Open Access

Anti‑VEGF treatment suppresses remodeling factors and restores epithelial barrier function through the E‑cadherin/β‑catenin signaling axis in experimental asthma models

  • Authors:
    • Ahmet Türkeli
    • Özge Yilmaz
    • Meral Karaman
    • Esra Toprak Kanik
    • Fatih Firinci
    • Sevinç İnan
    • Hasan Yüksel
  • View Affiliations

  • Published online on: April 29, 2021     https://doi.org/10.3892/etm.2021.10121
  • Article Number: 689
  • Copyright: © Türkeli 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

Besides maintaining a physical barrier with adherens junctional (AJ) and tight junctional proteins, airway epithelial cells have important roles in modulating the inflammatory processes of allergic asthma. E‑cadherin and β‑catenin are the key AJ proteins that are involved in airway remodeling. Various mediators such as transforming growth factor‑β (TGF‑β), epidermal growth factor (EGF), fibroblast growth factor (FGF), platelet derived growth factor (PDGF), insulin‑like growth factor (IGF), tumor necrosis factor‑α (TNF‑α) and angiogenic factors, such as vascular endothelial growth factor (VEGF), are released by the airway epithelium in allergic asthma. The signaling pathways activated by these growth factors trigger epithelial‑mesenchymal transition (EMT), which contributes to fibrosis and subsequent downregulation of E‑cadherin. The present study used a mouse asthma model to investigate the effects of anti‑VEGF, anti‑TNF and corticosteroid therapies on growth factor and E‑cadherin/β‑catenin expression. The study used 38 male BALB/c mice, divided into 5 groups. A chronic mouse asthma model was created by treating 4 of the groups with inhaled and intraperitoneal ovalbumin (n= 8 per group). Saline, anti‑TNF‑α (etanercept), anti‑VEGF (bevacizumab) or a corticosteroid (dexamethasone) were applied to each group by intraperitoneal injection. No medication was administered to the control group (n=6). Immunohistochemistry for E‑cadherin, β‑catenin and growth factors was performed on lung tissues and protein expression levels assessed using H‑scores. Statistically significant differences were observed in E‑cadherin, β‑catenin, EGF, FG, and PFGF (P<0.001 for all) as well as the IGF H‑scores between the five groups (P<0.005). Only anti‑VEGF treatment caused E‑cadherin and β‑catenin levels to increase to the level of non‑asthmatic control groups (P>0.005). All treatment groups had reduced TGF‑β, PDGF and FGF H‑scores in comparison with the untreated asthma group (P=0.001). The EGF and IGF levels were not significantly different between the untreated asthmatic and non‑asthmatic controls. The results suggested that anti‑VEGF and TNF‑α inhibition treatments are effective in decreasing growth factors, in a similar manner to conventional corticosteroid treatments. Anti‑VEGF and TNF inhibition therapy may be an effective treatment for remodeling in asthma while offering an alternative therapeutic option to steroid protective agents. The data suggested that anti‑VEGF treatment offered greater restoration of the epithelial barrier than both anti‑TNF‑α and corticosteroid treatment.

Introduction

A strong link between allergen exposure, sensitization and asthma development has been reported (1). Epithelial permeability is indicated to be markedly increased in allergic asthma, allowing allergens and irritants to penetrate into the subepithelial area, thus stimulating type 2 immune responses and constituting an important process in the immunopathology of allergy (2). Various mediators, such as transforming growth factor β (TGF-β), epidermal growth factor (EGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), insulin-like growth factor (IGF), tumor necrosis factor-α (TNF-α) and angiogenic factors are released by airway epithelium, inflammatory cells and structural cells (3-6). The signaling pathways activated by these growth factors trigger epithelial-mesenchymal transition (EMT), which contributes to fibrosis and subsequent downregulation of E-cadherin (6). Published studies suggest that EMT commonly occurs in epithelial cells in asthma and may contribute to airway remodeling (7,8). Remodeling comprises structural alterations to the airway wall, including epithelial cell loss, goblet cell hyperplasia, hypertrophy of the airway smooth muscle, basement membrane thickening and increased angiogenesis (9).

Protection of the integrity of the barrier function of the airway epithelium depends on adherens junctional (AJ) and tight junctional (TJ) proteins that link cells together (10,11). AJ proteins are part of two main complexes, the E-cadherin- β-catenin complex and the nectin-afadin complex, that have important roles in arranging cell-cell adhesion and intercellular motility (12). It has been reported that the expression of E-cadherin, β-catenin and zonula occludens-1 are reduced and that their functions are impaired in asthmatic epithelial cells in comparison with healthy cells (13-17).

Inhaled or oral corticosteroids are the most common drugs used for asthma therapy in the majority of patients with asthma (18). Despite providing clinical and symptomatic improvement, existing therapies are insufficient to prevent the onset of airway remodeling (9). The influence of various therapeutic agents on epithelial barrier dysfunction, inhibition of airway remodeling and controlling adaptive immunity guide the development of further therapeutic strategies, targets and rational treatment (19).

Vascular endothelial growth factor (VEGF) is a signaling protein and the main regulator of endothelial cell growth. It is released by many cells that stimulate the formation of blood vessels and is a classic profibrotic growth factor (20). The overexpression of VEGF in the lung can lead to significant airway remodeling (21). Therefore, antagonizing VEGF is important in the prevention of remodeling and in preserving epithelial barrier function (3,22).

An example of a proinflammatory cytokine that plays a role in the pathogenesis of asthma is TNF-α (4,23). TNF-α is usually secreted by macrophages, lymphocytes and mast cells in the lungs (24,25). However, lung epithelial cells may also release TNF-α (25,26). It has been reported that patients with severe asthma have a high amount of TNF-α in the airways in comparison to healthy individuals (24,25). Tissue damage occurs via a TNF-α driven effect in the airways and remodeling may develop as a result (24). Therefore, anti-TNF therapy may be useful to treat severe asthma. Co-prevention of remodeling and tissue injury may be possible by antagonism of TNF-α.

The present study aimed to use an experimental asthma mouse model, to investigate the effects of anti-VEGF, anti-TNF and corticosteroid therapies on growth factors and E-cadherin-β-catenin expression.

Materials and methods

Study animals

Male BALB/c mice (n=38; age, 6-8 weeks; weight, 18-20 g) were used to simulate chronic asthma. The mice were kept in a pathogen-free animal facility at a controlled temperature of 20-24˚C with 60±10% relative humidity and had free access to food and water on a 12-h light-dark cycle. The study was performed in accordance with the suggestions specified in the Guide for the Care and Use of Experimental Animals (27) and ethics approval was obtained from the Ethics Committee of Dokuz Eylül University.

Sensitization protocol

Mice were divided into five groups: i) Control group (n=6); ii) untreated asthma group (n=8); iii) TNF-α receptor blocker group (n=8); iv) anti-VEGF group (n=8); and v) corticosteroid group (n=8). Mice in the control group were not administered medication. The chronic asthma model was applied according to a previously described protocol in all groups except for the control group (28). The duration of the experiment was 10 weeks (2 weeks of ovalbumin injection and 8 weeks of nebulization). Mice were sensitized by injecting 10 µg (0.1 ml) ovalbumin (grade V; Sigma-Aldrich; Merck KGaA) intraperitoneally on the 1st and 14th day of the experiment. After the 3rd week of the experiment, mice were nebulized with 2.5% OVA aerosol in sterile saline for 30 min, 3 days per week for 8 weeks. Nebulized saline inhalation was applied to the mice in the control group. The mice were sacrificed after ketamine hydrochloride (50 mg/ml and 200 mg/kg) anesthesia was applied via the IP route on the first day after nebulized treatment was completed.

Study design

Chronic asthma developed in 32 mice. The treatments given to the study group were as follows: i) Control group, medication was not applied; ii) untreated asthma group, IP saline was administered once per week for 2 weeks; iii) TNF-α receptor blocker group, IP etanercept (Wyeth Europa Ltd.) was administered at a dose of 0.01 mg/dose (0.5 mg/kg) twice per week for two weeks; iv) anti-VEGF group, IP bevacizumab (Avastin®; Roche Diagnostics) was administered at a dose of 0.15 mg/dose 5 mg/kg once per week for 2 weeks and v) corticosteroid group, IP dexamethasone (Dekort®; Deva Holding A.S.) was administered at a dose of 1 mg/kg for 7 days. The treatments commenced at the same stage as the nebulization treatment, but in the case of the corticosteroid group only, the treatments commenced in the second week of nebulization treatment.

Histopathological and immunohistochemical evaluations

Murine lung tissue was fixed at room temperature with 10 % buffered formalin for three days, and a 5 mm thick horizontal slice was obtained from the middle zone of the left lung. Formalin-fixed lung tissue was embedded in paraffin for use in histopathological evaluation. Tissue blocks were serially cut into 5 µm sections. The sections were stained with hematoxylin and eosin (H&E) for histological evaluation (29). Immunostaining for E-cadherin (cat. no. sc-8426; mouse monoclonal IgG1; Santa Cruz Biotechnology, Inc.), β-catenin (cat. no. E-5: sc-7963; mouse monoclonal IgG1; Santa Cruz Biotechnology, Inc.), TGF-β1 (cat. no. bs-0086R; rabbit polyclonal IgG1; BIOSS), FGF-1 (cat. no. C-19: sc-1884; goat polyclonal IgG; Santa Cruz Biotechnology, Inc.), PDGF-A (cat. no. N-30: sc-128; rabbit polyclonal IgG; Santa Cruz Biotechnology, Inc.), EGF (cat. no. EGF-10: sc-57088; mouse monoclonal IgG1; Santa Cruz Biotechnology, Inc.) and IGF-1 (cat. no. H-70: sc-9013; rabbit polyclonal IgG; Santa Cruz Biotechnology, Inc.) was performed according to manufacturer's instructions (30). Sections were washed three times (5 min each) with PBS, followed by incubation at 4˚C for 1 h with biotinylated IgG and then with streptavidin-peroxidase conjugate. For anti-mouse and anti-rabbit primary antibodies, the Histostain Plus IHC secondary antibody system was used (cat. no. 85-9643; broad spectrum; Invitrogen; Thermo Fisher Scientific, Inc.). For anti-goat primary antibodies, ImmunoCruz goat ABC Staining System was used (cat. no. sc-2023; Santa Cruz Biotechnology, Inc.). The staining process was performed according to the manufacturer's instructions. After washing with PBS three times for 5 min, these sections were colored with 3,3'-diaminobenzidine for signal detection and then counterstained with Mayer's hematoxylin at room temperature for 2 min. All antibodies were used at 1:100 dilution. The sections were mounted using Entellan® mounting medium (HX265767; Merck KGaA) and evaluated using a light microscope (model BX40; Olympus Corporation). Control samples were processed similarly except for the use of IgG instead of a primary antibody (30).

The samples were evaluated by two experienced pathologists blinded to the clinical and serological characteristics of the rodents. Based on the nuclear and intracytoplasmic density of immunostaining of the airway epithelium, an H-SCORE [∑ Pi (I+1)] was calculated for all growth factors, E-cadherin and β-catenin. Both the ratio of positive immunostained cells to all cells in the selected fields and the respective H-scores were calculated. I is the intensity score (0, 1, 2 or 3 corresponding to the presence of negative, weak, intermediate or strong staining, respectively). Pi is the percentage of epithelial cells stained with each intensity between 0 and 100% (30). There were 25 slides per animal. From each slide 10 different areas (x400 magnification) were evaluated under a microscope and the percentage of cells with different staining densities was determined.

Statistical analysis

Statistical analysis was performed using SPSS 15.0 (SPSS, Inc.). The statistical significance between the 5 groups was assessed by Kruskal-Wallis test followed by the Dunn-Bonferroni post-hoc test. H-SCORES were compared between pairs of groups (two by two) using the Mann-Whitney U test. P<0.05 was accepted as statistically significant.

Results

TGF-β1 H-scores

TGF-β1 H-scores were compared between the five groups, and there was a statistically significant difference (P<0.001). The highest TGF-β1 H-scores were observed in the untreated asthma group and the lowest scores were observed in the TNF-α blocker group (202.5 vs. 151). There were no statistically significant differences among the TGF-β1 H-scores of the etanercept, bevacizumab and dexamethasone treatment groups and the non-asthmatic group (P>0.001, for all). The differences among the TGF-β1 H-scores of the etanercept, bevacizumab and dexamethasone treatment groups and the untreated asthma group were statistically significant (P<0.001 for all; Table I; Figs. 1 and 2).

Table I

Growth factors H-scores in the study groups.

Table I

Growth factors H-scores in the study groups.

 Study groups 
ProteinNon-asthmatic controlUntreated asthmaEtanerceptBevacizumabDexamethasone P-valuea
TGF-β202.5 (180-240)270 (254-280)b151 (130-172.5)c157.5 (150-187.5)c217.5 (177.5-225)c<0.001
PDGF150 (138-163)216.5 (207-226.5)b150 (135-169)c145 (133-169)c138 (130-152.5)c<0.001
FGF237 (225-259)351 (329-360)b263 (252-280)c217 (187-236)c266 (245-280)c<0.001
EGF232.5 (210-240)290 (260-310)232.5 (182.5-240)210 (140-225)d260 (210-300)0.005
IGF117.5 (90-130)97.5 (89-100)95 (90-100)80 (72.5-80)147 (130-165)<0.001

[i] aKruskal-Wallis statistical analysis (expressed as median-interquartile range). The Bonferroni correction was applied for the post-hoc analysis results, where P<0.005 was regarded as a statistically significant difference for the two group comparisons;

[ii] bMann-Whitney U-test, P<0.005 vs. non-asthmatic control;

[iii] cMann-Whitney U-test, P<0.001 vs. untreated asthma;

[iv] dMann-Whitney U-test, P=0.002 vs. untreated asthma.

PDGF-A H-scores

PDGF-A H-scores were compared between the five groups, and there was a statistically significant difference (P<0.001). The highest PDGF-A H-scores were observed in the untreated asthma group and the lowest scores were seen in the corticosteroid group (216.5 vs. 138). No statistically significant difference was found among the PDGF-A H-scores of the etanercept, bevacizumab and dexamethasone treatment groups in comparison with the non-asthmatic group (P>0.005, for all). The comparison of PDGF-A H-scores among the etanercept, bevacizumab, dexamethasone treatment groups and the untreated asthma group were statistically significantly (P<0.001 for all; Table I; Figs. 1 and 2).

FGF-1 H-scores

When FGF-1 H-scores were compared between the five groups, there was a statistically significant difference (P<0.001). The highest FGF-1 H-score was observed in the untreated asthma group and the lowest score in the anti VEGF group (351 vs. 217). When the FGF-1 H-scores of the etanercept, bevacizumab and dexamethasone treatment groups were compared with the non-asthmatic group no statistically significant difference was found (P>0.005, for all). The differences among the FGF-1 H-scores of the etanercept, bevacizumab, dexamethasone treatment groups and the untreated asthma group were statistically significant (P<0.001 for all; Table I; Figs. 1 and 2).

EGF H-scores

The EGF H-scores between the five groups were significant (P=0.001). The highest EGF-H-score was observed in the untreated asthma group and the lowest score in the anti-VEGF group (290 vs. 210; P=0.002). The comparison of EGF H-scores between the bevacizumab treatment group and the untreated asthma group indicated a statistically significant difference (P=0.002). When the EGF H-scores of the etanercept and dexamethasone treatment groups were compared with the non-asthmatic group, no statistically significant difference was observed (P>0.005; Table I; Figs. 1 and 2).

IGF H-scores

There was a statistically significant difference when the IGF H-scores were compared between the five groups (P<0.001). The highest IGF H-score was found in the corticosteroid group, and the lowest score was in the anti-VEGF group (147 vs. 80; P=0.001). The IGF H-scores of untreated asthma, etanercept, bevacizumab and dexamethasone treatment groups were not significantly different from those in healthy controls (P>0.005, for all). The differences between the IGF H-score in the corticosteroid group in comparison to the untreated asthma, etanercept and anti-VEGF groups was statistically significant (P=0.001 for all; Table I; Fig. 1).

E-cadherin H-scores

The H-scores of E-cadherin between the five groups were significantly different (P<0.001). The highest E-cadherin H-score was observed in the non-asthmatic control group and the lowest score was in the untreated asthma group (225 vs. 125; P=0.002). The E-cadherin H-scores of the anti-VEGF group were closest to the non-asthmatic control groups (193.5 vs. 225; P=0.023; Table II). The H-scores of the TNF-α receptor blocker and corticosteroid treatment groups were significantly lower than the control group (P=0.003 and P=0.002, respectively). Although the H-scores of the TNF-α receptor blocker and corticosteroid treatment groups were higher than the untreated asthma group, the differences were not significant (158.5 vs. 125 and 138 vs. 125; P=0.013 and P=0.138, respectively; Table II; Figs. 1 and 3).

Table II

E-cadherin and β-catenin H-scores in the study groups.

Table II

E-cadherin and β-catenin H-scores in the study groups.

 Study groups 
ProteinNon-asthmatic controlUntreated asthmaEtanerceptBevacizumabDexamethasone P-valuea
E-cadherin225 (200-234)b125 (118-40)158.5 (138-172.5)c193.5 (181-195)d138 (129-161)c<0.001
β-catenin191.5 (188-200)b120 (115-127.5)138 (125-154)c175 (150-182.5)d135 (130-140)c<0.001

[i] aKruskal-Wallis statistical analysis (expressed as median-interquartile range). The Bonferroni correction was applied for the post-hoc analysis results, where P<0.005 was regarded as a statistically significant difference for the two group comparisons;

[ii] bMann-Whitney U-test, P=0.002 vs. untreated asthma;

[iii] cMann-Whitney U-test, P<0.005 vs. Non-asthmatic control;

[iv] dMann-Whitney U-test, P>0.05 vs. Non-asthmatic control.

β-catenin H-scores

There was a statistically significant difference in β-catenin H-scores (P<0.001) between the five groups. The highest β-catenin H-score was found in the non-asthmatic control group and lowest score was in the untreated asthma group (191.5 vs. 120; P=0.002). Among the etanercept, bevacizumab and dexamethasone treatment groups, the β-catenin H-scores of the bevacizumab group were closest to the non-asthmatic control groups (175 vs. 191.5; P=0.002). The TNF-α receptor blocker and corticosteroid treatment groups had lower β-catenin H-scores than the control group (P=0.002 and P=0.002, respectively). Although the β-catenin H-scores of the etanercept and the corticosteroid treatment groups were higher than those in the untreated asthma group, the differences were not significant (138 vs. 120 and 135 vs. 120; P=0.056 and P=0.023, respectively; Table II; Figs. 1 and 3).

Discussion

The results suggested that the levels of TGF-β, PDGF and FGF were increased in the untreated asthma group compared with non-asthmatic controls. All treatment groups had reduced TGF-β, PDGF and FGF H-scores in comparison with the untreated asthma group. It was determined that E-cadherin and β-catenin levels were reduced in the untreated asthma group compared to the non-asthmatic control and that their levels increased in non-asthmatic controls following treatment with anti-VEGF treatment.

In asthma, growth factors can trigger inflammation and structural changes in the airways (e.g., epithelial barrier dysfunction and remodeling). This is a process in which EMT causes epithelial cells to dedifferentiate into more mobile mesenchymal cells, such as fibroblasts (6,31,32). EMT can be stimulated by various signals and molecules, including tyrosine kinase receptors, including FGF, IGF, EGF, PDGF and VEGF, and the TGF-β and Wnt/β-catenin pathways (5,6). The studies conducted to date have indicated that the asthmatic airway epithelium is an important source of profibrogenic growth factors, including FGF, IGF, EGF, TGF-β, PDGF and VEGF (6,33-35). Similar to these studies, the present study determined that TGF-β, PDGF and FGF levels were higher in asthmatic mice than control mice; however, IGF and EGF levels were not different from the control group in the present study.

The airway epithelium creates a structural and immunological barrier against environmental stimuli, such as aeroallergens, microorganisms and particulate matter (36). Changes in the asthmatic epithelium include epithelial shedding and the destruction of ciliary cells (37). E-cadherin is important in maintenance of the cytoskeleton and TJ functional integrity (38). Reduced E-cadherin activity causes TJ barrier integrity to deteriorate (38,39). In addition, E-cadherin plays a role in the repression of intracellular signaling pathways, the arrangement of the epithelium and cell proliferation and differentiation (37).

β-catenin is a membrane-bound protein and forms a key AJ component, interacting with E-cadherins and connecting them to the cytoskeleton. Disruption of this epithelial barrier function allows easier access of inhalant allergens to the antigen-presenting cells in the submucosa, causing stimulation of the Th2 type mediated immune response and EMT (13,14,38). A gradual loss of E-cadherin is considered a hallmark of EMT (5,6). Membrane levels of E-cadherin-β- catenin have been found to be reduced in the asthmatic epithelial cell in comparison with healthy cells (13-17). When the E-cadherin-β-catenin structure is disrupted, β-catenin is released into the cytosol and translocates to the nucleus, where it activates the Wnt/β-catenin signaling pathway and contributes to the EMT; this process is involved in tissue remodeling (7,14,40-42).

Studies on mice have shown that repetitive antigen load causes extensive E-cadherin loss and that there is a correlation between the number of allergen exposures and E-cadherin levels (39,43). In biopsies of the bronchial mucosa of patients with atopic asthma, E-cadherin levels in the respiratory epithelia were shown to be lower than non-atopic and non-asthmatic cases (17,44,45). Similarly, a study published by our group showed that patients with asthma had lower levels of E-cadherin in their airways (46). Similar to previous studies, the present study suggested that E-cadherin and β-catenin levels were lower in asthmatic model mice.

VEGF has significant roles in inflammation, angiogenesis and the vascular permeability of airways and in subepithelial collagen deposition, airway smooth muscle hyperplasia and the development of physiological abnormalities of the airway (33). Bevacizumab is a humanized monoclonal antibody that inhibits the activation of VEGF-R and tumor growth (20). TGF-β, IGF, FGF and PDGF were shown to stimulate the secretion of VEGF (33,47). Similar to these studies, the correlation of increased TGF-β levels and the decrease in E-cadherin and β-catenin levels was shown in the present study as well (11,14,32,40,45,48-50). TGF-β has also caused a significant increase in transcriptional activation of β-catenin (14,40). The available knowledge suggests that VEGF potentially affects the signal pathway, and thus triggers myofibroblast transformation (51). In the present study of asthmatic model mice, TGF-β, PDGF and FGF levels decreased with treatment to show no difference from non-asthmatic controls. These results suggest that EMT can be controlled by treatment approaches via VEGF.

Although the role of VEGF in the pathogenesis in asthma and remodeling is known, the effect of antagonizing VEGF on epithelial barrier integrity and AJ structure is not fully understood (33). Most knowledge on the effects of VEGF on epithelial barrier integrity was obtained from studies of the retinal endothelium (30,52). VEGF exposure causes decreased transepithelial resistance and increased vascular permeability. Permeability-enhancing agents also change AJ (53). After exposure to VEGF, the expression of VE-cadherin, β-catenin, occludin, claudin and ZO-1 proteins in the endothelium decreased in vitro (54-57). Furthermore, in diabetic retinopathy, VEGF-R1 antagonism has been shown to prevent vascular leakage and retinal leukostasis, degeneration and disorganization of ZO-1 and VE-cadherin (55,57). Similarly, in the present study, E-cadherin and β-catenin levels of asthmatic mice treated with anti-VEGF increased to levels of control mice without asthma. Overall, in the experimental asthma mouse model, anti-VEGF therapy appeared to cause an elevation in epithelial barrier proteins, E-cadherin and β-catenin levels. Therefore, the use of anti-VEGF therapy in asthma may be a therapeutic option for the restoration of epithelial barrier.

TNF-α is released from the airways in asthma and may play a role in the pathogenesis of allergic inflammation through the activation of transcription factors (4). TNF-α was shown to upregulate TGF-β expression in mice lung fibroblasts (58). TNF-α stimulates TGF-β expression in severe asthma, causing fibroblast growth and maturation into myofibroblasts (59). TNF-α enhances the effect of TGF-β1 on EMT induction in human bronchial epithelial cells (HBECs) (60). The effectiveness of a TNF-α receptor antagonist in severe asthma therapy has recently been studied (25). In the present study of asthmatic mice models, etanercept was shown to downregulate the expression of PDGF and FGF as well as TGF-β. Despite the apparent efficacy in treatment, the mechanism of action of anti-TNF-α is not fully understood, which is limitation of the present study. Further cellular and molecular mechanistic studies are needed to determine how these changes occur in the future. TNF-α causes epithelial barrier dysfunction in other tissues, and previous studies have shown that TNF-α in combination with IFN-γ disrupted the TJ in HBECs (61-63). Therefore, the inhibition of TNF-α can be considered as an option in maintaining epithelial integrity.

In another study, TNF-α stimulation in bronchial epithelial cell culture has been shown to reduce E-cadherin and β-catenin expression; however, improvements were observed with TNF-α inhibition and corticosteroid therapy (48). In contrast to these studies, E-cadherin and β-catenin expression partially increased with corticosteroid treatment and TNF-α inhibition in the present study, but did reach the levels of healthy controls. This may have been caused by the difference between inflammatory processes of Th2 type inflammation caused by the OVA-induced asthma model and Th1 type inflammation caused by TNF-α.

In experimental asthma model studies, corticosteroids were shown to decrease the release of TGF-β while dexamethasone was shown to decline the release of TGF-β, IGF, and FGF. ICS was shown to inhibit IGF expression (23,64,65). All glucocorticosteroids have been shown to inhibit PDGF and collagen I-induced proliferation and hypocontractility in airway smooth muscle cells (66). The anti-angiogenic effects of dexamethasone on VEGF release in FGF-treated human airway smooth muscle cells via the p38MAPK pathway were also demonstrated in vitro (47). Similar to these studies, the present study suggested that dexamethasone decreased TGF-β, PDGF and FGF levels.

By inducing tight junction formation and increasing transepithelial resistance, corticosteroids play a crucial role in the function and maintenance of cell-cell contact. However, the potential effects of returning to the original state of AJs (E-cadherin, β-catenin) and healing the epithelial barrier are not fully clear (18,48,67). Glucocorticoid administration in Doerner and Zuraw (68) did not substantially change E-cadherin mRNA downregulation mediated by TGF-β1. Song et al (67) showed that E-cadherin distribution could be partially salvaged following pretreatment with dexamethasone in experimental asthma models. While the downregulation of hypoxia-induced loss of ZO-1 expression was inhibited by dexamethasone in the human corneal epithelia, it did not affect the E-cadherin vs. β-catenin (69). In the present study, E-cadherin and β-catenin expression did not reach the level of healthy controls with dexamethasone treatment similar to the aforementioned studies.

The limitations of our study are the inability to look at the molecular level of β-catenin in terms of quantity, only the adherens proteins, and the inability to show a barrier function such as transepithelial resistance as physical data. Further cellular and molecular mechanistic studies are needed to determine how these changes occur in the future.

In the present study, the effects of conventional corticosteroid treatment, VEGF inhibition treatments and TNF-α antagonistic treatment on growth factor and E-cadherin-β-catenin expression were compared. These proteins play roles in the pathogenesis of asthma (3-6). Growth factors TGF-β, PDGF and FGF expression levels were high in asthmatic mice models and E-cadherin and β-catenin expressions were low. Anti-VEGF and TNF-α inhibition treatments are effective in decreasing growth factors similar to conventional corticosteroid treatments. However, corticosteroid and TNF-α inhibition treatment were not effective in increasing E-cadherin and β-catenin levels. This leads to epithelial barrier function disturbance. Anti-VEGF treatment increased the expression of AJ proteins in bronchial epithelium, and VEGF and TNF inhibition may provide an alternative therapeutic option to steroid-sparing agents. They are also a more effective treatment of epithelial barrier restoration and remodeling in asthma. However, before making firm recommendations, further clinical studies are needed to investigate the in vivo efficacy and safety profile of anti-VEGF treatment in asthma.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HY and OY were involved in the conception, hypotheses delineation and design of the study. AT wrote and HY revised the manuscript. FF, AT and ETK took part in the acquisition of the data. SI and MK participated in the analysis and interpretation of the data. HY and OY confirmed the authenticity of the raw data. All authors confirmed that they have read and approved the final manuscript.

Ethics approval and consent to participate

Ethics approval was obtained from the Ethics Committee of Dokuz Eylül University and the Animal Experiment Unit for the implementation of all experimental procedures.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Leynaert B, Le Moual N, Neukirch C, Siroux V and Varraso R: Environmental risk factors for asthma developement. Presse Med. 48:262–273. 2019.PubMed/NCBI View Article : Google Scholar : (In French).

2 

Goleva E, Berdyshev E and Leung DY: Epithelial barrier repair and prevention of allergy. J Clin Invest. 129:1463–1474. 2019.PubMed/NCBI View Article : Google Scholar

3 

Lee HY, Hur J, Kim IK, Kang JY, Yoon HK, Lee SY, Kwon SS, Kim YK and Rhee CK: Effect of nintedanib on airway inflammation and remodeling in a murine chronic asthma model. Exp Lung Res. 43:187–196. 2017.PubMed/NCBI View Article : Google Scholar

4 

Kim J and Remick DG: Tumor necrosis factor inhibitors for the treatment of asthma. Curr Allergy Asthma Rep. 7:151–156. 2007.PubMed/NCBI View Article : Google Scholar

5 

Guntur VP and Reinero CR: The potential use of tyrosine kinase inhibitors in severe asthma. Curr Opin Allergy Clin Immunol. 12:68–75. 2012.PubMed/NCBI View Article : Google Scholar

6 

Hasan NAHM, Harith HH, Israf DA and Tham CL: The differential effects of commercial specialized media on cell growth and transforming growth factor beta 1-induced epithelial-mesenchymal transition in bronchial epithelial cells. Mol Biol Rep. 47:3511–3519. 2020.PubMed/NCBI View Article : Google Scholar

7 

Yuksel H and Türkeli A: Airway epithelial barrier dysfunction in the pathogenesis and prognosis of respiratory tract diseases in childhood and adulthood. Tissue Barriers. 5(e1367458)2017.PubMed/NCBI View Article : Google Scholar

8 

Chu S, Zhang X, Sun Y, Liang Y, Sun J, Lu M, Huang J, Jiang M and Ma L: Atrial natriuretic peptide inhibits epithelial-mesenchymal transition (EMT) of bronchial epithelial cells through cGMP/PKG signaling by targeting Smad3 in a murine model of allergic asthma. Exp Lung Res. 45:245–254. 2019.PubMed/NCBI View Article : Google Scholar

9 

Zhang J and Dong L: Status and prospects: Personalized treatment and biomarker for airway remodeling in asthma. J Thorac Dis. 12:6090–6101. 2020.PubMed/NCBI View Article : Google Scholar

10 

Hellings PW and Steelant B: Epithelial barriers in allergy and asthma. J Allergy Clin Immunol. 145:1499–1509. 2020.PubMed/NCBI View Article : Google Scholar

11 

Post S, Nawijn MC, Jonker MR, Kliphuis N, van den Berge M, van Oosterhout AJ and Heijink IH: House dust mite-induced calcium signaling instigates epithelial barrier dysfunction and CCL20 production. Allergy. 68:1117–1125. 2013.PubMed/NCBI View Article : Google Scholar

12 

Niessen CM: Tight junctions/adherens junctions: Basic structure and function. J Invest Dermatol. 127:2525–2532. 2007.PubMed/NCBI View Article : Google Scholar

13 

Knight DA, Stick SM and Hackett TL: Defective function at the epithelial junction: A novel therapeutic frontier in asthma? J Allergy Clin Immunol. 128:557–558. 2011.PubMed/NCBI View Article : Google Scholar

14 

Heijink IH, Postma DS, Noordhoek JA, Broekema M and Kapus A: House dust mite-promoted epithelial-to-mesenchymal transition in human bronchial epithelium. Am J Respir Cell Mol Biol. 42:69–79. 2010.PubMed/NCBI View Article : Google Scholar

15 

Cao N, Wang J, Xu X, Xiang M and Dou J: PACAP38 improves airway epithelial barrier destruction induced by house dust mites allergen. Immunobiology. 224:758–764. 2019.PubMed/NCBI View Article : Google Scholar

16 

Yao L, Chen S, Tang H, Huang P, Wei S, Liang Z, Chen X, Yang H, Tao A, Chen R, et al: Transient receptor potential ion channels mediate adherens junctions dysfunction in a toluene diisocyanate-induced murine asthma model. Toxicol Sci. 168:160–170. 2019.PubMed/NCBI View Article : Google Scholar : Erratum in Toxicol Sci 170, 247, 2019.

17 

de Boer WI, Sharma HS, Baelemans SM, Hoogsteden HC, Lambrecht BN and Braunstahl GJ: Altered expression of epithelial junctional proteins in atopic asthma: Possible role in inflammation. Can J Physiol Pharmacol. 86:105–112. 2008.PubMed/NCBI View Article : Google Scholar

18 

Sekiyama A, Gon Y, Terakado M, Takeshita I, Kozu Y, Maruoka S, Matsumoto K and Hashimoto S: Glucocorticoids enhance airway epithelial barrier integrity. Int Immunopharmacol. 12:350–357. 2012.PubMed/NCBI View Article : Google Scholar

19 

McLellan K, Shields M, Power U and Turner S: Primary airway epithelial cell culture and asthma in children-lessons learnt and yet to come. Pediatr Pulmonol. 50:1393–1405. 2015.PubMed/NCBI View Article : Google Scholar

20 

Takahashi S: Vascular endothelial growth factor (VEGF), VEGF receptors and their inhibitors for antiangiogenic tumor therapy. Biol Pharm Bull. 34(1785e1798)2011.PubMed/NCBI View Article : Google Scholar

21 

Hur GY and Broide DH: Genes and pathways regulating decline in lung function and airway remodeling in asthma. Allergy Asthma Immunol Res. 11:604–621. 2019.PubMed/NCBI View Article : Google Scholar

22 

Huang C, Dong H, Zou M, Luo L, Hu Y, Xie Z, Le Y, Liu L, Zou F and Cai S: Bevacizumab reduced auto-phosphorylation of VEGFR2 to protect HDM-induced asthma mice. Biochem Biophys Res Commun. 478:181–186. 2016.PubMed/NCBI View Article : Google Scholar

23 

Herbert C, Hettiaratchi A, Webb DC, Thomas PS, Foster PS and Kumar RK: Suppression of cytokine expression by roflumilast and dexamethasone in a model of chronic asthma. Clin Exp Allergy. 38:847–856. 2008.PubMed/NCBI View Article : Google Scholar

24 

Ghebre MA, Pang PH, Desai D, Hargadon B, Newby C, Woods J, Rapley L, Cohen SE, Herath A, Gaillard EA, et al: Severe exacerbations in moderate-to-severe asthmatics are associated with increased pro-inflammatory and type 1 mediators in sputum and serum. BMC Pulm Med. 19(144)2019.PubMed/NCBI View Article : Google Scholar

25 

Malaviya R, Laskin JD and Laskin DL: Anti-TNFα therapy in inflammatory lung diseases. Pharmacol Ther. 180:90–98. 2017.PubMed/NCBI View Article : Google Scholar

26 

Mukhopadhyay S, Hoidal JR and Mukherjee TK: Role of TNFalpha in pulmonary pathophysiology. Respir Res. 7(125)2006.PubMed/NCBI View Article : Google Scholar

27 

National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animal: Guide for the Care and Use of Laboratory Animals, 8th edition. National Academies Press (US), Washington, DC, 2011.

28 

Temelkovski J, Hogan SP, Shepherd DP, Foster PS and Kumar RK: An improved murine model of asthma: Selective airway inflammation, epithelial lesions and increased methacholine responsiveness following chronic exposure to aerosolised allergen. Thorax. 53:849–856. 1998.PubMed/NCBI View Article : Google Scholar

29 

Cardiff RD, Miller CH and Munn RJ: Manual hematoxylin and eosin staining of mouse tissue sections. Cold Spring Harb Protoc. 2014:655–658. 2014.PubMed/NCBI View Article : Google Scholar

30 

Yuksel H, Yilmaz O, Baytur YB and Ozbilgin K: Prenatal administration of granulocyte-macrophage colony-stimulating factor increases mesenchymal vascular endothelial growth factor expression and maturation in fetal rat lung. Exp Lung Res. 34:550–558. 2008.PubMed/NCBI View Article : Google Scholar

31 

Wang T, Zhou Q and Shang Y: MiRNA-451a inhibits airway remodeling by targeting Cadherin 11 in an allergic asthma model of neonatal mice. Int Immunopharmacol. 83(106440)2020.PubMed/NCBI View Article : Google Scholar

32 

Gong JH, Cho IH, Shin D, Han SY, Park SH and Kang YH: Inhibition of airway epithelial-to-mesenchymal transition and fibrosis by kaempferol in endotoxin-induced epithelial cells and ovalbumin-sensitized mice. Lab Invest. 94:297–308. 2014.PubMed/NCBI View Article : Google Scholar

33 

Makinde T, Murphy RF and Agrawal DK: Immunomodulatory role of vascular endothelial growth factor and angiopoietin-1 in airway remodeling. Curr Mol Med. 6:831–841. 2006.PubMed/NCBI View Article : Google Scholar

34 

Hough KP, Curtiss ML, Blain TJ, Liu RM, Trevor J, Deshane JS and Thannickal VJ: Airway remodeling in asthma. Front Med (Lausanne). 7(191)2020.PubMed/NCBI View Article : Google Scholar

35 

Hasan NAHM, Harith HH, Israf DA and Tham CL: The differential effects of commercial specialized media on cell growth and transforming growth factor beta 1-induced epithelial-mesenchymal transition in bronchial epithelial cells. Mol Biol Rep. 47:3511–3519. 2020.PubMed/NCBI View Article : Google Scholar

36 

Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM and Wegmann M: More than just a barrier: The immune functions of the airway epithelium in asthma pathogenesis. Front Immunol. 11(761)2020.PubMed/NCBI View Article : Google Scholar

37 

Post S, Heijink IH, Hesse L, Koo HK, Shaheen F, Fouadi M, Kuchibhotla VNS, Lambrecht BN, Van Oosterhout AJM, Hackett TL, et al: Characterization of a lung epithelium specific E-cadherin knock-out model: Implications for obstructive lung pathology. Sci Rep. 8(13275)2018.PubMed/NCBI View Article : Google Scholar

38 

Cai J, Culley MK, Zhao Y and Zhao J: The role of ubiquitination and deubiquitination in the regulation of cell junctions. Protein Cell. 9:754–769. 2018.PubMed/NCBI View Article : Google Scholar

39 

Goto Y, Uchida Y, Nomura A, Sakamoto T, Ishii Y, Morishima Y, Masuyama K and Sekizawa K: Dislocation of E-cadherin in the airway epithelium during an antigen-induced asthmatic response. Am J Respir Cell Mol Biol. 23:712–718. 2000.PubMed/NCBI View Article : Google Scholar

40 

Baarsma HA, Menzen MH, Halayko AJ, Meurs H, Kerstjens HA and Gosens R: β-catenin signaling is required for TGF-β1-induced extracellular matrix production by airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 301:L956–L965. 2011.PubMed/NCBI View Article : Google Scholar

41 

Kim HT, Yin W, Nakamichi Y, Panza P, Grohmann B, Buettner C, Guenther S, Ruppert C, Kobayashi Y, Guenther A, et al: WNT/RYK signaling restricts goblet cell differentiation during lung development and repair. Proc Natl Acad Sci USA. 116:25697–25706. 2019.PubMed/NCBI View Article : Google Scholar

42 

Jia XX, Zhu TT, Huang Y, Zeng XX, Zhang H and Zhang WX: Wnt/β-catenin signaling pathway regulates asthma airway remodeling by influencing the expression of c-Myc and cyclin D1 via the p38 MAPK-dependent pathway. Exp Ther Med. 18:3431–3438. 2019.PubMed/NCBI View Article : Google Scholar

43 

Evans SM, Blyth DI, Wong T, Sanjar S and West MR: Decreased distribution of lung epithelial junction proteins after intratracheal antigen or lipopolysaccharide challenge: Correlation with neutrophil influx and levels of BALF sE-cadherin. Am J Respir Cell Mol Biol. 27:446–454. 2002.PubMed/NCBI View Article : Google Scholar

44 

Xiao C, Puddicombe SM, Field S, Haywood J, Broughton-Head V, Puxeddu I, Haitchi HM, Vernon-Wilson E, Sammut D, Bedke N, et al: Defective epithelial barrier function in asthma. J Allergy Clin Immunol. 128:549–56.e1-12. 2011.PubMed/NCBI View Article : Google Scholar

45 

Trautmann A, Kruger K, Akdis M, Muller-Wening D, Akkaya A, Brocker EB, Blaser K and Akdis CA: Apoptosis and loss of adhesion of bronchial epithelial cells in asthma. Int Arch Allergy Immunol. 138:142–150. 2005.PubMed/NCBI View Article : Google Scholar

46 

Yuksel H, Türkeli A, Taneli F, Horasan GD, Kanik ET, Kizilkaya M, Gözükara C and Yilmaz O: E-cadherin as an epithelial barrier protein in exhaled breath condensate. J Breath Res. 8(046006)2014.PubMed/NCBI View Article : Google Scholar

47 

Willems-Widyastuti A, Vanaudenaerde BM, Vos R, Dilisen E, Verleden SE, De Vleeschauwer SI, Vaneylen A, Mooi WJ, de Boer WI, Sharma HS, et al: Azithromycin attenuates fibroblast growth factors induced vascular endothelial growth factor via p38(MAPK) signaling in human airway smooth muscle cells. Cell Biochem Biophys. 67:331–339. 2013.PubMed/NCBI View Article : Google Scholar

48 

Carayol N, Campbell A, Vachier I, Mainprice B, Bousquet J, Godard P and Chanez P: Modulation of cadherin and catenins expression by tumor necrosis factor-alpha and dexamethasone in human bronchial epithelial cells. Am J Respir Cell Mol Biol. 26:341–347. 2002.PubMed/NCBI View Article : Google Scholar

49 

Moheimani F, Roth HM, Cross J, Reid AT, Shaheen F, Warner SM, Hirota JA, Kicic A, Hallstrand TS, Kahn M, et al: Disruption of β-catenin/CBP signaling inhibits human airway epithelial-mesenchymal transition and repair. Int J Biochem Cell Biol. 68:59–69. 2015.PubMed/NCBI View Article : Google Scholar

50 

Winton HL, Wan H, Cannell MB, Thompson PJ, Garrod DR, Stewart GA and Robinson C: Class specific inhibition of house dust mite proteinases which cleave cell adhesion, induce cell death and which increase the permeability of lung epithelium. Br J Pharmacol. 124:1048–1059. 1998.PubMed/NCBI View Article : Google Scholar

51 

Park HY, Kim JH and Park CK: VEGF induces TGF-β1 expression and myofibroblast transformation after glaucoma surgery. Am J Pathol. 182:2147–2154. 2013.PubMed/NCBI View Article : Google Scholar

52 

Chatterjee S, Wang Y, Duncan MK and Naik UP: Junctional adhesion molecule-A regulates vascular endothelial growth factor receptor-2 signaling-dependent mouse corneal wound healing. PLoS One. 8(e63674)2013.PubMed/NCBI View Article : Google Scholar

53 

Wallez Y and Huber P: Endothelial adherens and tight junctions in vascular homeostasis, inflammation and angiogenesis. Biochim Biophys Acta. 1778:794–809. 2008.PubMed/NCBI View Article : Google Scholar

54 

Díaz-Coránguez M, Lin CM, Liebner S and Antonetti DA: Norrin restores blood-retinal barrier properties after vascular endothelial growth factor-induced permeability. J Biol Chem. 295:4647–4660. 2020.PubMed/NCBI View Article : Google Scholar

55 

He J, Wang H, Liu Y, Li W, Kim D and Huang H: Blockade of vascular endothelial growth factor receptor 1 prevents inflammation and vascular leakage in diabetic retinopathy. J Ophthalmol. 2015(605946)2015.PubMed/NCBI View Article : Google Scholar

56 

Wisniewska-Kruk J, Hoeben KA, Vogels IM, Gaillard PJ, Van Noorden CJ, Schlingemann RO and Klaassen I: A novel co-culture model of the blood-retinal barrier based on primary retinal endothelial cells, pericytes and astrocytes. Exp Eye Res. 96:181–190. 2012.PubMed/NCBI View Article : Google Scholar : Erratum in Exp Eye Res 138, 167, 2015.

57 

Harhaj NS, Felinski EA, Wolpert EB, Sundstrom JM, Gardner TW and Antonetti DA: VEGF activation of protein kinase C stimulates occludin phosphorylation and contributes to endothelial permeability. Invest Ophthalmol Vis Sci. 47:5106–5115. 2006.PubMed/NCBI View Article : Google Scholar

58 

Sullivan DE, Ferris M, Nguyen H, Abboud E and Brody AR: TNF-alpha induces TGF-beta1 expression in lung fibroblasts at the transcriptional level via AP-1 activation. J Cell Mol Med. 13:1866–1876. 2009.PubMed/NCBI View Article : Google Scholar

59 

Brightling C, Berry M and Amrani Y: Targeting TNF-alpha: A novel therapeutic approach for asthma. J Allergy Clin Immunol. 121:5–10; quiz 11-12. 2008.PubMed/NCBI View Article : Google Scholar

60 

Câmara J and Jarai G: . Epithelial-mesenchymal transition in primary human bronchial epithelial cells is Smad-dependent and enhanced by fibronectin and TNF-alpha. Fibrogenesis Tissue Repair. 3(2)2010.PubMed/NCBI View Article : Google Scholar

61 

Hardyman MA, Wilkinson E, Martin E, Jayasekera NP, Blume C, Swindle EJ, Gozzard N, Holgate ST, Howarth PH, Davies DE, et al: TNF-α-mediated bronchial barrier disruption and regulation by src-family kinase activation. J Allergy Clin Immunol. 132:665–675.e8. 2013.PubMed/NCBI View Article : Google Scholar

62 

Coyne CB, Vanhook MK, Gambling TM, Carson JL, Boucher RC and Johnson LG: Regulation of airway tight junctions by proinflammatory cytokines. Mol Biol Cell. 13:3218–3234. 2002.PubMed/NCBI View Article : Google Scholar

63 

Pohl C, Hermanns MI, Uboldi C, Bock M, Fuchs S, Dei-Anang J, Mayer E, Kehe K, Kummer W and Kirkpatrick CJ: Barrier functions and paracellular integrity in human cell culture models of the proximal respiratory unit. Eur J Pharm Biopharm. 72:339–349. 2009.PubMed/NCBI View Article : Google Scholar

64 

Doherty T and Broide D: Cytokines and growth factors in airway remodeling in asthma. Curr Opin Immunol. 19:676–680. 2007.PubMed/NCBI View Article : Google Scholar

65 

Wang Z, Li W, Guo Q, Wang Y, Ma L and Zhang X: Insulin-like growth factor-1 signaling in lung development and inflammatory lung diseases. BioMed Res Int. 2018(6057589)2018.PubMed/NCBI View Article : Google Scholar

66 

Dekkers BG, Pehlic A, Mariani R, Bos IS, Meurs H and Zaagsma J: Glucocorticosteroids and β2-adrenoceptor agonists synergize to inhibit airway smooth muscle remodeling. J Pharmacol Exp Ther. 342:780–787. 2012.PubMed/NCBI View Article : Google Scholar

67 

Song J, Zhao H, Dong H, Zhang D, Zou M, Tang H, Liu L, Liang Z, Lv Y, Zou F, et al: Mechanism of E-cadherin redistribution in bronchial airway epithelial cells in a TDI-induced asthma model. Toxicol Lett. 220:8–14. 2013.PubMed/NCBI View Article : Google Scholar

68 

Doerner AM and Zuraw BL: TGF-β1 induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells is enhanced by IL-1β but not abrogated by corticosteroids. Respir Res. 10(100)2009.PubMed/NCBI View Article : Google Scholar

69 

Kimura K, Teranishi S, Kawamoto K and Nishida T: Protective effect of dexamethasone against hypoxia-induced disruption of barrier function in human corneal epithelial cells. Exp Eye Res. 92:388–393. 2011.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2021
Volume 22 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Türkeli A, Yilmaz Ö, Karaman M, Kanik ET, Firinci F, İnan S and Yüksel H: Anti‑VEGF treatment suppresses remodeling factors and restores epithelial barrier function through the E‑cadherin/β‑catenin signaling axis in experimental asthma models. Exp Ther Med 22: 689, 2021
APA
Türkeli, A., Yilmaz, Ö., Karaman, M., Kanik, E.T., Firinci, F., İnan, S., & Yüksel, H. (2021). Anti‑VEGF treatment suppresses remodeling factors and restores epithelial barrier function through the E‑cadherin/β‑catenin signaling axis in experimental asthma models. Experimental and Therapeutic Medicine, 22, 689. https://doi.org/10.3892/etm.2021.10121
MLA
Türkeli, A., Yilmaz, Ö., Karaman, M., Kanik, E. T., Firinci, F., İnan, S., Yüksel, H."Anti‑VEGF treatment suppresses remodeling factors and restores epithelial barrier function through the E‑cadherin/β‑catenin signaling axis in experimental asthma models". Experimental and Therapeutic Medicine 22.1 (2021): 689.
Chicago
Türkeli, A., Yilmaz, Ö., Karaman, M., Kanik, E. T., Firinci, F., İnan, S., Yüksel, H."Anti‑VEGF treatment suppresses remodeling factors and restores epithelial barrier function through the E‑cadherin/β‑catenin signaling axis in experimental asthma models". Experimental and Therapeutic Medicine 22, no. 1 (2021): 689. https://doi.org/10.3892/etm.2021.10121