Open Access

Vitamin D3 alleviates cigarette smoke extract‑mediated epithelial‑mesenchymal transition and fibrogenesis by upregulating CC16 expression in bronchial epithelial cells

  • Authors:
    • Yajun Mao
    • Hong Feng
  • View Affiliations

  • Published online on: March 29, 2022     https://doi.org/10.3892/etm.2022.11284
  • Article Number: 357
  • Copyright: © Mao 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

Vitamin D3 supplementation has been previously reported to inhibit the occurrence and development of chronic obstructive pulmonary disease (COPD). However, the underlying mechanism remains unclear. Epithelial‑mesenchymal transition (EMT) and fibrogenesis have been associated with the development of COPD. The aim of the present study was to investigate the potential effects and mechanism of vitamin D3 in an in vitro model of cigarette smoke (CS)‑induced EMT and fibrosis, with specific focus on the role of club cell protein 16 (CC16). CS extract (CSE) at different concentrations (5, 10 and 20%) was used to treat 16‑HBE cells to induce EMT and fibrogenesis following which they were treated with vitamin D3. Subsequently, the 20% CSE group was selected for further experiments, where 16‑HBE cells were divided into the following five groups: The control group; the CSE group; the low‑dose vitamin D3 group (250 nM); the medium‑dose vitamin D3 group (500 nM); and the high‑dose vitamin D3 group (1,000 nM). Western blot analysis was used to detect the protein expression levels of the EMT‑related proteins E‑cadherin, N‑cadherin, Slug and α‑SMA, fibrogenesis‑related proteins collagen Ⅳ and fibronectin 1, proteins involved in the TGF‑β1/SMAD3 signaling pathway and CC16. Immunofluorescence was used to measure the protein expression levels of E‑cadherin, N‑cadherin and collagen Ⅳ. Specific CC16 knockdown was performed using short hairpin RNA transfection to investigate the role of CC16. The results of the present study found that vitamin D3 could increase the protein expression level of CC16 to inhibit the activation of the TGF‑β1/SMAD3 signaling pathway; thereby reducing the 20% increase in CSE‑induced EMT‑ and fibrogenesis‑related protein expression levels. Following CC16 knockdown, the inhibitory effects of vitamin D3 on EMT‑ and fibrogenesis‑related protein expression were partially reversed. To conclude, these results suggest that vitamin D3 can inhibit the protein expression levels of EMT‑ and fibrogenesis‑related proteins induced by CSE, at least partially through the function of CC16. These findings are expected to provide novel theoretical foundations and ideas for the pathogenesis and treatment of COPD.

Introduction

Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation to and from the lung, which has become a growing global health issue (1,2). In addition, air pollution and genetic factors are important risk factors for COPD (3-6). However, currently available therapeutic options for COPD, including methylxanthines, β2-agonists, anticholinergics, corticosteroids and phosphodiesterase 4 inhibitors, were not able to completely halt the progression of COPD pathogenesis nor reduce the mortality rate (7-9). Therefore, there remains to be an urgent demand to explore novel treatment strategies to prevent the development of COPD.

Although the pathogenesis of COPD has not been fully elucidated, it has been proposed that epithelial-mesenchymal transition (EMT) and fibrogenesis may be involved (10,11). During EMT, epithelial cells gradually lose their epithelial phenotype and acquire typical mesenchymal characteristics (12-15). These are characterized by enhanced mitotic ability and extracellular matrix production, accompanied by the upregulation of N-cadherin, slug and α-SMA expression and the downregulation of E-cadherin expression (12-15). The main pathological changes that occur during fibrosis are increases in fibrous connective tissue generation in the organ tissue, decreases in parenchymal cells (16). Continuous EMT and fibrosis progression can lead to the destruction of organ structure and functional decline (17). This serves to be one of the main mechanism of small airway narrowing and is now considered to be the most important mechanism of COPD progression (18). Epithelial cell fibrogenesis process is frequently accompanied with upregulation of fibrogenesis-related protein expression, such as collagen IV and fibronectin 1 (FN1) (19). Previous studies have provided evidence that EMT is active in the airway of smokers, particularly in patients with COPD who smoke, suggesting that smoking-induced EMT can contribute to the pathogenesis of COPD (15,20).

Patients with COPD and acute obstructive pulmonary disease were frequently found to have vitamin D deficiency (21). Vitamin D (calcitonin) deficiency was reported to be positively associated with the severity of COPD (21). In addition, patients with COPD or more severe disease were found to have lower serum vitamin D levels (22). A previous study has suggested that treatment with vitamin D in patients with COPD and vitamin D deficiency can reduce the risk of moderate-to-severe disease (23). Furthermore, a recent study on patients with COPD caused by COVID-19 showed that although vitamin D deficiency is not a determinant of disease severity, supplementation does confer a positive role in alleviation (24). Cigarette smoke extract (CSE) can inhibit vitamin D-induced vitamin D receptor (VDR) translocation (25). Mathyssen et al (26) previously documented that the use of vitamin D supplements may reduce CSE-mediated cellular inflammatory responses by upregulating cathelicidin expression in bronchial epithelial cells. However, the underlying molecular mechanism remain unknown (26). In another previous study, although vitamin D deficiency exacerbated pulmonary fibrosis and EMT (27), no significant associations between the two conditions could be found. Vitamin supplements have been found to delay the process of fibrosis (28). However, the role of vitamins in CSE-induced fibrogenesis in bronchial epithelial cells remains unclear.

Club cell protein 16 (CC16) is the most abundant protein in the bronchoalveolar lavage fluid (29,30). It is encoded by the secretoglobin family 1A member 1 gene and serves an anti-inflammatory and antioxidant role in vivo (29,30). Previous studies have shown that CC16 mainly exerts anti-inflammatory effects in smoke-exposed lungs, such that COPD has been associated with CC16 deficiency (31,32). Smokers and patients with COPD were found to exhibit reduced airway CC16 immunostaining, which decreased further with increasing COPD severity (31-33). Exposing mice to CSE was found to reduce the airway expression of CC16(34). Therefore, CC16 is becoming of interest as a target molecule for the treatment of COPD. CC16 has been associated with the occurrence and development of COPD, with the severity and prognosis of the disease (34). The dynamic changes in CC16 expression can be used to predict changes in the condition of patients with COPD and to evaluate the clinical outcome. Combined with the comprehensive analysis of other common clinical indicators, the length of the stay in hospital can be shortened as analyzed by linear correlation analysis and multiple linear regression analysis (33,35,36). Therefore, the present study hypothesized that CC16 is associated with lung fibrogenesis during COPD. However, the effects of vitamin D3 on the expression of CC16 after CSE exposure and its underlying molecular mechanism remain unclear.

In COPD, the bronchial epithelium is the first immune barrier triggered by cigarette smoke (37). The present study aimed to investigate the effect of vitamin D3 supplementation on EMT and fibrogenesis in bronchial epithelial cells after CSE treatment. In addition, the role of CC16 in these processes and the expression of EMT and fibrogenesis-related markers were detected.

Materials and methods

Bioinformatics Methods

The interaction between CC16 and FN1 was analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database (version 11.5; https://string-db.org/cgi/input.pl).

Bronchial epithelial cell culture

The 16-HBE cells were purchased from the American Type Culture Collection and were cultured in DMEM (Sigma-Aldrich; Merck KGaA) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin G sodium and 100 µg/ml streptomycin sulfate (Invitrogen; Thermo Fisher Scientific, Inc.). They were cultured at 37˚C in a humidified incubator with 5% CO2. The cells were cultured until they reached 70-80% confluence.

CC16 short hairpin (sh) RNA

The pLentiLox 3.7 lentiviral plasmid shRNAs targeting CC16 and scramble control shRNA were purchased from Hanbio Biotechnology Co., Ltd. The cells were seeded into a 96-well plate for 24 h at 37˚C until 70-80% confluence, before the cells were transfected with the shRNAs (50 nM) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. The efficiency of transfection was determined using reverse transcription-quantitative PCR (RT-qPCR) and western blot analyzes after 48 h of transfection. The sequences were as follows: ShRNA-CC16-1, sense 5'-CCAGAGAAAGCATCATTAA-3', antisense 5'-TTAATGATGCTTTCTCTGG-3'; shRNA-CC16-2, sense 5'-CCCAAAGCTCACTGTGTAA-3', antisense 5'-TTACACAGTGAGCTTTGGG-3'; shRNA-NC, sense 5'-GATCCCCTTCTCCGAACG-3', antisense 5'-AGCTAAAAATTCTCCGAAC-3'.

Preparation of CSE

The smoke from 15 lit cigarettes (Sichuan China Tobacco Industry Co., Ltd.) was slowly inhaled into a 50 ml syringe, and then injected it into DMEM pre-heated in a 37˚C water bath (38). The pH of DMEM was adjusted to 7.4 and sterilized using a 0.22-µm filter (EMD Millipore) and stored at -80˚C. Serum-free DMEM was used to dilute 100% CSE to the required CSE concentrations (5, 10 and 20%). Cells were treated with vitamin D3 (250, 500 or 1,000 nM; Sigma-Aldrich; Merck KGaA) or vehicle (0.1% ethanol) for 30 min prior to 24 h treatment at 37˚C with CSE and vitamin D3 or vehicle, after shRNA transfection (when it was required) (26).

Cell Counting Kit (CCK)-8 assay

Cell viability was performed using CCK-8 assay. A total of 1x104 16-HBE cells/well were seeded into 96-well plates and pre-treated with CSE followed by vitamin D3. In total, 10 µl CCK-8 reagent (Abcam) was added into each well and the samples were incubated at 37˚C for 1-4 h. Subsequently, the absorbance in each well was measured at 460 nm using a microplate spectrophotometer (Bio-Rad Laboratories, Inc.).

RT-qPCR

Total RNA was extracted from the cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). The concentration was assessed using a Nanodrop® 2000 spectrophotometer (Thermo Fisher Scientific, Inc.) before being reverse transcribed into cDNA. cDNA was synthesized using the PrimeScript™ RT kit (cat. no. RR037A; Takara Bio, Inc.) under the following conditions: 15 min at 37˚C and 5 sec at 85˚C. The reactions were performed using SYBR Green Taq Mix (cat. no. RR096A; Takara Bio, Inc.) in a StepOnePlus™ PCR system (Thermo Fisher Scientific, Inc.) under the following conditions: 45˚C for 3 min, 95˚C for 10 sec, 40 cycles at 95˚C for 15 sec then 58˚C for 1 min. The primer sequences used were as follows: CC16 forward, 5'-CAGAGACGGAACCAGAGACG-3' and reverse, 5'-CAGATCTCTGCAGAAGCGGA-3' and GAPDH forward, 5'-GGAGCGAGATCCCTCCAAAAT-3' and reverse, 5'-GGCTGTTGTCATACTTCTCATGG-3'. Gene expression was evaluated using the 2-ΔΔCq method (39) using GAPDH as a reference gene.

Western blot analysis

The total protein was extracted from the cells using RIPA lysate buffer (Beyotime Institute of Biotechnology) containing protease inhibitors, phosphatase inhibitors and phenylmethylsulfonyl fluoride (all from Beyotime Institute of Biotechnology). The protein concentration was determined using a BCA protein determination kit (Beyotime Institute of Biotechnology). A total of 30 µg protein per lane extract was separated using 6-12% SDS-PAGE and transferred onto PVDF membranes. Subsequently, the PVDF membranes were blocked with TBS containing 5% BSA (Sigma-Aldrich; Merck KGaA) for 2 h at room temperature, before being incubated overnight at 4˚C with the following primary antibodies: E-cadherin (1:1,000; cat. no. 14472; Cell Signaling Technology, Inc.), N-cadherin (1:1,000; cat. no. 13116; Cell Signaling Technology, Inc.), slug (1:1,000; cat. no. 9585; Cell Signaling Technology, Inc.), α-smooth muscle actin (1:2,000; α-SMA; cat. no. NBP2-78836; Novus Biologicals, LLC), collagen Ⅳ (1:1,000; cat. no. ab6586; Abcam), FN1 (1:1,000; cat. no. 26836; Cell Signaling Technology, Inc.), TGF-β1 (1:2,000; cat. no. sc-130348; Santa Cruz Biotechnology, Inc.), SMAD3 (1:1,000; cat. no. 9523; Cell Signaling Technology, Inc.), phosphorylated (p-) SMAD3 (1:1,000; cat. no. 9520; Cell Signaling Technology, Inc.) and CC16 (5 µg/ml; cat. no. RD181022220-01; BioVender). The membranes were then incubated with the goat anti-mouse IgG H&L HRP-conjugated secondary antibodies (1:20,000; cat. no. ab205719; Abcam) and goat anti-rabbit IgG H&L (1:50,000; cat. no. ab205718; Abcam) at room temperature for 2 h. Afterwards, they were washed for 25 min with TBS-0.1% Tween-20. After using an enhanced chemiluminescence reagent (Thermo Fisher Scientific, Inc.), the protein bands were detected using a Bio-Rad Imaging system (Bio-Rad Laboratories, Inc.) and analyzed using ImageJ software (version 1.43; National Institutes of Health).

Immunofluorescence staining

The cells were fixed in 4% paraformaldehyde (Beyotime Institute of Biotechnology) for 1 h at room temperature, washed in PBS and blocked for 15 min in QuickBlock™ Blocking buffer for immunol staining (Beyotime Institute of Biotechnology) at room temperature. The samples were then incubated with primary antibodies overnight at 4˚C. The following primary antibodies used targeted E-cadherin (1:200; cat. no. 14472; Cell Signaling Technology, Inc.), N-cadherin (1:200; cat. no. 13116; Cell Signaling Technology, Inc.), collagen Ⅳ (1:500; cat. no. ab6586; Abcam). Subsequently, the samples were incubated with corresponding Alexa Fluor® 488-conjugated secondary antibodies (Goat Anti-Rabbit IgG H&L; 1:1,000; cat. no. 150077; Abcam and Goat Anti-Mouse IgG H&L; 1:1,000; cat. no. 150113; Abcam) for 1 h at room temperature after washing with PBS. Then these samples were incubated for 5 min at room temperature with DAPI (1 µg/ml; cat. no. D1306; Invitrogen; Thermo Fisher Scientific, Inc.), and observations were performed using a fluorescence microscope (magnification, x100; Olympus Corporation) and were analyzed with ImageJ version 1.43 software.

Statistical analysis

Statistical analysis was performed using GraphPad 8.0 (GraphPad Software, Inc.). Data were expressed as means ± SEM of three independent experiments. Comparisons were performed using one-way analysis of variance with Tukey-Kramer post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

CES reduces cell viability and induces EMT

After treating the 16-HBE cells with different concentrations of CSE (5, 10 and 20%) for 24 h, cell viability was decreased in a dose-dependent manner, comparing with that in the control group (Fig. 1A). CSE at a concentration of 20% was used for subsequent experiments. Western blot analysis revealed that the protein expression level of E-cadherin was decreased, whilst the protein expression levels of N-cadherin, slug and α-SMA were increased, in a dose-dependent manner following CSE treatment (Fig. 1B and C). The results of immunofluorescence were consistent with those in the western blot analysis results, revealing a decrease in E-cadherin protein expression and an increase in N-cadherin protein expression compared with those in the control group (Fig. 1D). These results suggest that CSE promoted the transformation of these 16-HBE epithelial cells into mesenchymal-like cells.

CSE induces fibrogenesis in 16-HBE cells

It was first found that CC16 interacted with FN1, which is associated with fibrogenesis (40), from analysis using the STRING database (Fig. 2A). Therefore, western blot analysis was used to measure the protein expression level of fibrogenesis-related proteins. Compared with that in the control group, the protein expression level of CC16 in 16-HBE cells were decreased following CSE treatment. Furthermore, the protein expression levels of the fibrogenesis-related proteins collagen IV and FN1, in addition to those in the TGF-β1/SMAD3 signaling pathway, were increased in the CSE treatment groups in a dose-dependent manner compared with that in the control group (Fig. 2B). Immunofluorescence results also showed that the expression level of collagen IV was also increased in the CSE treatment group compared with that in the control group (Fig. 2C). In summary, these results suggest that CSE induced fibrogenesis in the 16-HBE cell line.

Supplementation of vitamin D3 inhibits EMT caused by CSE in the 16-HBE cells

Vitamin D3 treatment did not appear to cause toxicity to 16-HBE cells treated with 20% CSE, instead increasing cell viability (Fig. 3A). Western blot analysis found that the protein expression level of E-cadherin was increased, whilst the protein expression levels of N-cadherin and slug were decreased following supplementation with vitamin D3, compared with those in the 20% CSE treatment alone group for 24 h (Fig. 3B and C). The results from immunofluorescence were consistent with those in western blot analysis (Fig. 3C), suggesting that vitamin D3 supplementation can reverse EMT induced by CSE.

Vitamin D3 supplementation inhibits fibrogenesis caused by CSE in the 16-HBE cell line

Compared with that in the CSE alone group, the protein expression level of CC16 was increased following supplementation with vitamin D3. Western blot analysis was used to also measure the protein expression level of fibrogenesis-related proteins. The results showed that compared with that in the CSE alone group, the protein expression level of fibrogenesis-related proteins collagen IV, FN1, TGF-β1 and SMAD phosphorylation were decreased following administration with vitamin D3 (Fig. 4A). Immunofluorescence results showed that compared with that in the CSE alone group, the protein expression level of collagen IV was inhibited following supplementation with vitamin D3 (Fig. 4B). These results suggest that vitamin D3 can inhibit the CSE-induced fibrogenesis of bronchial epithelial cells (Fig. 4B).

Inhibitory effects of vitamin D3 on EMT and fibrogenesis is weakened following transfection with shRNAs targeting CC16 in the 16-HBE cells

To assess the role of CC16 in fibrogenesis and EMT, specific shRNAs were used to inhibit the expression level of CC16 in the 16-HBE cell line. Compared with that in the scrambled shRNA control group, both shRNA-CC16-1 and shRNA-CC16-2 significantly reduced CC16 expression in the 16-HBE cell line, with shRNA-CC16-1 being more potent (Fig. 5A). According to western blot analysis, CC16 knockdown markedly reduced the protein expression level of E-cadherin whilst promoting the protein expression levels of N-cadherin, slug and α-SMA, compared with those in the vitamin D3 treatment group (Fig. 5B). Subsequent western blot analysis revealed that knocking down CC16 expression led to significant increases in the protein expression levels of fibrogenesis-related proteins compared with those in the vitamin D3 treatment group (Fig. 5C). These results suggest that after transfection with shRNA-CC16-1, the inhibitory effects of vitamin D3 on EMT and fibrogenesis were reversed in the 16-HBE cell line. In addition, these suppressive effects of vitamin D3 on CSE-induced EMT and fibrogenesis was at least partially mediated by CC16.

Discussion

COPD is a type of chronic bronchitis and/or emphysema with the pathological characteristics of airflow obstruction, which can further develop into common chronic diseases such as pulmonary heart disease and respiratory failure (41). COPD is associated with aberrant inflammatory reactions caused by harmful gases and harmful particles (42). Among these harmful factors, cigarette smoke considered to be one of the most common harmful particles that can cause COPD (43). Metal substances (such as Arsenic, cadmium and lead) and tobacco derivatives contained within cigarette smoke increases the risk of lung adenocarcinoma, such that ~6 million deaths are caused by tobacco use every year worldwide (44-46). Previous studies have shown that smoking contributes to lung remodeling prior to COPD, where CSE induces bronchial epithelial damage by increasing the susceptibility to respiratory infections (47-49). During this process, cells tend to undergo EMT and anchor-independent growth (50). In the present study, 16-HBE cells exposed to CSE exhibited EMT characteristics in a dose-dependent manner, with the highest N-cadherin, slug and α-SMA protein expression levels and the lowest E-cadherin protein expression levels being observed following 20% CSE exposure. Previous studies have found signs of peribronchiolar fibrosis in the small airways of patients with COPD, which EMT appearing to be involved in this process (47,51). The present study observed that the expression of fibrogenesis-related proteins collagen IV and FN1 were significantly upregulated in 16-HBE cells after being treated with 20% CSE for 24 h. These results suggest that CSE can induce EMT and fibrogenesis in bronchial epithelial cells.

According to analysis using the STRING database, CC16 is associated with the fibrosis-related protein FN1. CC16 is a major protein that is secreted by club cells in the bronchial epithelium and is eliminated by the kidneys (30). After observing this association between CC16 and fibrogenesis, the expression levels of CC16 was measured after CSE exposure. Previous study reported that CC16 can exert anti-inflammatory properties in smoke-exposed lungs of mice and proposed that COPD may be associated with CC16 deficiency (33). In the present study, downregulation of CC16 expression in CSE treated 16-HBE cells was also observed.

Over the past decade, a number of studies have shown that vitamins serve an important role in COPD (52-54). Vitamin A is essential for the preservation and integrity of the lung epithelium (55). Compared with that in the healthy group, the levels and intake of vitamin A in patients with COPD was significantly reduced whilst serum vitamin A levels were also found to be positively associated with the severity of COPD (56). In a previous clinical study that involved Korean patients with COPD, vitamin C was found to exert protective effects against COPD (57). In addition, another previous study hypothesized that the protective effects of vitamin C against COPD was due to its antioxidant effects (52). Vitamin B12, vitamin E and vitamin K were also found to serve key roles in inhibiting the occurrence and development of COPD (58-61). However, accumulating evidence has found that vitamin D, which is a class of fat-soluble vitamins that includes vitamin D3 and D2, can exert a role in COPD (62,63). Its reported physiological effects include regulation of calcium and phosphorus metabolism, cell proliferation and differentiation, immune regulation and promotion of bone growth (64). Therefore, vitamin D has been of interest for the potential treatment and prevention of various diseases, such as chronic kidney disease and cystic fibrosis (65-67). In a previous in vivo study, vitamin D3 supplementation was found to alleviate lung injury in rats with COPD whilst also reducing cell apoptosis in the lung tissues (68). Clinical studies have also shown that vitamin D3 supplementation can alleviate moderate or severe COPD to reduce the incidence of upper respiratory tract infections (23-69). Furthermore, CSE has been reported to inhibit vitamin D-induced VDR translocation (25), but the vitamin D3/VDR axis can inhibit emphysema in patients with COPD (70). However, the specific mechanism of the protective effects of vitamin D3 against COPD remains unclear. Therefore, the present study investigated the effects of vitamin D3 on CSE-induced EMT and fibrogenesis in 16-HBE cells.

A previous study found that combining aerobic exercise with vitamin D3 supplementation can upregulate the levels of serum CC16 in patients with lung injury caused by smoking (71). However, the specific mechanism was not elucidated (71). Compared with the findings reported by Mathyssen et al (26), this previous study found no clear association between vitamin D3 supplementation and the degree of EMT of bronchial epithelial cells (71). In the present study, EMT and fibrogenesis was induced in 16-HBE cells following exposure to CSE, which were alleviated by vitamin D3 in a dose-dependent manner. In addition, to verify the association between Vitamin D3 and CC16 in this process, CC16 shRNA was used to knock down its expression. The results showed that after CC16 expression was reduced, the protective effects of vitamin D3 on 16-HBE cells were significantly reversed, with EMT and fibrogenesis restored. This suggests that vitamin D3 can mediate protective effects by increasing the protein expression of CC16. This provides a novel mechanism to further the understanding of the protective role of vitamin D3 against COPD. However, further investigation is required.

During EMT, the TGF-β1/SMAD signaling pathway serves an important role (72). A previous study has shown that smoking can increase TGF-β1 production (73). The levels of EMT and TGF-β in the airway epithelial cells of patients with COPD were associated with the severity of peribronchial fibrosis and airway obstruction (74). In the present study, upregulation of TGF-β1 expression was observed in CSE-treated 16-HBE cells. A previous study also suggested that the TGF-β1/SMAD2/3 pathway can be a potential therapeutic target for EMT in malignant tumor cells derived from epithelial cells (75). Compared with that in heathy individuals, the expression level of TGF-β1 and its downstream signal, SMAD2/3 was found to be significantly increased in the airways of patients with COPD (73). The present study showed that SMAD3 was activated in 16-HBE cells exposed to CSE. After treating the cells with vitamin D3, the expression of TGF-β1 and the activation of SMAD3 was partially inhibited whilst the expression of CC16 was upregulated.

Bronchial epithelial cells were used for the present study. However, in vivo models must be applied for verification. In addition, it is of importance to investigate further the exact mechanistic relationship between vitamin D3 and CC16.

In conclusion, vitamin D3 supplementation could inhibit CSE-mediated EMT and fibrogenesis by increasing the protein expression levels of CC16. After CC16 expression was knocked down using shRNA, the inhibitory effects of vitamin D3 on EMT and fibrogenesis was reversed, suggesting that vitamin D3 protected the bronchial epithelial cells by inhibiting EMT and fibrogenesis. To the best of our knowledge, the present study was the first to suggest that vitamin D3 can serve a inhibitory role in EMT and fibrogenesis by regulating CC16. This may furthering the understanding of the mechanism underlying the effects exerted by vitamin D3 supplementation in preventing the development of COPD.

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

HF guided the project, analyzed the data and wrote the manuscript. YM conceived the technical details and designed the experiments. HF and YM performed the experiments. YM and HF confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al: Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the global burden of disease study 2010. Lancet. 380:2095–2128. 2012.PubMed/NCBI View Article : Google Scholar

2 

Pauwels RA and Rabe KF: Burden and clinical features of chronic obstructive pulmonary disease (COPD). Lancet. 364:613–620. 2004.PubMed/NCBI View Article : Google Scholar

3 

Churg A, Brauer M, del Carmen Avila-Casado M, Fortoul TI and Wright JL: Chronic exposure to high levels of particulate air pollution and small airway remodeling. Environ Health Perspect. 111:714–718. 2003.PubMed/NCBI View Article : Google Scholar

4 

Brakema EA, Tabyshova A, Kasteleyn MJ, Molendijk E, van der Kleij RMJJ, van Boven JFM, Emilov B, Akmatalieva M, Mademilov M, Numans ME, et al: High COPD prevalence at high altitude: Does household air pollution play a role? Eur Respir J. 53(1801193)2018.PubMed/NCBI View Article : Google Scholar

5 

Hall R, Hall IP and Sayers I: Genetic risk factors for the development of pulmonary disease identified by genome-wide association. Respirology. 24:204–214. 2019.PubMed/NCBI View Article : Google Scholar

6 

Barrera C, Rocchi S, Degano B, Soumagne T, Laurent L, Bellanger AP, Laplante JJ, Millon L, Dalphin JC and Reboux G: Microbial exposure to dairy farmers' dwellings and COPD occurrence. Int J Environ Health Res. 29:387–399. 2019.PubMed/NCBI View Article : Google Scholar

7 

Vasques F, Camporota L and Barrett NA: Nonantibiotic pharmacological treatment of severe chronic obstructive pulmonary disease exacerbations. Semin Respir Crit Care Med. 41:842–850. 2020.PubMed/NCBI View Article : Google Scholar

8 

Kim V and Criner GJ: Chronic bronchitis and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 187:228–237. 2013.PubMed/NCBI View Article : Google Scholar

9 

Matera MG, Calzetta L, Rogliani P, Cesario A and Cazzola M: New treatments for COPD in the elderly. Curr Pharm Des. 20:5968–5982. 2014.PubMed/NCBI View Article : Google Scholar

10 

Eapen MS, Sharma P, Thompson IE, Lu W, Myers S, Hansbro PM and Sohal SS: Heparin-binding epidermal growth factor (HB-EGF) drives EMT in patients with COPD: Implications for disease pathogenesis and novel therapies. Lab Invest. 99:150–157. 2019.PubMed/NCBI View Article : Google Scholar

11 

Barnes PJ: Small airway fibrosis in COPD. Int J Biochem Cell Biol. 116(105598)2019.PubMed/NCBI View Article : Google Scholar

12 

Clapéron A, Mergey M, Ho-Bouldoires TH, Vignjevic D, Wendum D, Chrétien Y, Merabtene F, Frazao A, Paradis V, Housset C, et al: EGF/EGFR axis contributes to the progression of cholangiocarcinoma through the induction of an epithelial-mesenchymal transition. J Hepatol. 61:325–332. 2014.PubMed/NCBI View Article : Google Scholar

13 

Han YT, Chen XH, Gao H, Ye JL and Wang CB: Physcion inhibits the metastatic potential of human colorectal cancer SW620 cells in vitro by suppressing the transcription factor SOX2. Acta Pharmacol Sin. 37:264–275. 2016.PubMed/NCBI View Article : Google Scholar

14 

Kannan A, Krishnan A, Ali M, Subramaniam S, Halagowder D and Sivasithamparam ND: Caveolin-1 promotes gastric cancer progression by up-regulating epithelial to mesenchymal transition by crosstalk of signalling mechanisms under hypoxic condition. Eur J Cancer. 50:204–215. 2014.PubMed/NCBI View Article : Google Scholar

15 

Hou W, Hu S, Li C, Ma H, Wang Q, Meng G, Guo T and Zhang J: Cigarette smoke induced lung barrier dysfunction, EMT, and tissue remodeling: A possible link between COPD and lung cancer. Biomed Res Int. 2019(2025636)2019.PubMed/NCBI View Article : Google Scholar

16 

Nicholson AG, Colby TV and Wells AU: Histopathological approach to patterns of interstitial pneumonia in patient with connective tissue disorders. Sarcoidosis Vasc Diffuse Lung Dis. 19:10–17. 2002.PubMed/NCBI

17 

Wynn TA and Ramalingam TR: Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat Med. 18:1028–1040. 2012.PubMed/NCBI View Article : Google Scholar

18 

Hogg JC, Pare PD and Hackett TL: The contribution of small airway obstruction to the pathogenesis of chronic obstructive pulmonary disease. Physiol Rev. 97:529–552. 2017.PubMed/NCBI View Article : Google Scholar

19 

Kyung SY, Kim DY, Yoon JY, Son ES, Kim YJ, Park JW and Jeong SH: Sulforaphane attenuates pulmonary fibrosis by inhibiting the epithelial-mesenchymal transition. BMC Pharmacol Toxicol. 19(13)2018.PubMed/NCBI View Article : Google Scholar

20 

Sohal SS, Mahmood MQ and Walters EH: Clinical significance of epithelial mesenchymal transition (EMT) in chronic obstructive pulmonary disease (COPD): Potential target for prevention of airway fibrosis and lung cancer. Clin Transl Med. 3(33)2014.PubMed/NCBI View Article : Google Scholar

21 

Islam S, Sarkar NK, Mujahid AA, Bennoor KS, Hossain SS, Attar MM, Jahan R, Hossain MA, Chowdhury HA and Ali L: Association of serum vitamin D (25OHD) level with acute exacerbation of chronic obstructive pulmonary disease. Mymensingh Med J. 28:441–448. 2019.PubMed/NCBI

22 

Baneen U and Naseem S: Correlation of severity of chronic obstructive pulmonary disease with serum vitamin-D level. J Family Med Prim Care. 8:2268–2277. 2019.PubMed/NCBI View Article : Google Scholar

23 

Martineau AR, James WY, Hooper RL, Barnes NC, Jolliffe DA, Greiller CL, Islam K, McLaughlin D, Bhowmik A, Timms PM, et al: Vitamin D3 supplementation in patients with chronic obstructive pulmonary disease (ViDiCO): A multicentre, double-blind, randomised controlled trial. Lancet Respir Med. 3:120–130. 2015.PubMed/NCBI View Article : Google Scholar

24 

Chaabouni M, Feki W, Chaabouni K and Kammoun S: Vitamin D supplementation to prevent COVID-19 in patients with COPD: A research perspective. Adv Respir Med. 88:364–365. 2020.PubMed/NCBI View Article : Google Scholar

25 

Uh ST, Koo SM, Kim YK, Kim KU, Park SW, Jang AS, Kim DJ, Kim YH and Park CS: Inhibition of vitamin d receptor translocation by cigarette smoking extracts. Tuberc Respir Dis (Seoul). 73:258–265. 2012.PubMed/NCBI View Article : Google Scholar

26 

Mathyssen C, Serre J, Sacreas A, Everaerts S, Maes K, Verleden S, Verlinden L, Verstuyf A, Pilette C, Gayan-Ramirez G, et al: Vitamin D modulates the response of bronchial epithelial cells exposed to cigarette smoke extract. Nutrients. 11(2138)2019.PubMed/NCBI View Article : Google Scholar

27 

Li SR, Tan ZX, Chen YH, Hu B, Zhang C, Wang H, Zhao H and Xu DX: Vitamin D deficiency exacerbates bleomycin-induced pulmonary fibrosis partially through aggravating TGF-β/smad2/3-mediated epithelial-mesenchymal transition. Respir Res. 20(266)2019.PubMed/NCBI View Article : Google Scholar

28 

Tzilas V, Bouros E, Barbayianni I, Karampitsakos T, Kourtidou S, Ntassiou M, Ninou I, Aidinis V, Bouros D and Tzouvelekis A: Vitamin D prevents experimental lung fibrosis and predicts survival in patients with idiopathic pulmonary fibrosis. Pulm Pharmacol Ther. 55:17–24. 2019.PubMed/NCBI View Article : Google Scholar

29 

Almuntashiri S, Zhu Y, Han Y, Wang X, Somanath PR and Zhang D: Club cell secreted protein cc16: Potential applications in prognosis and therapy for pulmonary diseases. J Clin Med. 9(4039)2020.PubMed/NCBI View Article : Google Scholar

30 

Egron C, Labbé A, Rochette E, Mulliez A, Bernard A and Flore A: Urinary club cell protein 16 (CC16): Utility of its assay during acute bronchiolitis. Pediatr Pulmonol. 55:490–495. 2020.PubMed/NCBI View Article : Google Scholar

31 

Pang M, Liu HY, Li T, Wang D, Hu XY, Zhang XR, Yu BF, Guo R and Wang HL: Recombinant club cell protein 16 (CC16) ameliorates cigarette smokeinduced lung inflammation in a murine disease model of COPD. Mol Med Rep. 18:2198–2206. 2018.PubMed/NCBI View Article : Google Scholar

32 

Zhu L, Di PY, Wu R, Pinkerton KE and Chen Y: Repression of CC16 by cigarette smoke (CS) exposure. PLoS One. 10(e0116159)2015.PubMed/NCBI View Article : Google Scholar

33 

Laucho-Contreras ME, Polverino F, Tesfaigzi Y, Pilon A, Celli BR and Owen CA: Club cell protein 16 (CC16) augmentation: A potential disease-modifying approach for chronic obstructive pulmonary disease (COPD). Expert Opin Ther Targets. 20:869–883. 2016.PubMed/NCBI View Article : Google Scholar

34 

Laucho-Contreras ME, Polverino F, Gupta K, Taylor KL, Kelly E, Pinto-Plata V, Divo M, Ashfaq N, Petersen H, Stripp B, et al: Protective role for club cell secretory protein-16 (CC16) in the development of COPD. Eur Respir J. 45:1544–1556. 2015.PubMed/NCBI View Article : Google Scholar

35 

Rong B, Fu T, Gao W, Li M, Rong C, Liu W and Liu H: Reduced serum concentration of CC16 is associated with severity of chronic obstructive pulmonary disease and contributes to the diagnosis and assessment of the disease. Int J Chron Obstruct Pulmon Dis. 15:461–470. 2020.PubMed/NCBI View Article : Google Scholar

36 

Zemans RL, Jacobson S, Keene J, Kechris K, Miller BE, Tal-Singer R and Bowler RP: Multiple biomarkers predict disease severity, progression and mortality in COPD. Respir Res. 18(117)2017.PubMed/NCBI View Article : Google Scholar

37 

Danov O, Wolff M, Bartel S, Böhlen S, Obernolte H, Wronski S, Jonigk D, Hammer B, Kovacevic D, Reuter S, et al: Cigarette smoke affects dendritic cell populations, epithelial barrier function, and the immune response to viral infection with H1N1. Front Med (Lausanne). 7(571003)2020.PubMed/NCBI View Article : Google Scholar

38 

Li D, Hu J, Wang T, Zhang X, Liu L, Wang H, Wu Y, Xu D and Wen F: Silymarin attenuates cigarette smoke extract-induced inflammation via simultaneous inhibition of autophagy and ERK/p38 MAPK pathway in human bronchial epithelial cells. Sci Rep. 6(37751)2016.PubMed/NCBI View Article : Google Scholar

39 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

40 

Miguel V, Ramos R, García-Bermejo L, Rodríguez-Puyol D and Lamas S: The program of renal fibrogenesis is controlled by microRNAs regulating oxidative metabolism. Redox Biol. 40(101851)2021.PubMed/NCBI View Article : Google Scholar

41 

López-Campos JL, Tan W and Soriano JB: Global burden of COPD. Respirology. 21:14–23. 2016.PubMed/NCBI View Article : Google Scholar

42 

Raherison C and Girodet PO: Epidemiology of COPD. Eur Respir Rev. 18:213–221. 2009.PubMed/NCBI View Article : Google Scholar

43 

Vij N, Chandramani-Shivalingappa P, Van Westphal C, Hole R and Bodas M: Cigarette smoke-induced autophagy impairment accelerates lung aging, COPD-emphysema exacerbations and pathogenesis. Am J Physiol Cell Physiol. 314:C73–C87. 2018.PubMed/NCBI View Article : Google Scholar

44 

Pinto E, Cruz M, Ramos P, Santos A and Almeida A: Metals transfer from tobacco to cigarette smoke: Evidences in smokers' lung tissue. J Hazard Mater. 325:31–35. 2017.PubMed/NCBI View Article : Google Scholar

45 

Paumgartten FJR, Gomes-Carneiro MR and de Oliveira AC: The impact of tobacco additives on cigarette smoke toxicity: A critical appraisal of tobacco industry studies. Cad Saude Publica. 33 (Suppl 3)(e00132415)2017.PubMed/NCBI View Article : Google Scholar

46 

D'Angelo D, Ahluwalia IB, Pun E, Yin S, Palipudi K and Mbulo L: Current cigarette smoking, access, and purchases from retail outlets among students aged 13-15 years-global youth tobacco survey, 45 countries, 2013 and 2014. MMWR Morb Mortal Wkly Rep. 65:898–901. 2016.PubMed/NCBI View Article : Google Scholar

47 

Xu H, Ling M, Xue J, Dai X, Sun Q, Chen C, Liu Y, Zhou L, Liu J, Luo F, et al: Exosomal microRNA-21 derived from bronchial epithelial cells is involved in aberrant epithelium-fibroblast cross-talk in COPD induced by cigarette smoking. Theranostics. 8:5419–5433. 2018.PubMed/NCBI View Article : Google Scholar

48 

Amatngalim GD, Schrumpf JA, Dishchekenian F, Mertens TC, Ninaber DK, van der Linden AC, Pilette C, Taube C, Hiemstra PS, van der Does AM, et al: Aberrant epithelial differentiation by cigarette smoke dysregulates respiratory host defence. Eur Respir J. 51(1701009)2018.PubMed/NCBI View Article : Google Scholar

49 

Willey JC, Grafstrom RC, Moser CE Jr, Ozanne C, Sundquvist K and Harris CC: Biochemical and morphological effects of cigarette smoke condensate and its fractions on normal human bronchial epithelial cells in vitro. Cancer Res. 47:2045–2049. 1987.PubMed/NCBI

50 

Vaz M, Hwang SY, Kagiampakis I, Phallen J, Patil A, O'Hagan HM, Murphy L, Zahnow CA, Gabrielson E, Velculescu VE, et al: Chronic cigarette smoke-induced epigenomic changes precede sensitization of bronchial epithelial cells to single-step transformation by KRAS mutations. Cancer Cell. 32:360–376. 2017.PubMed/NCBI View Article : Google Scholar

51 

Milara J, Peiró T, Serrano A and Cortijo J: Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax. 68:410–420. 2013.PubMed/NCBI View Article : Google Scholar

52 

Pirabbasi E, Shahar S, Manaf ZA, Rajab NF and Manap RA: Efficacy of ascorbic acid (Vitamin C) and/N-Acetylcysteine (NAC) supplementation on nutritional and antioxidant status of male chronic obstructive pulmonary disease (COPD) patients. J Nutr Sci Vitaminol (Tokyo). 62:54–61. 2016.PubMed/NCBI View Article : Google Scholar

53 

Piscaer I, Wouters EFM, Vermeer C, Janssens W, Franssen FME and Janssen R: Vitamin K deficiency: The linking pin between COPD and cardiovascular diseases? Respir Res. 18(189)2017.PubMed/NCBI View Article : Google Scholar

54 

Zhu M, Wang T, Wang C and Ji Y: The association between vitamin D and COPD risk, severity, and exacerbation: An updated systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis. 11:2597–2607. 2016.PubMed/NCBI View Article : Google Scholar

55 

Biesalski HK and Nohr D: Importance of vitamin-A for lung function and development. Mol Aspects Med. 24:431–440. 2003.PubMed/NCBI View Article : Google Scholar

56 

Caram LM, Amaral RA, Ferrari R, Tanni SE, Correa CR, Paiva SAR and Godoy I: Serum vitamin A and inflammatory markers in individuals with and without chronic obstructive pulmonary disease. Mediators Inflamm. 2015(862086)2015.PubMed/NCBI View Article : Google Scholar

57 

Park HJ, Byun MK, Kim HJ, Kim JY, Kim YI, Yoo KH, Chun EM, Jung JY, Lee SH and Ahn CM: Dietary vitamin C intake protects against COPD: The Korea national health and nutrition examination survey in 2012. Int J Chron Obstruct Pulmon Dis. 11:2721–2728. 2016.PubMed/NCBI View Article : Google Scholar

58 

Liu JT, Luo B, He XT, Li LY and Xu SG: The protective effects of vitamin E on lung injury caused by high temperature and PM2.5 in COPD rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 35:293–296. 2019.PubMed/NCBI View Article : Google Scholar : (In Chinese).

59 

Peh HY, Tan WSD, Chan TK, Pow CW, Foster PS and Wong WSF: Vitamin E isoform gamma-tocotrienol protects against emphysema in cigarette smoke-induced COPD. Free Radic Biol Med. 110:332–344. 2017.PubMed/NCBI View Article : Google Scholar

60 

Paulin FV, Zagatto AM, Chiappa GR and Muller PT: Addition of vitamin B12 to exercise training improves cycle ergometer endurance in advanced COPD patients: A randomized and controlled study. Respir Med. 122:23–29. 2017.PubMed/NCBI View Article : Google Scholar

61 

Piscaer I, van den Ouweland JMW, Vermeersch K, Reynaert NL, Franssen FM, Keene S, Wouters EF, Janssens W, Vermeer C and Janssen R: Low vitamin K status is associated with increased elastin degradation in chronic obstructive pulmonary disease. J Clin Med. 8(1116)2019.PubMed/NCBI View Article : Google Scholar

62 

Jolliffe DA, Greenberg L, Hooper RL, Mathyssen C, Rafiq R, de Jongh RT, Camargo CA, Griffiths CJ, Janssens W and Martineau AR: Vitamin D to prevent exacerbations of COPD: systematic review and meta-analysis of individual participant data from randomised controlled trials. Thorax. 74:337–345. 2019.PubMed/NCBI View Article : Google Scholar

63 

Milne S and Sin DD: Vitamin D deficiency in COPD: Biomarker, treatable trait, or just a common comorbidity? Chest. 157:755–756. 2020.PubMed/NCBI View Article : Google Scholar

64 

Gil Á, Plaza-Diaz J and Mesa MD: Vitamin D: Classic and novel actions. Ann Nutr Metab. 72:87–95. 2018.PubMed/NCBI View Article : Google Scholar

65 

Chau YY and Kumar J: Vitamin D in chronic kidney disease. Indian J Pediatr. 79:1062–1068. 2012.PubMed/NCBI View Article : Google Scholar

66 

Chesdachai S and Tangpricha V: Treatment of vitamin D deficiency in cystic fibrosis. J Steroid Biochem Mol Biol. 164:36–39. 2016.PubMed/NCBI View Article : Google Scholar

67 

Tripkovic L, Lambert H, Hart K, Smith CP, Bucca G, Penson S, Chope G, Hyppönen E, Berry J, Vieth R and Lanham-New S: Comparison of vitamin D2 and vitamin D3 supplementation in raising serum 25-hydroxyvitamin D status: A systematic review and meta-analysis. Am J Clin Nutr. 95:1357–1364. 2012.PubMed/NCBI View Article : Google Scholar

68 

Chen L, Yuan X, Zou L, Peng J and Hu X: Effects of 1,25-Dihydroxyvitamin D3 on the prevention of chronic obstructive pulmonary disease (COPD) in rats exposed to air pollutant particles less than 2.5 micrometers in diameter (PM2.5). Med Sci Monit. 24:356–362. 2018.PubMed/NCBI View Article : Google Scholar

69 

Hyun DG, Oh YM, Lee SW, Lee SD and Lee JS: Clinical phenotypes, comorbidities, and exacerbations according to serum 25-OH vitamin D and plasma fibrinogen levels in chronic obstructive pulmonary disease. J Korean Med Sci. 34(e195)2019.PubMed/NCBI View Article : Google Scholar

70 

Hu G, Dong T, Wang S, Jing H and Chen J: Vitamin D3-vitamin D receptor axis suppresses pulmonary emphysema by maintaining alveolar macrophage homeostasis and function. EBioMedicine. 45:563–577. 2019.PubMed/NCBI View Article : Google Scholar

71 

Nikniaz L, Ghojazadeh M, Nateghian H, Nikniaz Z, Farhangi MA and Pourmanaf H: The interaction effect of aerobic exercise and vitamin D supplementation on inflammatory factors, anti-inflammatory proteins, and lung function in male smokers: A randomized controlled trial. BMC Sports Sci Med Rehabil. 13(102)2021.PubMed/NCBI View Article : Google Scholar

72 

Mahmood MQ, Reid D, Ward C, Muller HK, Knight DA, Sohal SS and Walters EH: Transforming growth factor (TGF) β1 and smad signalling pathways: A likely key to EMT-associated COPD pathogenesis. Respirology. 22:133–140. 2017.PubMed/NCBI View Article : Google Scholar

73 

Wang L, Meng J, Wang C, Yang C, Wang Y and Li Y and Li Y: Hydrogen sulfide alleviates cigarette smoke-induced COPD through inhibition of the TGF-β1/smad pathway. Exp Biol Med (Maywood). 245:190–200. 2020.PubMed/NCBI View Article : Google Scholar

74 

Gohy ST, Hupin C, Fregimilicka C, Detry BR, Bouzin C, Chevronay HG, Lecocq M, Weynand B, Ladjemi MZ, Pierreux CE, et al: Imprinting of the COPD airway epithelium for dedifferentiation and mesenchymal transition. Eur Respir J. 45:1258–1272. 2015.PubMed/NCBI View Article : Google Scholar

75 

Camara 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

Related Articles

Journal Cover

May-2022
Volume 23 Issue 5

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
Mao Y and Mao Y: Vitamin D3 alleviates cigarette smoke extract‑mediated epithelial‑mesenchymal transition and fibrogenesis by upregulating CC16 expression in bronchial epithelial cells. Exp Ther Med 23: 357, 2022
APA
Mao, Y., & Mao, Y. (2022). Vitamin D3 alleviates cigarette smoke extract‑mediated epithelial‑mesenchymal transition and fibrogenesis by upregulating CC16 expression in bronchial epithelial cells. Experimental and Therapeutic Medicine, 23, 357. https://doi.org/10.3892/etm.2022.11284
MLA
Mao, Y., Feng, H."Vitamin D3 alleviates cigarette smoke extract‑mediated epithelial‑mesenchymal transition and fibrogenesis by upregulating CC16 expression in bronchial epithelial cells". Experimental and Therapeutic Medicine 23.5 (2022): 357.
Chicago
Mao, Y., Feng, H."Vitamin D3 alleviates cigarette smoke extract‑mediated epithelial‑mesenchymal transition and fibrogenesis by upregulating CC16 expression in bronchial epithelial cells". Experimental and Therapeutic Medicine 23, no. 5 (2022): 357. https://doi.org/10.3892/etm.2022.11284