin-loaded nanoparticles in an in vitro model of osteoarthritis. Sci Rep. 2023;13:15494.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang B, Yang LN, Yang LT, Liang Y, Guo F, Fu P, Ma L. Fisetin ameliorates fibrotic kidney disease in mice via inhibiting ACSL4-mediated tubular ferroptosis. Acta Pharmacol Sin. 2024;45:150–65.

    Article  CAS  PubMed  Google Scholar 

  15. Wu SJ, Huang WC, Cheng CY, Wang MC, Cheng SC, Liou CJ. Fisetin Suppresses the Inflammatory Response and Oxidative Stress in Bronchial Epithelial Cells. Nutrients. 2022;14:1841.

  16. Huang W, Li ML, Xia MY, Shao JY. Fisetin-treatment alleviates airway inflammation through inhbition of MyD88/NF-kappaB signaling pathway. Int J Mol Med. 2018;42:208–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Feng G, Jiang ZY, Sun B, Fu J, Li TZ. Fisetin alleviates Lipopolysaccharide-Induced Acute Lung Injury via TLR4-Mediated NF-kappaB signaling pathway in rats. Inflammation. 2016;39:148–57.

    Article  CAS  PubMed  Google Scholar 

  18. Lee S, Ro H, In HJ, Choi JH, Kim MO, Lee J, Hong ST, Lee SU. Fisetin inhibits TNF-alpha/NF-kappaB-induced IL-8 expression by targeting PKCdelta in human airway epithelial cells. Cytokine. 2018;108:247–54.

    Article  CAS  PubMed  Google Scholar 

  19. Agraval H, Yadav UCS. MMP-2 and MMP-9 mediate cigarette smoke extract-induced epithelial-mesenchymal transition in airway epithelial cells via EGFR/Akt/GSK3beta/beta-catenin pathway: amelioration by fisetin. Chem Biol Interact. 2019;314:108846.

    Article  CAS  PubMed  Google Scholar 

  20. Daina A, Michielin O, Zoete V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019;47:W357–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rebhan M, Chalifa-Caspi V, Prilusky J, Lancet D. GeneCards: integrating information about genes, proteins and diseases. Trends Genet. 1997;13:163.

    Article  CAS  PubMed  Google Scholar 

  22. Amberger JS, Bocchini CA, Schiettecatte F, Scott AF, Hamosh A. OMIM.org: online mendelian inheritance in man (OMIM(R)), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43:D789–798.

    Article  PubMed  Google Scholar 

  23. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, Chanda SK. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49:D605–12.

    Article  CAS  PubMed  Google Scholar 

  25. Hammad H, Lambrecht BN. The basic immunology of asthma. Cell. 2021;184:1469–85.

    Article  CAS  PubMed  Google Scholar 

  26. Kim MA, Shin YS, Pham le D, Park HS. Adult asthma biomarkers. Curr Opin Allergy Clin Immunol. 2014;14:49–54.

    Article  CAS  PubMed  Google Scholar 

  27. Reeves SR, Barrow KA, Kolstad TK, White MP, Rich LM, Wight TN, Debley JS. Fibroblast gene expression following asthmatic bronchial epithelial cell conditioning correlates with epithelial donor lung function and exacerbation history. Sci Rep. 2018;8:15768.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zuyderduyn S, Sukkar MB, Fust A, Dhaliwal S, Burgess JK. Treating asthma means treating airway smooth muscle cells. Eur Respir J. 2008;32:265–74.

    Article  CAS  PubMed  Google Scholar 

  29. Xu GN, Yang K, Xu ZP, Zhu L, Hou LN, Qi H, Chen HZ, Cui YY. Protective effects of anisodamine on cigarette smoke extract-induced airway smooth muscle cell proliferation and tracheal contractility. Toxicol Appl Pharmacol. 2012;262:70–9.

    Article  CAS  PubMed  Google Scholar 

  30. Bentley JK, Hershenson MB. Airway smooth muscle growth in asthma: proliferation, hypertrophy, and migration. Proc Am Thorac Soc. 2008;5:89–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. An Z, Liu Y, Song ZG, Tang H, Yuan Y, Xu ZY. Mechanisms of aortic dissection smooth muscle cell phenotype switch. J Thorac Cardiovasc Surg. 2017;154:1511–e15211516.

    Article  PubMed  Google Scholar 

  32. Henderson N, Markwick LJ, Elshaw SR, Freyer AM, Knox AJ, Johnson SR. Collagen I and thrombin activate MMP-2 by MMP-14-dependent and -independent pathways: implications for airway smooth muscle migration. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1030–1038.

    Article  CAS  PubMed  Google Scholar 

  33. Wang H, Zhong B, Geng Y, Hao J, Jin Q, Zhang Y, Dong L, Gao D, Li J, Hou W. TIPE2 inhibits PDGF-BB-induced phenotype switching in airway smooth muscle cells through the PI3K/Akt signaling pathway. Respir Res. 2021;22:238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang W, Guo J, Wang Y. MicroRNA-30b-5p promotes the proliferation and migration of human airway smooth muscle cells induced by platelet-derived growth factor by targeting phosphatase and tensin homolog deleted on chromosome ten. Bioengineered. 2021;12:3662–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem. 1994;269:5241–8.

    Article  CAS  PubMed  Google Scholar 

  36. Nayak AP, Deshpande DA, Penn RB. New targets for resolution of airway remodeling in obstructive lung diseases. F1000Res. 2018;7:F1000.

  37. Kume H. Role of Airway Smooth Muscle in inflammation related to Asthma and COPD. Adv Exp Med Biol. 2021;1303:139–72.

    Article  CAS  PubMed  Google Scholar 

  38. Yousefzadeh MJ, Zhu Y, McGowan SJ, Angelini L, Fuhrmann-Stroissnigg H, Xu M, Ling YY, Melos KI, Pirtskhalava T, Inman CL, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kashyap D, Garg VK, Tuli HS, Yerer MB, Sak K, Sharma AK, Kumar M, Aggarwal V, Sandhu SS. Fisetin and Quercetin: Promising Flavonoids with Chemopreventive Potential. Biomolecules 2019, 9.

  40. Brown A, Danielsson J, Townsend EA, Zhang Y, Perez-Zoghbi JF, Emala CW, Sr., Gallos G. Attenuation of airway smooth muscle contractility via flavonol-mediated inhibition of phospholipase-cbeta. Am J Physiol Lung Cell Mol Physiol. 2016;310:L747–758.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ashkar S, Weber GF, Panoutsakopoulou V, Sanchirico ME, Jansson M, Zawaideh S, Rittling SR, Denhardt DT, Glimcher MJ, Cantor H. Eta-1 (osteopontin): an early component of type-1 (cell-mediated) immunity. Science. 2000;287:860–4.

    Article  CAS  PubMed  Google Scholar 

  42. Samitas K, Zervas E, Vittorakis S, Semitekolou M, Alissafi T, Bossios A, Gogos H, Economidou E, Lotvall J, Xanthou G, et al. Osteopontin expression and relation to disease severity in human asthma. Eur Respir J. 2011;37:331–41.

    Article  CAS  PubMed  Google Scholar 

  43. Gela A, Kasetty G, Morgelin M, Bergqvist A, Erjefalt JS, Pease JE, Egesten A. Osteopontin binds and modulates functions of eosinophil-recruiting chemokines. Allergy. 2016;71:58–67.

    Article  CAS  PubMed  Google Scholar 

  44. Kohan M, Breuer R, Berkman N. Osteopontin induces airway remodeling and lung fibroblast activation in a murine model of asthma. Am J Respir Cell Mol Biol. 2009;41:290–6.

    Article  CAS  PubMed  Google Scholar 

  45. Shao J, Xia ZW, Li YZ, Yu SC, Deng WW. [Expression of cellular phenotype switching markers-matrix protein gla, mRNA and collagen I, III and V of human airway smooth muscle cells in vitro after TGF-beta1 stimulation]. Zhonghua Er Ke Za Zhi. 2006;44:531–4.

    PubMed  Google Scholar 

  46. Ma L, Zhang R, Li D, Qiao T, Guo X. Fluoride regulates chondrocyte proliferation and autophagy via PI3K/AKT/mTOR signaling pathway. Chem Biol Interact. 2021;349:109659.

    Article  CAS  PubMed  Google Scholar 

  47. Song M, Wang X, Luo Y, Liu Z, Tan W, Ye P, Fu Z, Lu F, Xiang W, Tang L, et al. Cantharidin suppresses gastric cancer cell migration/invasion by inhibiting the PI3K/Akt signaling pathway via CCAT1. Chem Biol Interact. 2020;317:108939.

    Article  CAS  PubMed  Google Scholar 

  48. Li T, Zhang L, Jin C, Xiong Y, Cheng YY, Chen K. Pomegranate flower extract bidirectionally regulates the proliferation, differentiation and apoptosis of 3T3-L1 cells through regulation of PPARgamma expression mediated by PI3K-AKT signaling pathway. Biomed Pharmacother. 2020;131:110769.

    Article  CAS  PubMed  Google Scholar 

  49. Adhami VM, Syed DN, Khan N, Mukhtar H. Dietary flavonoid fisetin: a novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem Pharmacol. 2012;84:1277–81.

    Article  CAS  PubMed  Google Scholar 

  50. Chang PR, Liou JW, Chen PY, Gao WY, Wu CL, Wu MJ, Yen JH. The neuroprotective effects of Flavonoid Fisetin against Corticosterone-Induced Cell Death through Modulation of ERK, p38, and PI3K/Akt/FOXO3a-Dependent pathways in PC12 cells. Pharmaceutics. 2023;15:2376.

  51. Xu L, He X, Zhou Y, Yu K, Yuan M, Zhang Q, Qin L, Zhang Q. Connectivity map analysis identifies Fisetin as a treatment compound for osteoporosis through activating the PI3K-AKT signaling pathway in mouse pre-osteoblastic MC3T3-E1 cells. Curr Pharm Biotechnol. 2021;22:2038–47.

    Article  CAS  PubMed  Google Scholar 

  52. Ma B, Athari SS, Mehrabi Nasab E, Zhao L. PI3K/AKT/mTOR and TLR4/MyD88/NF-kappaB signaling inhibitors attenuate pathological mechanisms of allergic asthma. Inflammation. 2021;44:1895–907.

    Article  CAS  PubMed  Google Scholar 

  53. Wang J, Li F, Yang M, Wu J, Zhao J, Gong W, Liu W, Bi W, Dong L. FIZZ1 promotes airway remodeling through the PI3K/Akt signaling pathway in asthma. Exp Ther Med. 2014;7:1265–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work were funded by the Traditional Chinese Medicine Science and Technology Project of Shandong province (Grant No. M-2022217), Science and Technology Innovation Development Project of Taian City (Grant No. 2021NS423).

Author information

Authors and Affiliations

Authors

Contributions

LYY and YQL designed the study and wrote the article. LB conducted the network analysis. LZ performed the animal experiments. LMJ conducted the cell experiments. ML and HC organized the experimental scheme. CJ revised the manuscript. All authors have approved the submitted version.All authors have agreed both to be personally accountable for the author’s own contributions and to ensure that questions related to the accuracy or integrity of any part of the work, even ones in which the author was not personally involved, are appropriately investigated, resolved, and the resolution documented in the literature.

Corresponding authors

Correspondence to Ling Meng, Chao He or Jin Chang.

Ethics declarations

Ethics approval and consent to participate

All animal experiments were approved by the Ethics Committee of the Second Affiliated Hospital of Shandong First Medical University (approval no: 2023-A-061).

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

12931_2024_3005_MOESM1_ESM.rar

Supplementary Material 1: Additional file 1: Fig S1. The acquisition of genes related to ASMC, asthma and fisetin. (A) ASMC-related genes extracted from Genecards database. (B) Asthma-related genes extracted from Genecards database. (C) ASMC-related genes extracted from OMIM database. (D) Asthma-related genes extracted from OMIM database. (E) Fisetin-related genes extracted from Swiss Target Prediction database.

Supplementary Material 2: Additional file 2. Fig S2. All Western blotting images.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Yin, Q., Liu, B. et al. Fisetin reduces ovalbumin-triggered airway remodeling by preventing phenotypic switching of airway smooth muscle cells. Respir Res 25, 370 (2024). https://doi.org/10.1186/s12931-024-03005-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-03005-8

Keywords