Download

ORIGINAL ARTICLE

MiR-506 targets polypyrimidine tract-binding protein 1 to inhibit airway inflammatory response and remodeling via mediating Wnt/β-catenin signaling pathway

Yuxiang Caia, Jifeng Tianb, Yufei Sua, Xiaolan Shic*

aDepartment of Emergency, Xi’an Children’s Hospital, Xi’an, Shaanxi, China

bDepartment of Integrated Traditional Chinese and Western Medicine, Xi’an Children’s Hospital, Xi’an, Shaanxi, China

cDepartment of Respiratory Asthma Center, Xi’an Children’s Hospital, Xi’an, Shaanxi, China

Abstract

Background: Airway remodeling, which contributes to the clinical course of childhood asthma, occurs due to airway inflammation and is featured by anomalous biological behaviors of airway smooth muscle cells (ASMCs). microRNA (miRNA) plays an essential role in the etiopathogenesis of asthma.

Objective: This research was aimed to characterize miR-506 in asthma and uncover potential regulatory machinery.

Material and methods: The asthmatic cell model was established by treating ASMCs with transforming growth factor-beta1 (TGF-β1) and assessed by the levels of interleukin (IL)-1β and interferon gamma (IFN-γ). Using real-time quantitative polymerase chain reaction, mRNA expression of miR-506 and polypyrimidine tract-binding protein 1 (PTBP1) was measured. Cell counting kit-8 and Transwell migration tests were used for estimating the capacity of ASMCs to proliferate and migrate. Luciferase reporter assay was used to corroborate whether miR-506 was directly bound to PTBP1. Expression of PTBP1, collagen I and III, and essential proteins of the wingless-related integration (Wnt)/β-catenin pathway (β-catenin, c-MYC and cyclin D1) was accomplished by Western blot analysis. The involvement of Wnt/β-catenin signaling in asthma was confirmed by Wnt signaling pathway inhibitor (IWR-1).

Results: miR-506 was poorly expressed in asthmatic tissues and cell model. Functionally, overexpression of miR-506 reduced aberrant proliferation, migration, inflammation and collagen deposition of ASMCs triggered by TGF-β1. Mechanically, miR-506 directly targeted the 3’ untranslated region (3-UTR) of PTBP1 and had a negative regulation on PTBP1 expression. Moreover, overexpression of miR-506 suppressed the induction of Wnt/β-catenin pathway. The administration of IWR-1 further validated negative correlation between miR-506 and the Wnt/β-catenin pathway in asthma.

Conclusion: Our data indicated that targeting miR-506/PTBP1/Wnt/β-catenin axis might point in a helpful direction for treating asthma in children.

Key words: airway smooth -muscle cells, childhood asthma, miR-506, PTBP1, Wnt/β-catenin

*Corresponding author: Xiaolan Shi, Department of Respiratory Asthma Center, Xi’an Children’s Hospital, No. 69, Xijuyuan Lane, Lianhu District, Xi’an, Shaanxi 710003, China. Email address: [email protected]

Received 19 May 2022; Accepted 12 October 2022; Available online 1 May 2023

DOI: 10.15586/aei.v51i3.676

Copyright: Cai Y, et al.
License: This open access article is licensed under Creative Commons Attribution 4.0 International (CC BY 4.0). http://creativecommons.org/licenses/by/4.0/

Introduction

Recently, childhood asthma has become one of the prevalent chronic illnesses in pediatric patients.1 Airway irritation tends to induce mucus accumulation, airway inflammatory response, and remodeling, leading to the onset of asthma.2 Asthma etiology is linked to the increased number of airway smooth muscle cells (ASMCs) along with their migration, and secreted adhesion molecules, cytokines, matrix components and metalloproteinases secreted by ASMCs; these secretions promoted the proliferation of ASMCs and aggravated airway remodeling.3,4 Therefore, the inhibition of pathobiological processes in ASMCs provides a therapeutic approach for childhood asthma.

Highly conserved microRNAs (miRNAs) are small non-coding RNAs that are responsible for regulating the post-transcriptional expression of genes. Research studies have shown the vital role of miRNAs in regulating ASMCs malignant phenotypes during asthma.5 For instance, miR-221 regulated the expression of p21WAF1 and p27kip1 to enhance the aberrant proliferation of ASMCs.6 In contrast, miR-217 interacted with zinc finger E-box binding homeobox 1 (ZEB1) to repress abnormal ASMCs survival and migration.7 The protective effects of miR-506 in progression of different cancers, especially in lung cancer, were reported in previous studies, which raised our concern.810 A research uncovered that miR-506-3p inhibitor contributed to non-small cell lung cancer tumorigenesis by boosting the proliferative, migratory as well as invasive properties of cells.11 In addition, overexpression of miR-506 was reported to downregulate Tubby-like protein 3, leading to an inhibitory pathological process in lung cancer.12 Moreover, miR-506 was apparently downregulated and its overexpression inhibited pulmonary fibrosis by reducing apoptosis and improving inflammation in a lung fibrosis model.13 However, the link between miR-506 and asthma etiology still needs to be explained.

Polypyrimidine tract-binding protein 1 (PTBP1), a member of the heterogeneous ribonucleoprotein subfamily, participates in pre-mRNA processing, and mRNA metabolism and transport.1416 PTBP1 has been verified to play an oncogenic function in multiple cancers, including lung cancer.17,18 Furthermore, abnormal expression of PTBP1 was involved in controlling integral metabolism and proliferation in pulmonary hypertension and fibrosis.19,20 PTBP1 was markedly upregulated in a vascular smooth muscle cells (VSMCs) injury model, and PTBP1 knockdown resulted in a reduced proliferation of VSMCs.21

Here, we established a transforming growth factor--beta1 (TGF-β1)-stimulated airway smooth muscle cells (AMSCs) model of asthma in vitro. In this model, we investigated the protective properties of miR-506 and the modulatory regimes involved. Our study offers novel insights for improving pediatric asthma treatment.

Materials and Methods

Clinical tissues

In this study, 50 asthma patients with a balanced gender ratio of 1:1 were recruited; patients’ varied between 5 and 12 years, with an average age of 8.2 ± 3.5 years. The control group included children who were diagnosed as having no history of respiratory and allergy-related diseases within 1 month by the respiratory asthma department of Xi’an Children’s Hospital. A total of 12 boys and 13 girls were enrolled in the control group of this study. The age of these children ranged from 6 to 13 years, with a mean age of 8.2 ± 3.1 years. All tissue samples were extracted by performing biopsy of the subjects according to a previous study,22 and cryopreserved for subsequent experiments.

The study was conducted in accordance with the Declaration of Helsinki, and was authorized by the Ethics Committee of Xi’an Children’s Hospital. Informed consent was taken for all subjects.

Cell culture and treatment

Human-derived ASMCs were obtained from Procell Life Science & Technology Co. Ltd. (Cat. No. CP-H014; Wuhan, China) and cultivated in Dulbecco’s modified Eagles Medium (DMEM; Cat. No. D5796; Sigma-Aldrich, St Louis, USA) enriched with 10% fetal bovine serum under the condition of 5% CO2 and 37°C. We developed an airway remodeling cell model as described by Cao et al.7 In short, ASMCs were exposed to 10-ng/mL TGF-β1 (Cat. No. 75362; Cell Signaling Technology, Shanghai, China) for 24 h. Besides, ASMCs were treated with 2.5-μM wingless-related integration (Wnt)/ β-catenin inhibitor (IWR-1; Cat. No. I0161; Sigma-Aldrich) for 24 h.

Cell transfection

In order to enhance expression of PTBP1, the amplified DNA fragment of PTBP1 was cloned into plasmid cloning (pc) DNA3.1 (Cat. No. V79020; Invitrogen, Carlsbad, CA, USA). MiR-506 mimic, miR-NC, anti-miR-506, and anti-miR-NC were obtained from GenePharma Co. (Shanghai, China). The specific sequences of mimics were as follow: miR-506 mimic (5'-UAAGGCACCCUUCUGAGUAGA-3'), miR-NC (5'-UUCUCCGAACGUGUCACGUTT-3'), anti-miR-506 (5'-UCUACUCAGAAGGGUGCCUUA-3'), and anti-miR-NC (5'-CAGUACUUUUGUGUAGUACAA-3'). The transfections of the above-mentioned plasmids and miRNAs in ASMCs were conducted with Lipofectamine 3000 (Cat. No. L3000015;, Invitrogen) in compliance with -manufacturer’s protocol.

Real-time quantitative polymerase chain reaction (RT-qPCR)

Using Trizol (Cat. No. 15596018; Invitrogen), total RNA was extracted from ASMCs. M-MLV (Cat. No. 28025021; Invitrogen) reverse transcriptase kit was used to obtain complementary DNA (cDNA). RT-qPCR was performed on a 7500 real-time PCR machine (Cat. No. 4351107; Applied Biosystems, Shanghai, China) with a TB Green Premix Ex Taq II (Cat. No. RR820Q; Takara, Dalian, China) and the relative mRNA level was quantified by the 2−ΔΔCt method. Reference controls for miRNAs and mRNAs were U6 and glyceraldehyde 3-phosphate dehydrogenase (GAPDH), respectively. The sequences of primers are listed in Table 1.

Table 1 Primers for RT-PCR of genes.

Genes Sequences (5' to 3')
miR-506-F TAAGGCACCCTTCT-GAGTAGA
miR-506-R GCGAGCACAGAATTAATACGAC
U6-F CTCGCTTCGGCAGCACA
U6-R ACGCTTCACGAATTTGCGT
PTBP1-F CTGAGGATCCATGTCTGGTTATTCTAGTG
PTBP1-R TTACTCTCGAGTTACTGGGAATATCCGGTT
GAPDH-F ACCACAGTCCATGCCATCAC
GAPDH-R TCCACCACCCTGTTGCTGTA

Western blot analysis

Radioimmunoprecipitation assay (RIPA) buffer (Cat. No. 89900; Sigma-Aldrich) was applied to lyse ASMCs to obtain total proteins. Then, protein quantification was executed with a bicinchoninic acid (BCA) protein assay kit (Cat. No. P0012S; Beyotime, Shanghai, China). Next, the proteins were separated by electrophoreses and transferred onto polyvinylidene fluoride (PVDF) membranes (Cat. No. VVLP02500; Millipore, MA, USA). The membranes were treated with 5% skimmed milk for 1 h. The following antibodies were used as primary antibodies: anti-PTBP1 (1:1000; ab5642; Abcam, Shanghai, China), anti-collagen I (1:1000; ab138492; Abcam), anti--collagen III (1:1000; ab6310; Abcam), anti-β-catenin (1:1000; ab32572; Abcam), anti-c-MYC (1:1000; ab32072; Abcam), anti-cyclin D1 (1:1000; ab16663; Abcam), and anti-GAPDH (1:5000; sc-47724; Santa Cruz Biotechnology, TX, USA). Incubation of primary antibodies was carried out overnight at 4°C; then, the membranes were washed and treated with horseradish peroxidase-conjugated secondary antibody (1:5000; sc-2004; Santa Cruz) for 1 h at room temperature. Eventually, the results were visualized using enhanced -chemiluminescence (ECL) reagents (Cat. No. 32209; Millipore). Using Image J (NIH, MD, USA), GAPDH was -utilized to normalize protein bands.

Luciferase reporter assay

An online website TargetScan 7.2 (http://www.targetscan.org/ vert_72/) predicted possible correlation between PTBP1 and miR-506. The 3’ untranslated regions (3-UTR) of PTBP1 mutant (MUT) and wild-type (WT) bound to miR-506 were inserted into psiCHECK (Cat. No. C8021; Promega, Madison, WI, USA). Co-transfection of these recombinant plasmids into ASMCs with miR-506 mimics or miR-NC was accomplished by using Lipofectamine 3000 (Invitrogen). Following 48-h transfection, renilla luciferase activity was utilized to standardize luciferase reporter activity on a Dual Luciferase Reporter Assay System (Promega).

Cell counting kit (CCK)-8 assay

First, ASMCs were added into 96-well plates (2 × 104 cells/well). Subsequently, the proliferative activity of ASMCs was determined with CCK-8 solution (Cat. No. CK04; Dojindo Molecular Technologies, Shanghai, China) as directed by the manufacturer. Finally, analysis of absorbance at 450 nm was recorded using a microplate reader (Cat. No. VL0000D2; Thermo Fisher Scientific, Waltham, MA, USA).

Enzyme-linked immunosorbent serological assay (ELISA)

The measurements of interleukin (IL)-1β and i nterferon gamma (IFN-γ) levels in cell supernatant were conducted using commercial ELISA kits (Cat. No. PI305 and PI511; Beyotime) according to instruction manual of the manufacturer. IL-1β and IFN-γ contents were also calculated based on the absorbance values at 450 nm.

Wound healing assay

ASMCs were planted into 24-well plates (5 × 104 cells per well). After cell confluency reached 90%, a sterile disposable pipette was used to make a scratch in the center of cell monolayer. Images were captured under a microscope (×100 magnification; Cat. No. CX43/33; Olympus, Tokyo, Japan) at 0 h and 48 h after scratching to quantify and assess the ability of migration.

Transwell assay

In addition, an 8-μm pore size Transwell chamber (Cat. No. 430157; Costar, Suzhou, China) was used to evaluate cell migration. After serum starvation for 24 h, resuspended ASMCs in serum-free medium was put in the top of 24-well Transwell chamber. The lower part of Transwell chamber was loaded with 10% DMEM. After being cultured for 48 h, the unmigrated cells were removed gently, and the migrated cells in the bottom of the chamber were fixed with formaldehyde and stained with crystal violet. Cell migration rate was calculated in five randomly selected fields of view with a light microscope (×100 magnification; Cat. No. CX43/33; Olympus).

Statistical analysis

All experiments were carried out thrice independently. SPSS 22.0 was used for all statistical analyses. The results were presented as mean ± standard error of mean (SEM). In order to identify the significance of variations between groups, Student’s t-test and ANOVA were applied. Spearman’s Rank test was applied to ascertain correlations between miR-506 and PTBP1 in asthmatic tissues. P < 0.05 was considered as statistically significant difference.

Results

miR-506 was poorly expressed in asthmatic tissues and TGF-β1-induced ASMCs

We first identified miR-506 expression in asthmatic tissues and a TGF-β1-stimulated in vitro cell model of asthma. We observed that miR-506 expression was markedly attenuated in asthmatic tissues, compared to normal tissues (Figure 1A). Likewise, a remarkable decline in miR-506 expression was discovered in TGF-β1-stimulated ASMCs, compared to untreated ASMCs. Meanwhile, the results showed that miR-506 mimic dramatically upregulated TGF-β1-induced miR-506 expression whereas anti-miR-506 effectively attenuated the role of TGF-β1 in miR-506 expression (Figure 1B).

Figure 1 miR-506 overexpression repressed abnormal proliferation and inflammatory responses of ASMCs induced by TGF-β1. (A) RT-qPCR analysis of miR-506 expression in asthmatic tissues. *P < 0.05 versus healthy patients. (B) Transfection of ASMCs with miR-506 mimic or anti-miR-506 was followed by a 24-h incubation with 10-ng/mL TGF-β1. RT-qPCR was conducted to determine transfection efficiency in ASMCs. (C) CCK-8 test was used to measure cell viability. (D and E) Contents of inflammation-related factors, IL-1β and IFN-γ, were measured in cell supernatant by ELISA. *P < 0.05 vs untreated group, #P < 0.05 vs TGF-β1 + miR-NC group or TGF-β1 + anti-miR-NC group. Data are shown as mean ± SEM (n = 3).

miR-506 inhibited TGF-β1-stimulated aberrant cellular proliferation and inflammatory responses of ASMCs

Subsequently, ASMCs were exposed to TGF-β1 to imitate asthma environment in vivo. CCK-8 assay showed that exposure to TGF-β1 elevated cell viability considerably. Interestingly, overexpression of miR-506 remarkably suppressed the abnormal proliferation of ASMCs evoked by TGF-β1 whereas anti-miR-506 exacerbated the malignant growth of ASMCs (Figure 1C). In addition, we found that TGF-β1 treatment elevated IL-1β levels and inhibited IFN-γ production in the supernatant of ASMCs culture system, while the upregulation of miR-506 reversed this effect, and anti-miR-506 aggravated TGF-β1-stimulated abnormal production of pro-inflammatory factors (Figures 1D and 1E). The findings revealed that miR-506 had an inhibitory effect on TGF-β1-stimulated aberrant cellular proliferation and inflammation of ASMCs.

miR-506 repressed TGF-β1-stimulated aberrant migration and collagen deposition of ASMCs

Next, we addressed the involvement of miR-506 in TGF-β1-stimulated aberrant migration of ASMCs. Administration of TGF-β1 dramatically increased the migratory capacity of ASMCs, while miR-506 mimic markedly depressed this effect (Figures 2A and 2B). Meanwhile, the downregulation of miR-506 induced elevation in the migratory capacity of ASMCs evoked by TGF-β1. Besides, excessive collagen deposition is also an essential factor leading to airway remodeling.23 Further validation experiments revealed that expression of collagen I and collagen III was remarkably enhanced via TGF-β1 treatment. In addition, anti-miR-506 markedly promoted this effect whereas miR-506 mimic mitigated this effect (Figure 2C). Altogether, these findings supported that miR-506 attenuated TGF-β1-stimulated aberrant ASMCs migration and collagen deposition.

Figure 2 miR-506 overexpression reduced abnormal migration and collagen deposition of ASMCs induced by TGF-β1. Transfection of ASMCs with miR-506 mimic or inhibitors was followed by a 24-h treatment with 10-ng/mL TGF-β1. (A) Wound healing test, and (B) Transwell migration test were adopted to evaluate migrative capacity in ASMCs. (C) Western blot analysis was used to quantify collagen I and collagen III protein expressions. *P < 0.05 vs untreated group, #P < 0.05 vs TGF-β1 + miR-NC group or TGF-β1 + anti-miR-NC group. Data are shown as mean ± SEM (n = 3).

miR-506 directly targeted to PTBP1

A recent study has demonstrated that PTBP1 was involved in the aberrant inflammation and migration of ASMCs evoked by TGF-β1.24 Interestingly, we used TargetScan 7.2 to identify PTBP1 as a potential target for miR-506 and predicted the matched regions between miR-506 and PTBP1 (Figure 3A). Further investigation revealed that miR-506 mimic markedly diminished the Luciferase activity of PTBP1-WT (Figure 3B). Moreover, PTBP1 expression was enhanced in TGF-β1-induced ASMCs, which was dramatically diminished by miR-506 mimic (Figures 3C and 3D). In addition, PTBP1 expression was dramatically elevated in asthma tissues, compared to healthy tissues and had a negative regulatory relationship with miR-506 expression (Figures 3E and 3F). The above data indicated that miR-506 directly targeted to PTBP1 and served as a negative regulator of PTBP1.

Figure 3 PTBP1 was verified as a downstream target of miRNA-506 in asthma. (A) miRNA database predicted the bond locations of miR-506 to PTBP1. (B) The relative luciferase activity of PTBP1 3'-UTR following transfection with miR-506 mimic was determined using a luciferase reporter experiment. *P < 0.05 vs miR-NC group. (C) Transfection of ASMCs with miR-506 mimic or inhibitors was followed by a 24-h treatment with 10-ng/mL TGF-β1. RT-qPCR analysis of PTBP1 mRNA expression in ASMCs. (D) Western blot analysis of PTBP1 protein expression in ASMCs. *P < 0.05 vs control group, #P < 0.05 vs TGF-β1 + miR-NC group. (E) RT-qPCR was used to check miR-506 expression in asthmatic tissues. *P < 0.05 vs healthy patients. (F) Relationship between miR-506 and PTBP1 was analyzed. Data are shown as mean ± SEM (n = 3).

miR-506/PTBP1 axis repressed TGF-β1-stimulated aberrant cellular proliferation and inflammatory responses of ASMCs by regulating Wnt/β-catenin signaling pathway

Wnt/β-catenin signaling pathway exerts a profound effect on pathogenesis of asthma.25 Therefore, we tried to unveil the correlation between miR-506 and Wnt/β-catenin signaling pathway. The result pointed out that TGF-β1 treatment significantly enhanced expression of key target proteins (β-catenin, c-MYC, and cyclin D1) in Wnt signaling pathway. miR-506 overexpression significantly suppressed the TGF-β1-triggered activation of Wnt signaling pathway, while PTBP1 overexpression reversed these effects (Figure 4A).

Figure 4 The alleviating effects of miR-506 on TGF-β1-stimulated aberrant proliferation, migration, and inflammatory responses of ASMCs were mediated by an altered PTBP1/Wnt/β-catenin axis. (A) Western blot analysis of the protein expressions of β-catenin, c-MYC, and cyclin D1. *P < 0.05 vs untreated group, #P < 0.05 vs TGF-β1 + pcDNA3.1 group. $P < 0.05 vs TGF-β1 + miR-506 group. (B) Cell proliferation was monitored by CCK-8 test. (C and D) Cell migration was evaluated by wound healing and Transwell experiments. (E and F) ELISA detection was performed to quantify the production of IL-1β and IFN-γ. *P < 0.05 vs TGF-β1 + pcDNA3.1 group, #P < 0.05 vs TGF-β1 + miR-506 + pcDNA3.1 group. $P < 0.05 vs TGF-β1 + miR-506 + PTBP1 group. Data are shown as mean ± SEM (n = 3).

Rescue assays were performed in TGF-β1-induced ASMCs using IWR-1 inhibitor. As depicted in Figures 4B–D, miR-506 mimic markedly repressed TGF-β1-stimulated aberrant proliferative and migratory activity of ASMCs whereas the upregulation of PTBP1 dramatically abated these effects. Additionally, miR-506 overexpression suppressed the generation of IL-1β and enhanced the secretion of IFN-γ in asthmatic cell model whereas opposite effects were observed in the PTBP1 overexpression groups. Moreover, the repressive role of miR-506 mimics in proliferation, migration as well as inflammatory responses of TGF-β1-stimulated ASMCs was greatly counteracted by PTPB1 overexpression, which was markedly reversed by exogenous IWR-1 (Figures 4B–F). Overall, miR-506/PTBP1 axis inhibited proliferation, migration, and cellular inflammatory responses of TGF-β1-stimulated ASMCs through inactivated Wnt/β-catenin pathway.

Discussion

The present treatment for severe or gentle asthma is primarily focused on inhalation of corticosteroids, long-acting β2-agonists, and leukotriene antagonists. Although traditional therapy has made great progress, treatment of persistent and uncontrolled asthma in children is still a problem.26 Molecular targeted therapy has become a novel alternative for asthma patients, especially in controling airway inflammatory responses and remodeling. Our data indicated that miR-506 modulated the alterations of proliferation, migration, inflammation, and collagen deposition in a TGF-β1-induced asthmatic cell model by targeting PTBP1 to inactivate the Wnt/β-catenin pathway.

Growing research suggests that miRNAs is instrumental in controlling the progression of asthma.2729 For example, miR-142-5p and miR-130a-3p have been shown to be engaged in pulmonary inflammation and airway phenotypic alterations.30 MiR-30a inhibited autophagy, and hence reduced airway fibrosis in a mouse model.31 MiR-506 is a newly identified miRNA involved in multiple biological processes, mostly reported in cancer. Notably, the sequences that contained single nucleotide polymorphism rs3809865, significantly associated with asthma, was predicted to bind to the seed regions of miR-506,32 which raised our speculation that miR-506 may be associated with asthma. At the same time, Wang et al. observed that miR-506-3p expression was declined in ovalbumin (OVA)-based asthma model, and its overexpression reduced growth and induced apoptosis of ASMCs.33 Consistently, our data suggested that upregulated miR-506 suppressed ASMCs proliferation and migration during asthma progression. Simultaneously, upregulated miR-506 reduced inflammation and collagen deposition in an asthmatic in vitro cell model evoked by TGF-β1. Additionally, a previous research confirmed a positive role of miR-506 in the fibrosis and inflammatory response of nonalcoholic fatty liver disease.34 Therefore, it implied that miR-506 presented a protective role in the development of asthma.

miRNA-mediated gene regulation has a variety of regulatory modes by targeting different regions, such as coding regions,35 3'-UTR,36 or 5'-UTR.37 Among them, the binding of miRNAs to the 3'-UTR of target genes is deemed as a classic way to participate in the regulatory mechanism of airway remodeling.38 For example, Sun et al. documented that miR-874 inhibited TNF-α-stimulated human fetal ASMCs remodeling by targeting Signal transducer and activator of transcription 3 (STAT3).39 In a study conducted by Chen et al., miR-23b directly bound to Smad3 to suppress ASMCs proliferation.40. Interestingly, PTBP1 was considered as a crucial RNA-binding protein in asthmatic tissues and a common miRNA-target.41,42 However, there were only two studies regarding the effects of PTBP1 in asthma etiology. In our work, the direct target of miR-506 was identified as PTBP1. Consistent with our findings, another research demonstrated that miR-506-3p mediated the direct regulation of PTBP1 in gastric cancer.43 Besides, our research indicated that PTBP1 was greatly upregulated in asthmatic tissues and promoted abnormal biological behavior in TGF-β1-stimulated ASMCs. Similarly, PTBP1 silencing was reported to inhibit the malignant phenotype of VSMCs and lung cancer cells.18,21 Additionally, we observed a promotive impact of PTBP1 on TGF-β1-stimulated inflammatory responses in ASMCs. Consistently, a recent study has shown that the overexpression of PTBP1 enhanced ASMCs proliferation, migration, and inflammation by upregulating neuro-oncological ventral antigen 1.24

As a well-known pathway, Wnt/β-catenin pathway is extremely relevant for cell growth and development, and has been connected to recent findings of asthma. For instance, in TGF-β1-induced bronchial smooth muscle cell, miR-188 mediated the blockage of Wnt/β-catenin signaling pathway to dampen cell proliferation.44 Induction of Wnt/β-catenin signaling pathway mediated by miR-21 exacerbated airway inflammation and oxidative damage in asthma.45 Therefore, this further aroused us to speculate whether Wnt/β-catenin signaling took part in miR-506 modulation of asthma pathogenesis. Previous study has indicated that a positive regulatory relationship existed between PTPB1 and Wnt/β-catenin signaling in glioma.46 Furthermore, several evidences indicated that miR-506 regulated the tumorigenesis of osteosarcoma via Wnt/β-catenin signaling pathway.47 Our data pointed out that overexpression of miR-506 inactivated Wnt/β-catenin pathway to mitigate TGF-β1-mediated adverse progression of asthma, while PTBP1 exacerbated TGF-β1-stimulated airway remodeling and inflammation in ASMCs by triggering the Wnt/β-catenin signaling pathway.

The limitation of this study was its sample size, which was limited; moreover, the animal model of asthma was not created. Further investigation of mechanism is required to verify the findings of this study, which would help to provide a theoretical basis to develop new agents for asthma therapy.

Conclusion

To conclude, this research implied that miR-506 targeted PTPB1 to alleviate TGF-β1-induced airway inflammatory response and remodeling in ASMCs by suppression of Wnt/β-catenin pathway. This would figure out the possibility of miR-506 in treating asthma.

Conflict of interest

The authors declare no potential conflicts of interest with respect to research, authorship, and/or publication of this paper.

Data availability

All data are available from corresponding author with reasonable requirements.

Author Contributions

Xiaolan Shi designed experiments. Jifeng Tian analyzed clinical samples. Yufei Su performed testing and laboratory support. Yuxiang Cai conducted data analysis. Yuxiang Cai and Xiaolan Shi wrote and edited the manuscript. All authors adopt the publication of the final manuscript.

REFERENCES

1. Huang Y, Tzeng J, Maguire R, Hoyo C, Allen T. The association between neuraxial anesthesia and the development of childhood asthma—A secondary analysis of the newborn epigenetics study cohort. Curr Med Res Opin. 2020 Apr;36(6): 1025–32. 10.1080/03007995.2020.1747417

2. Barnes PJ. Endogenous inhibitory mechanisms in asthma. Am J Respir Crit Care Med. 2000 Mar;161(2):S176–81. 10.1164/ajrccm.161.supplement_2.a1q4-6

3. Liu Y, Miao Y, Gao X, Wang YY, Wang H, Zheng YW, et al. MicroRNA-200a affects the proliferation of airway smooth muscle cells and airway remodeling by targeting FOXC1 via the PI3K/AKT signaling pathway in ovalbumin-induced asthmatic mice. Cell Physiol Biochem. 2018 Nov;50(6):2365–89. 10.1159/000495097

4. Chen YF, Huang G, Wang YM, Cheng M, Zhu FF, Zhong JN, et al. Exchange protein directly activated by cAMP (Epac) protects against airway inflammation and airway remodeling in asthmatic mice. Respir Res. 2019 Dec;20(1):285. 10.1186/s12931-019-1260-2

5. Munitz A, Karo-Atar D, Foster PS. Asthma diagnosis: MicroRNAs to the rescue. JJ Allergy Clin Immunol. 2016 May;137(5):1447–48. 10.1016/j.jaci.2016.02.013

6. Perry MM, Baker JE, Gibeon DS, Adcock IM, Chung KF. Airway smooth muscle hyperproliferation is regulated by microRNA-221 in severe asthma. Am J Respir Cell Mol Biol. 2014 Jan;50(1):7–17. 10.1165/rcmb.2013-0067OC

7. Gao Y, Wang B, Luo H, Zhang Q, Xu M. miR-217 represses TGF-β1-induced airway smooth muscle cell proliferation and migration through targeting ZEB1. Biomed Pharmacother. 2018 Dec;108:27–35. 10.1016/j.biopha.2018.09.030

8. Peng T, Zhou L, Zuo L, Luan Y. MiR-506 functions as a tumor suppressor in glioma by targeting STAT3. Oncol Rep. 2016 Feb;35(2):1057–64. 10.3892/or.2015.4406

9. Guo S, Yang P, Jiang X, Li X, Wang Y, Zhang X, et al. Genetic and epigenetic silencing of mircoRNA-506-3p enhances COTL1 oncogene expression to foster non-small lung cancer progression. Oncotarget. 2017 Jan;8(1):644–57. 10.18632/oncotarget.13501

10. Hossian A, Sajib MS, Tullar PE, Mikelis CM, Mattheolabakis G. Multipronged activity of combinatorial miR-143 and miR-506 inhibits lung cancer cell cycle progression and angiogenesis in vitro. Sci Rep. 2018 Jul;8(1):10495. 10.1038/s41598-018-28872-2

11. Li L, Wei H, Zhang H, Xu F, Che G. Circ_100565 promotes proliferation, migration and invasion in non-small cell lung cancer through upregulating HMGA2 via sponging miR-506-3p. Cancer Cell Int. 2020 May;20:160. 10.1186/s12935-020-01241-8

12. Li ZH, Zhou JH, Chen SN, Pan L, Feng Y, Luo MQ, et al. MicroRNA-506 has a suppressive effect on the tumorigenesis of nonsmall-cell lung cancer by regulating tubby-like protein 3. Bioengineered. 2021 Dec;12(2):10176–86. 10.1080/21655979.2021.2001216

13. Zhu M, An Y, Zhang X, Wang Z, Duan H. Experimental pulmonary fibrosis was suppressed by microRNA-506 through NF-kappa-mediated apoptosis and inflammation. Cell Tissue Res. 2019 Nov;378(2):255–65. 10.1007/s00441-019-03054-2

14. Spellman R, Rideau A, Matlin A, Gooding C, Robinson F, McGlincy N, et al. Regulation of alternative splicing by PTB and associated factors. Biochem Soc Trans. 2005 Jun;33(3): 457–60. 10.1042/BST0330457

15. Sawicka K, Bushell M, Spriggs KA, Willis AE. Polypyrimidine-tract-binding protein: A multifunctional RNA-binding protein. Biochem Soc Trans. 2008 Aug;36(4):641–47. 10.1042/BST0360641

16. Wollerton MC, Gooding C, Wagner EJ, Garcia-Blanco MA, Smith CW. Autoregulation of polypyr0imidine tract binding protein by alternative splicing leading to nonsense--mediated decay. Mol Cell. 2004 Jan;13(1):91–100. 10.1016/S1097-2765(03)00502-1

17. Llorian M, Schwartz S, Clark TA, Hollander D, Tan L-Y, Spellman R, et al. Position-dependent alternative splicing activity revealed by global profiling of alternative-splicing events regulated by PTB. Nat Struct Mol Biol. 2010 Sep;17(9):1114. 10.1038/nsmb.1881

18. Cho CY, Chung SY, Lin S, Huang JS, Chen YL, Jiang SS, et al. PTBP1-mediated regulation of AXL mRNA stability plays a role in lung tumorigenesis. Sci Rep. 2019 Nov;9(1):1–12. 10.1038/s41598-019-53097-2

19. Caruso P, Dunmore BJ, Schlosser K, Schoors S, Dos Santos C, Perez-Iratxeta C, et al. Identification of microRNA-124 as a major regulator of enhanced endothelial cell glycolysis in pulmonary arterial hypertension via PTBP1 (polypyrimidine tract binding protein) and pyruvate kinase M2. Circulation. 2017 Dec;136(25):2451–67. 10.1161/CIRCULATIONAHA.117.028034

20. Xu T, Yan W, Wu Q, Xu Q, Yuan J, Li Y, et al. MiR-326 inhibits inflammation and promotes autophagy in silica-induced pulmonary fibrosis through targeting TNFSF14 and PTBP1. Chem Res Toxicol. 2019 Nov;32(11):2192–203. 10.1021/acs.chemrestox.9b00194

21. Wang Z, Gan X, Qiu C, Yang D, Sun X, Zeng Z. Role of polypyrimidine tract-binding protein 1/yin yang 2 signaling in regulating vascular smooth muscle cell proliferation and neointima hyperplasia. Toxicol Appl Pharmacol. 2019 Nov;383:114747. 10.1016/j.taap.2019.114747

22. Yick CY, Zwinderman AH, Kunst PW, Grünberg K, Mauad T, Chowdhury S, et al. Gene expression profiling of laser-microdissected airway smooth muscle tissue in asthma and atopy. Allergy. 2014 Sep;69(9):1233–40. 10.1111/all.12452

23. Tang X, Nian H, Li X, Yang Y, Wang X, Xu L, et al. Effects of the combined extracts of Herba Epimedii and Fructus Ligustrilucidi on airway remodeling in the asthmatic rats with the treatment of budesonide. BMC Complement Altern Med. 2017 Aug;17(1):380. 10.1186/s12906-017-1891-0

24. Cheng Y, Wang N, Zhao L, Liu C, Wang J, Ma C, et al. Knockdown of NOVA1 inhibits inflammation and migration of asthmatic airway smooth muscle cells to regulate PTEN/Akt pathway by targeting PTBP1. Mol Immunol. 2021 Oct;138: 31–37. 10.1016/j.molimm.2021.07.016

25. Koopmans T, Gosens R. Revisiting asthma therapeutics: Focus on WNT signal transduction. Drug Discov Today. 2018 Jan;23(1):49–62. 10.1016/j.drudis.2017.09.001

26. Porcaro F, Ullmann N, Allegorico A, Di Marco A, Cutrera R. Difficult and severe asthma in children. Children. 2020 Dec;7(12):286. 10.3390/children7120286

27. Yu Y, Men S, Zhang Y. miR-20a-5p ameliorates ovalbumin (OVA)-induced mouse model of allergic asthma through targeting ATG7-regulated cell death, fibrosis and inflammation. Int Immunopharmacol. 2021 Jun;95:107342. 10.1016/j.intimp.2020.107342

28. Wang J, Dong X, Yu Z, Ge L, Lu L, Ding L, et al. Borneol inhibits CD4+T cells proliferation by down-regulating miR-26a and miR-142-3p to attenuate asthma. Int Immunopharmacol. 2021 Jan;90:107223. 10.1016/j.intimp.2020.107223

29. Fan Q, Jian Y. MiR-203a-3p regulates TGF-β1-induced epithelial-mesenchymal transition (EMT) in asthma by regulating Smad3 pathway through SIX1. Biosci Rep. 2020 Feb;40(2):BSR20192645. 10.1042/BSR20192645

30. Shi J, Chen M, Ouyang L, Wang Q, Guo Y, Huang L, et al. miR-142-5p and miR-130a-3p regulate pulmonary macrophage polarization and asthma airway remodeling. Immunol Cell Biol. 2020 Oct;98(9):715–25. 10.1111/imcb.12369

31. Li BB, Chen YL, Pang F. MicroRNA-30a targets ATG5 and attenuates airway fibrosis in asthma by suppressing autophagy. Inflammation. 2020 Feb;43(1):44–53. 10.1007/s10753-019-01076-0

32. Zhang Y, Han Y, Dong L, Yu H, Cheng L, Zhao X, et al. Genetic variation of ITGB3 is associated with asthma in Chinese Han children. PLoS ONE. 2013 Feb;8(2):e56914. 10.1371/journal.pone.0056914

33. Wang ML, Qiao H. Effect of miR-506-3p on proliferation and apoptosis of airway smooth muscle cells in asthmatic mice by regulating CCL2 gene expression and mediating TLR4/NF-κB signaling pathway activation. Mol Biotechnol. 2021 May;63(5):410–23. 10.1007/s12033-021-00309-8

34. Jin SS, Lin XF, Zheng JZ, Wang Q, Guan HQ. lncRNA NEAT1 regulates fibrosis and inflammatory response induced by nonalcoholic fatty liver by regulating miR-506/GLI3. Eur Cytokine Netw. 2019 Sep 1;30(3):98–106. 10.1684/ecn.2019.0432

35. Huang S, Wu S, Ding J, Lin J, Wei L, Gu J, et al. MicroRNA-181a modulates gene expression of zinc finger family members by directly targeting their coding regions. Nucleic Acids Res. 2010 Nov;38(20):7211–18. 10.1093/nar/gkq564

36. Bartel DP. MicroRNAs: Target recognition and regulatory functions. Cell. 2009 Jan;136(2):215. 10.1016/j.cell.2009.01.002

37. Panda AC, Itishri S, Kulkarni SD, Martindale JL, Kotb A, Arya V, et al. miR-196b-mediated translation regulation of mouse insulin2 via the 5'UTR. PLoS ONE. 2014 Jul;9(7):e101084. 10.1371/journal.pone.0101084

38. Shao Y, Chong L, Lin P, Li H, Zhu L, Wu Q, et al. MicroRNA-133a alleviates airway remodeling in asthtama through PI3K/AKT/mTOR signaling pathway by targeting IGF1R. J Cell Physiol. 2019 Apr;234(4):4068–80. 10.1002/jcp.27201

39. Sun M, Huang Y, Li F, Li H, Zhang B, Jin L. MicroRNA-874 inhibits TNF-α-induced remodeling in human fetal airway smooth muscle cells by targeting STAT3. Respir Physiol Neurobiol. 2018 May;251:34–40. 10.1016/j.resp.2018.02.008

40. Chen M, Shi J, Zhang W, Huang L, Lin X, Lv Z, et al. MiR-23b controls TGF-β1-induced airway smooth muscle cell proliferation via direct targeting of Smad3. Pulm Pharmacol Ther. 2017 Feb;42:33–42. 10.1016/j.pupt.2017.01.001

41. Kang H, Heo S, Shin JJ, Ji E, Tak H, Ahn S, et al. A miR-194/PTBP1/CCND3 axis regulates tumor growth in human hepatocellular carcinoma. J Pathol. 2019 Nov;249(3):395–408. 10.1002/path.5325

42. Cui J, Placzek WJ. PTBP1 enhances miR-101-guided AGO2 targeting to MCL1 and promotes miR-101-induced apoptosis. Cell Death Dis. 2018 May;9(5):552. 10.1038/s41419-018-0551-8

43. Ding Y, Gao S, Zheng J, Chen X. Blocking lncRNA-SNHG16 sensitizes gastric cancer cells to 5-Fu through targeting the miR-506-3p-PTBP1-mediated glucose metabolism. Cancer Metab. 2022 Nov;10(1):20. 10.1186/s40170-022-00293-w.

44. Shan L, Liu S, Zhang Q, Zhou Q, Shang Y. Human bone marrow--mesenchymal stem cell-derived exosomal microRNA-188 reduces bronchial smooth muscle cell proliferation in asthma through suppressing the JARID2/Wnt/β-catenin axis. Cell Cycle. 2022 Feb;21(4):352–67. 10.1080/15384101.2021.2020432

45. Zou Y, Zhou Q, Zhang Y. MicroRNA-21 released from mast cells-derived extracellular vesicles drives asthma in mice by potentiating airway inflammation and oxidative stress. Am J Transl Res. 2021 Jul;13(7):7475–91.

46. He X, Sheng J, Yu W, Wang K, Zhu S, Liu Q. LncRNA MIR155HG promotes temozolomide resistance by activating the Wnt/ β-catenin pathway via binding to PTBP1 in glioma. Cell Mol Neurobiol. 2021 Aug;41(6):1271–84. 10.1007/s10571-020-00898-z

47. Yao J, Qin L, Miao S, Wang X, Wu X. Overexpression of miR-506 suppresses proliferation and promotes apoptosis of osteosarcoma cells by targeting astrocyte elevated gene-1. Oncol Lett. 2016 Sep;12(3):1840–48. 10.3892/ol.2016.4827