The profibrotic effect of downregulated Na,K‑ATPase β1 subunit in alveolar epithelial cells during lung fibrosis

  • Authors:
    • Biyun Li
    • Xiaoxi Huang
    • Xuefeng Xu
    • Wen Ning
    • Huaping Dai
    • Chen Wang
  • View Affiliations

  • Published online on: May 16, 2019     https://doi.org/10.3892/ijmm.2019.4201
  • Pages: 273-280
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by progressive lung scarring and excessive extracellular matrix depositon. When stimulated, alveolar epithelial cells (AECs) are aberrantly activated, the expression of profibrotic molecules is enhanced, and lung fibrosis is promoted, but the mechanism for this is unclear. It has been reported that a downregulation of the Na,K‑ATPase β1 subunit in renal epithelial cells is involved in renal fibrosis development, but the role of this protein in lung fibrosis remains unknown. In the present study, the expression of the Na,K‑ATPase β1 subunit was revealed to be markedly decreased in AECs of patients with IPF and a bleomycin‑induced pulmonary fibrosis mouse model. Treatment with transforming growth factor β‑1 led to significantly downregulation of the Na,K‑ATPase β1 subunit in lung adenocarcioma A549 cells. Furthermore, the knockdown of the Na,K‑ATPase β1 subunit in A549 cells resulted in the upregulation of profibrotic molecules, activation of the neurogenic locus notch homolog protein 1 and extracellular signal‑regulated kinase 1/2 signaling pathways and induction of endoplasmic reticulum stress. These findings reveal that the downregulation of the Na,K‑ATPase β1 subunit enhances the expression of profibrotic molecules in AECs and may contribute to IPF pathogenesis.

Introduction

Idiopathic pulmonary fibrosis (IPF) is a prevalent and progressive fatal fibrotic lung disease with few available effective therapies (1-4). It mainly occurs in elderly adults with a median survival time of 2-3 years (5,6), and the etiology of IPF remains unclear. A prevailing hypothesis for IPF pathogenesis is that abnormal wound healing in response to ongoing alveolar epithelial microinjuries causes fibroblast activation and excess extracellular matrix deposition, ultimately resulting in lung damage (7-9).

Alveolar epithelial cells (AECs) serve a crucial role in IPF pathogenesis (10). Persistent microinjuries to AECs are thought to be a trigger of lung fibrosis. The origins of lung injury are often varied and complex. Exposure to smoke, various types of dust, gastroesophageal reflux and viral infection can induce AEC injury (11-15). Rebuilding AECs is a key component of normal wound healing following injury. This requires a carefully programmed response, including the proliferation and migration of type II AECs. However, type II AECs isolated from the lungs of patients with IPF are aberrantly activated with increased collagen, α-smooth muscle actin (α-SMA) and fibronectin, and decreased expression of E-cadherin (16). The role of epithelial-mesenchymal transition (EMT) in lung fibrosis remains controversial (17,18). Furthermore, AEC senescence, endoplasmic reticulum (ER) stress, fibroblast resistance to apoptosis, insufficient autophagy, ubiquitination dysfuction, abnormal macrophage activation, gene mutation and epigenetic changes are involved in IPF development (19-25).

The Na,K-ATPase β1 subunit has been reported to be involved in organ fibrosis. Rajasekaran et al (26) demonstrated that Na,K-ATPase β1 subunit expression is significantly decreased in renal fibrotic tissues. The knockdown of this subunit in porcine kidney LLC-PK1 cells induced EMT, as did its downregulation in retinal pigment epithelial cells (27). Na,K-ATPase, also known as a sodium pump, transports 3 Na+ and 2 K+ ions in opposite directions across the cell membrane to maintain osmotic equilibrium. This protein pump is composed of 3 subunits, α, β and γ. The functional α subunit has 4 isoforms (α1, α2, α3 and α4), whereas the β (β1, β2 and β3) and γ (isoforms 1-7) subunits are regulatory (28). Additional functions of Na,K-ATPase have been identified in the regulation of cell proliferation, cell motility, and apoptosis (29,30).

In the present study, the expression of Na,K-ATPase β1 subunit was revealed to be decreased in AECs of patients with IPF and in a bleomycin-induced pulmonary fibrosis mouse model. Based on this observation, the role of the downregulation of the Na,K-ATPase β1 subunit in AECs during lung fibrosis was investigated.

Materials and methods

Tissue samples form patients

Lung tissue samples from 13 patients with IPF and 5 healthy donors (all male; age 51.08±10.03 years) were obtained from the China-Japan Friendship Hospital (Beijing, China) during surgical lung biopsy and lung transplantation for inclusion in the present study. The diagnosis of IPF was based on the 2011 American Thoracic Society/European Respiratory Society/Japanese Respiratory Society/Latin American Thoracic Association Guidelines for Diagnosis and Management (5). All patients provided signed consent and the study was approved by the Ethics Committee of the China-Japan Friendship Hospital (approval no. 2017-25-1).

Animal model

C57BL/6N mice (50 mice; male; 7-8 weeks old; 22-24 g) were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). The animals were maintained at a controlled temperature of 24±1°C with a 12/12 h light-dark cycle, and were fed a standard diet. Water was freely available. The mice were randomly divided into 2 groups: For the model of pulmonary fibrosis, a dose of 2 mg/kg bleomycin (Nippon Kayaku Co., Ltd., Tokyo, Japan) was intratracheally administered, and the control mice were injected intratracheally with the same volume of saline. The bleomycin and saline were adminstrated only once. The mice were euthanized on day 21 with an intraperitoneal injection of 1% pentobarbital sodium (100 mg/kg animal weight). This study was approved by the Animal Ethics Committee of China-Japan Friendship Hospital (Beijing, China).

Immunohistochemistry and immunofluorescence

The preparation of the human and mouse lung specimens for histology was performed as previously described (23,31). Briefly, the samples were dehydrated, paraffin-embedded, and cut into 4-μm sections. The tissue sections were deparaffinized and rehydrated. Following a microwave treatment for 20 min in EDTA buffer and subsequent cooling, the endogenous peroxidase activity was blocked with 0.3% hydrogen peroxide in methanol for 15 min in the dark. Following blocking in 5% goat serum (OriGene Technologies, Inc., Beijing, China) for 20 min, the sections were incubated with antibodies against the Na,K-ATPase β1 subunit (cat. no. ab193669; 1:600 dilution) and fibronectin (cat. no. ab2413; 1:500 dilution) (both Abcam, Cambridge, UK) overnight at 4°C as described previously (32), the samples were observed using an optical microscope (magnification, ×100) and were analyzed using Aperio Imagescope version 12.0 software (Leica Microsystems, Ltd., Milton Keynes, UK).

For immunofluorescence, the mouse lung tissue sections (4 μm) were de-paraffinized, hydrated using xylene, 100, 95, 85 and 70% ethanol, and PBS solution. The non-specific binding was blocked with 10% goat serum, and the samples were incubated overnight at 4°C with the desired primary antibodies against the Na,K-ATPase β1 subunit (cat. no. ab2873; 1:500 dilution) and prosurfactant protein C (cat. no. ab90716; 1:4,000 dilution) (both Abcam), and then incubated with a specific fluorescence-conjugated secondary IgG (fluorescein isothiocyanate-conjugated, cat. no. ZF-0311; rhodamine B isothiocyanate-conjugated, cat. no. ZF-0313; both 1:100 dilution; OriGene Technologies, Inc.) for 1 h in a light-protected chamber at room temperature. Subsequently, the sections were counterstained with DAPI (cat. no. P0131; Beyotime Institute of Biotechnology, Haimen, China) at room temperature and immunofluorescence signals were detected immediately using fluorescence microscopy (magnification, ×100).

Cell culture and small interfering (si)RNA transfection

Human lung carcinoma epithelial A549 cells were purchased from the American Type Culture Collection (Manassas, VA, USA). A549 is a human AEC line with similar characteristics to type II AECs. It has been used as a stable AEC line in a number of studies (33,34). The cells were maintained in RPMI-1640 medium with 10% fetal bovine serum (both Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 100 U/ml penicillin and 100 mg/ml streptomycin (both Hyclone; GE Healthcare Life Sciences, Logan, UT, USA). Transforming growth factor β-1 (TGF-β1; 10 ng/ml; R&D Systems, Inc., Minneapolis, MN, USA) was added to subconfluent cultures and the same volume of citric acid was added to the control cells. The cells were maintained in a humidified incubator at 37°C in 95% air (21% O2) and 5% CO2.

The A549 cells were seeded in 6-well plates and incubated overnight. Na,K-ATPase β1 subunit siRNA (5 μM; sequence, 5′-AAU GUU CUC ACC GUA CGC Ctt-3′) and negative control siRNA (5 μM; cat. no. 4390843; Silencer® Select Negative Control; Thermo Fisher Scientific, Inc.) were separately mixed with Lipofectamine® 3000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and Opti-MEM medium (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The cells were incubated for 24 h for the measurement of RNA levels and 48 h for cell morphology observation under an optical microscope and protein detection experiments.

RNA purification and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

As previously described (35), total RNA was isolated from the cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. RT was performed on 1 μg total RNA with oligo(dT) primers in 25-μl reactions using the Omniscript RT kit (Tiangen Biotech Co., Ltd., Beijing, China) at 37°C for 60 min according to the manufacturer's instructions. The qPCR was performed on an ABI 7500 instrument (Applied Biosystems; Thermo Fisher Scientific, Inc.) using SYBR-Green PCR reagents (Tiangen Biotech Co., Ltd.). The thermocycling conditions used were as follows: Initial denaturation at 94°C for 2 min and 40 cycles of denaturation at 94°C for 15 sec, annealing at 55°C for 20 sec and extension at 69°C for 35 sec. The primers used were: β-actin forward, 5′-AGGCCAACCGTGAAAAGATG-3′; and reverse, 5′-AGAGCATAGCCCTCGTAGATGG-3′; and Na,K-ATPase β1 subunit forward, 5′-ATGTGCCCAGTGAACCGAAA-3′; and reverse, 5′-TCCAGAGCAATTTCCCAGCC-3′. The relative expression of the target gene was calculated using the 2−ΔΔCq method (36), normalized to the levels of β-actin.

Protein extraction and western blot analysis

Total cell lysates were obtained using radioimmunoprecipitation assay buffer (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) containing 1:100 phenylmethylsulfonyl fluoride, phosphatase inhibitors and protease inhibitor. The cell lysates were resuspended in protein loading buffer containing 5% mercaptoethanol. The protein concentration was determined using a bicinchoninic acid assay kit. Western blotting was performed as previously described (23). The denatured proteins (20 μg per lane) were separated by 10% SDS-PAGE using a Mini-Protein electrophoresis module assembly (both Bio-Rad Laboratories, Inc., Hercules, CA, USA) at 80 mV and transferred to nitrocellulose membranes (Merck KGaA, Darmstadt, Germany) for 100-120 min using the Mini Trans-Blot electrophoresis transfer cell (Bio-Rad Laboratories, Inc.) at 300 mA, according to the molecular weight. The primary antibodies used were anti-human and mouse α-SMA (cat. no. ab124964; 1:5,000 dilution), fibronectin (cat. no. ab2413; 1:2,000 dilution), β-actin (cat. no. ab6267; 1:3,000 dilution), Na,K-ATPase β1 subunit (cat. no. ab2873; 1:600 dilution) (all Abcam), cleaved Notch1 (cat. no. 4147T), extracellular signal-regulated kinase (ERK)1/2 (cat. no. 9101), phosphorylated ERK1/2 (cat. no. 8544), and immunoglobulin heavy chain-binding protein (BiP, cat. no. 3177T) (all 1:1,000 dilution; Cell Signaling Technology, Inc., Danvers, MA, USA). The membranes were incubated with the primary antibodies overnight at 4°C and treated with IRDyeCW800 (green)- or IRDyeCW800 (red)-conjugated affinity purified anti-rabbit (cat. no. 925-32211) or anti-mouse (cat. no. 925-32210) IgG (both 1:15,000 dilution; LI-COR Biosciences, Lincoln, NE, USA). The intensity of the bands was evaluated using a LI-COR Odyssey infrared double-fluorescence imaging system (LI-COR Image Studio Software version 4.0; LI-COR Biosciences).

Statistical analysis

Data are expressed as mean ± standard error of the mean. Two-tailed Student's t-test was performed for the comparison of mRNA and protein expression levels. The statistical analyses were performed using the Prism software version 5.0 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate statistically significant differences.

Results

Na,K-ATPase β1 subunit expression is downregulated in lung fibrosis

To examine the Na,K-ATPase β1 subunit expression level in lung fibrosis, immunohistochemistry was performed on lung tissue sections from patients with IPF. The Na,K-ATPase β1 subunit was mainly expressed in the cytoplasm of AECs. The staining of this subunit was visibly diminished in the fibrotic and lesion-adjacent areas compared with that in the healthy lung tissue (Fig. 1A). Na,K-ATPase β1 subunit expression was also investigated in tissue from a bleomycin-induced pulmonary fibrosis mouse model. The results of the western blot and immunohistochemistry analyses demonstrated that Na,K-ATPase β1 subunit expression was markedly decreased in the lung tissue of the bleomycin group compared with that of control (Fig. 1B and C).

Na,K-ATPase β1 subunit expression in type II AECs in mouse lung tissue

In contrast to the human lung tissue, the immunohistochemistry of the mouse lung tissue revealed that not all AECs express the Na,K-ATPase β1 subunit (Fig. 1B). Therefore, immunofluorescence was used to identify which type of AECs express Na,K-ATPase β1 subunit in mouse lung. As displayed in Fig. 2, the Na,K-ATPase β1 subunit was primarily expressed in the cell membrane of type II AECs.

TGF-β1 treatment decreases Na,K-ATPase β1 subunit expression in A549 cells

TGF-β1 is an important profibrotic cytokine. To assess the level of Na,K-ATPase β1 subunit in TGF-β1-stimulated AECs, total protein was extracted from the cells following treatment with 10 ng/ml TGF-β1 for 48 and 72 h. The results indicated that the treatment led to a significant decrease in the protein expression of Na,K-ATPase β1 subunit at the two tested time points (Fig. 3).

Knockdown of Na,K-ATPase β1 subunit mediates changes in the cell morphology of A549 cells

Due to the observed significant difference between the expression of Na,K-ATPase β1 subunit in lung fibrosis and normal lung samples, the effects of the downregulation of the Na,K-ATPase β1 subunit on AECs was explored. The Na,K-ATPase β1 subunit expression in A549 cells was knocked down using siRNA interference. As demonstrated in Fig. 4A, Na,K-ATPase β1 subunit mRNA was significantly decreased 24 h post-transfection, as were the protein expression levels at 48 h (Fig. 4B and C). In addition, the knockdown of Na,K-ATPase β1 subunit resulted in an altered spindle morphology in the A549 cells (Fig. 4D).

Knockdown of the Na,K-ATPase β1 subunit promotes the upregulation of profibrotic proteins in A549 cells

To further investigate the role of the Na,K-ATPase β1 subunit in AECs during lung fibrosis, the expression of fibrosis-associated proteins fibronectin, α-SMA and E-cadherin was examined in A549 cells following siRNA silencing of the Na,K-ATPase β1 subunit. The results revealed that the fibronectin and α-SMA levels were increased, but E-cadherin expression was not significantly altered, compared with that in the cells transfected with NC-siRNA (Fig. 5).

Knockdown of the Na,K-ATPase β1 subunit activates ERK1/2 and neurogenic locus notch homolog protein 1 (Notch1) signaling and induces ER stress

The downstream signaling pathway of the Na,K-ATPase β1 subunit was investigated. As observed in Fig. 6A, phosphorylated ERK1/2 and cleaved Notch1 were significantly upregulated in cells with Na,K-ATPase β1 subunit silencing, suggesting that a deficiency of this protein may lead to the activation of the ERK1/2 and Notch1 signaling pathways, contributing to lung fibrosis. In addition, the knockdown of Na,K-ATPase β1 subunit resulted in a significant increase in the expression of BiP, an ER-stress protein marker (Fig. 6B), suggesting that downregulation of this ion pump causes ER stress in A549 cells.

Discussion

IPF is a chronic and lethal interstitial lung disease. It is generally accepted that the initial progression of IPF is stimulated by the aberrant activation of AECs in response to repetitive microinjury. In the present study, the Na,K-ATPase β1 subunit protein expression was revealed to be downregu-lated in lung fibrosis, mainly in AECs, enhancing profibrotic protein expression, activating the ERK1/2 and Notch1 signaling pathways, and inducing ER stress, consequently leading to lung fibrosis.

The present study has demonstrated that the expression of the Na,K-ATPase β1 subunit is different in human and mouse lung tissues. It is expressed in type I and II AECs, and located in the cytoplasm of AECs in human lungs. However, in mouse lungs, the Na,K-ATPase β1 subunit is mainly expressed in type II AECs and is located in the cell membrane. The human Na,K-ATPase β1 subunit exhibits two bands in immunoblot analyses, where the lower 40-kDa band represents the intracellular immature fraction of the subunit and the higher molecular weight band represents the mature plasma membrane form. However, the mouse Na,K-ATPase β1 subunit results in only a single 42-kDa band (37).

The Na,K-ATPase β1 subunit belongs to the N-linked glycoproteins, and as a regulatory subunit, its main fuction is to assist the folding of the α subunit and its transport from the ER to the plasma membrance (38). Furthermore, the Na,K-ATPase β1 subunit is a molecular partner of Wolframin, an ER protein involved in ER stress (39). The results of the present study indicated that the knockdown of this subunit led to the upregulation of BiP, whereas the level of DNA damage-inducible transcript 3 protein was not altered (data not shown). Over the past decades, accumulating evidence has suggested that ER stress serves an important role in the pathogenesis of lung fibrosis, as ER stress markers are highly expressed in AECs in IPF. ER stress in lung fibrosis induces AEC injury and apotosis, causing inflammation and cell phenotype alteration (20,40-43).

The present data revealed that the knockdown of Na,K-ATPase β1 subunit led to the enhanced expression of profibrotic proteins fibronectin and α-SMA, but no changes in epithelial marker E-cadherin were observed, suggesting that AECs undergo incomplete activation and partly maintain epithelial characteristics (44). Treatment of A549 cells with TGF-β1 resulted in a decrease in Na,K-ATPase β1 subunit expression, which may affect electrolyte metabolism in AECs. Further investigation is required to clarify the role of Na,K-ATPase α1 subunit-mediated electrolyte metabolism dysfuction in the pathegenesis of lung fibrosis.

Attempts to use plasmids to overexpress Na,K-ATPase β1 subunit in A549 cells proved unsuccessful in the present study. Ouabain, an inhibitor of Na,K-ATPase, leads to the upregulation of Na,K-ATPase β1 subunit expression and suppresses EMT (26,45). In addition, a previous study of the present group revealed that ouabain ameliorates bleomycin-induced pulmonary fibrosis (46). Therefore, it can be inferred that this inhibitor suppresses EMT due to the upregulation of of Na,K-ATPase β1 subunit expression, providing direction of subsequent studies.

In conclusion, Na,K-ATPase β1 subunit expression is down-regulated in clinical human IPF samples, in the lung tissue of a bleomycin-induced pulmonary fibrosis mouse model, and in TGF-β1-stimulated lung carcinoma A549 cells. Additionally, Na,K-ATPase β1 subunit deficiency in A549 cells upregulates profibrotic protein expression, activates ERK1/2 and Notch1 signaling pathways and induces ER stress. Therefore, the results of the present study suggest that decreased expression of Na,K-ATPase β1 subunit in AECs serves a crucial role in the progression of lung fibrosis.

Funding

This study was supported by grants from the National Natural Science Foundation of China (nos. 81430001 and 81470258).

Availability of data and materials

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

Authors' contributions

BL, WN, HD and CW designed the experiments. BL performed the experiments and drafted the manuscript. BL, XX and XH analyzed the data. XX, WN, HD and CW revised the manuscript. All authors have read and approved the final version for publication.

Ethics approval and consent to participate

This study was approved by the Ethics Committee (approval no. 2017-25-1) and the Animal Ethics Committee (approval no. 2017-18-2) of China-Japan Friendship Hospital, Beijing, China.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

This abstract was presented at the ERS International Congress, September 15-19, 2018 in Paris, France and was published as Abstract no. PA3720 in the European Respiratory Journal 52 (Suppl 62) 2018. The authors thank Professor Ying Li, Department of Medical Research, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China, and Professor Yunchao Su, Department of Pharmacology and Toxicology, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA, for their excellent technical assistance.

References

1 

Coultas DB, Zumwalt RE, Black WC and Sobonya RE: The epidemiology of interstitial lung diseases. Am J Respir Crit Care Med. 150:967–972. 1994. View Article : Google Scholar : PubMed/NCBI

2 

Mannino DM, Etzel RA and Parrish RG: Pulmonary fibrosis deaths in the United States, 1979-1991. An analysis of multiple-cause mortality data. Am J Respir Crit Care Med. 153:1548–1552. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Noble PW: Idiopathic pulmonary fibrosis: Natural history and prognosis. Clin Chest Med. 27(1 Suppl 1): S11–S16. v2006. View Article : Google Scholar : PubMed/NCBI

4 

Raghu G, Weycker D, Edelsberg J, Bradford WZ and Oster G: Incidence and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 174:810–816. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA, et al: An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 183:788–824. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Cai M, Zhu M, Ban C, Su J, Ye Q, Liu Y, Zhao W, Wang C and Dai H: Clinical features and outcomes of 210 patients with idiopathic pulmonary fibrosis. Chin Med J (Engl). 127:1868–1873. 2014.

7 

Selman M, King TE and Pardo A; American Thoracic Society; European Respiratory Society; American College of Chest Physicians: Idiopathic pulmonary fibrosis: Prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med. 134:136–151. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Thannickal VJ, Toews GB, White ES, Lynch JP III and Martinez FJ: Mechanisms of pulmonary fibrosis. Annu Rev Med. 55:395–417. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Zoz DF, Lawson WE and Blackwell TS: Idiopathic pulmonary fibrosis: A disorder of epithelial cell dysfunction. Am J Med Sci. 341:435–438. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Selman M and Pardo A: Role of epithelial cells in idiopathic pulmonary fibrosis: From innocent targets to serial killers. Proc Am Thorac Soc. 3:364–372. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Baumgartner KB, Samet JM, Stidley CA, Colby TV and Waldron JA: Cigarette smoking: A risk factor for idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 155:242–248. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Taskar VS and Coultas DB: Is idiopathic pulmonary fibrosis an environmental disease? Proc Am Thorac Soc. 3:293–298. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Raghu G, Freudenberger TD, Yang S, Curtis JR, Spada C, Hayes J, Sillery JK, Pope CE II and Pellegrini CA: High prevalence of abnormal acid gastro-oesophageal reflux in idiopathic pulmonary fibrosis. Eur Respir J. 27:136–142. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Hubbard R, Lewis S, Richards K, Johnston I and Britton J: Occupational exposure to metal or wood dust and aetiology of cryptogenic fibrosing alveolitis. Lancet. 347:284–289. 1996. View Article : Google Scholar : PubMed/NCBI

15 

Kelly BG, Lok SS, Hasleton PS, Egan JJ and Stewart JP: A rearranged form of Epstein-Barr virus DNA is associated with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 166:510–513. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Marmai C, Sutherland RE, Kim KK, Dolganov GM, Fang X, Kim SS, Jiang S, Golden JA, Hoopes CW, Matthay MA, et al: Alveolar epithelial cells express mesenchymal proteins in patients with idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 301:L71–L78. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Rock JR, Barkauskas CE, Cronce MJ, Xue Y, Harris JR, Liang J, Noble PW and Hogan BL: Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc Natl Acad Sci USA. 108:E1475–E1483. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D and Chapman HA: Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA. 103:13180–13185. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Jiang C, Liu G, Luckhardt T, Antony V, Zhou Y, Carter AB, Thannickal VJ and Liu RM: Serpine 1 induces alveolar type II cell senescence through activating p53-p21-Rb pathway in fibrotic lung disease. Aging Cell. 16:1114–1124. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Korfei M, Ruppert C, Mahavadi P, Henneke I, Markart P, Koch M, Lang G, Fink L, Bohle RM, Seeger W, et al: Epithelial endoplasmic reticulum stress and apoptosis in sporadic idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 178:838–846. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Im J, Kim K, Hergert P and Nho RS: Idiopathic pulmonary fibrosis fibroblasts become resistant to Fas ligand-dependent apoptosis via the alteration of decoy receptor 3. J Pathol. 240:25–37. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Araya J, Kojima J, Takasaka N, Ito S, Fujii S, Hara H, Yanagisawa H, Kobayashi K, Tsurushige C, Kawaishi M, et al: Insufficient autophagy in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 304:L56–L69. 2013. View Article : Google Scholar

23 

Geng J, Huang X, Li Y, Xu X, Li S, Jiang D, Liang J, Wang C and Dai H: Down-regulation of USP13 mediates phenotype transformation of fibroblasts in idiopathic pulmonary fibrosis. Respir Res. 16:1242015. View Article : Google Scholar : PubMed/NCBI

24 

Nie Y, Sun L, Wu Y, Yang Y, Wang J, He H, Hu Y, Chang Y, Liang Q, Zhu J, et al: AKT2 regulates pulmonary inflammation and fibrosis via modulating macrophage activation. J Immunol. 198:4470–4480. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Peljto AL, Zhang Y, Fingerlin TE, Ma SF, Garcia JG, Richards TJ, Silveira LJ, Lindell KO, Steele MP, Loyd JE, et al: Association between the MUC5B promoter polymorphism and survival in patients with idiopathic pulmonary fibrosis. JAMA. 309:2232–2239. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Rajasekaran SA, Huynh TP, Wolle DG, Espineda CE, Inge LJ, Skay A, Lassman C, Nicholas SB, Harper JF, Reeves AE, et al: Na,K-ATPase subunits as markers for epithelial-mesenchymal transition in cancer and fibrosis. Mol Cancer Ther. 9:1515–1524. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Mony S, Lee SJ, Harper JF, Barwe SP and Langhans SA: Regulation of Na,K-ATPase beta1-subunit in TGF-β2-mediated epithelial-to-mesenchymal transition in human retinal pigmented epithelial cells. Exp Eye Res. 115:113–122. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Pierre SV and Xie Z: The Na,K-ATPase receptor complex: Its organization and membership. Cell Biochem Biophys. 46:303–316. 2006. View Article : Google Scholar

29 

Xie Z and Askari A: Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem. 269:2434–2439. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Barwe SP, Anilkumar G, Moon SY, Zheng Y, Whitelegge JP, Rajasekaran SA and Rajasekaran AK: Novel role for Na,K-ATPase in phosphatidylinositol 3-kinase signaling and suppression of cell motility. Mol Biol Cell. 16:1082–1094. 2005. View Article : Google Scholar :

31 

Dong Y, Geng Y, Li L, Li X, Yan X, Fang Y, Zheng X, Dong S, Liu X, Yang X, et al: Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary fibrosis in mice. J Exp Med. 212:235–252. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Yogo Y, Fujishima S, Inoue T, Saito F, Shiomi T, Yamaguchi K and Ishizaka A: Macrophage derived chemokine (CCL22), thymus and activation-regulated chemokine (CCL17), and CCR4 in idiopathic pulmonary fibrosis. Respir Res. 10:802009. View Article : Google Scholar : PubMed/NCBI

33 

Wang YC, Liu JS, Tang HK, Nie J, Zhu JX, Wen LL and Guo QL: miR-221 targets HMGA2 to inhibit bleomycin-induced pulmonary fibrosis by regulating TGFβ1/Smad3-induced EMT. Int J Mol Med. 38:1208–1216. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Zheng Q, Tong M, Ou B, Liu C, Hu C and Yang Y: Isorhamnetin protects against bleomycin-induced pulmonary fibrosis by inhibiting endoplasmic reticulum stress and epithelial-mesenchymal transition. Int J Mol Med. 43:117–126. 2019.

35 

Xu X, Wan X, Geng J, Li F, Yang T and Dai H: Rapamycin regulates connective tissue growth factor expression of lung epithelial cells via phosphoinositide 3-kinase. Exp Biol Med (Maywood). 238:1082–1094. 2013. View Article : Google Scholar

36 

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. View Article : Google Scholar

37 

Tokhtaeva E, Sachs G and Vagin O: Assembly with the Na, K-ATPase alpha(1) subunit is required for export of beta(1) and beta(2) subunits from the endoplasmic reticulum. Biochemistry. 48:11421–11431. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Lemas MV, Hamrick M, Takeyasu K and Fambrough DM: 26 amino acids of an extracellular domain of the Na,K-ATPase alpha-subunit are sufficient for assembly with the Na,K-ATPase beta-subunit. J Biol Chem. 269:8255–8259. 1994.PubMed/NCBI

39 

Zatyka M, Ricketts C, da Silva Xavier G, Minton J, Fenton S, Hofmann-Thiel S, Rutter GA and Barrett TG: Sodium-potassium ATPase 1 subunit is a molecular partner of Wolframin, an endoplasmic reticulum protein involved in ER stress. Hum Mol Genet. 17:190–200. 2008. View Article : Google Scholar

40 

Lawson WE, Crossno PF, Polosukhin VV, Roldan J, Cheng DS, Lane KB, Blackwell TR, Xu C, Markin C, Ware LB, et al: Endoplasmic reticulum stress in alveolar epithelial cells is prominent in IPF: Association with altered surfactant protein processing and herpesvirus infection. Am J Physiol Lung Cell Mol Physiol. 294:L1119–L1126. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Mulugeta S, Nguyen V, Russo SJ, Muniswamy M and Beers MF: A surfactant protein C precursor protein BRICHOS domain mutation causes endoplasmic reticulum stress, proteasome dysfunction, and caspase 3 activation. Am J Respir Cell Mol Biol. 32:521–530. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Maguire JA, Mulugeta S and Beers MF: Endoplasmic reticulum stress induced by surfactant protein C BRICHOS mutants promotes proinflammatory signaling by epithelial cells. Am J Respir Cell Mol Biol. 44:404–414. 2011. View Article : Google Scholar :

43 

Ulianich L, Garbi C, Treglia AS, Punzi D, Miele C, Raciti GA, Beguinot F, Consiglio E and Di Jeso B: ER stress is associated with dedifferentiation and an epithelial-to-mesenchymal transition-like phenotype in PC Cl3 thyroid cells. J Cell Sci. 121:477–486. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Morbini P, Inghilleri S, Campo I, Oggionni T, Zorzetto M and Luisetti M: Incomplete expression of epithelial-mesenchymal transition markers in idiopathic pulmonary fibrosis. Pathol Res Pract. 207:559–567. 2011. View Article : Google Scholar : PubMed/NCBI

45 

La J, Reed E, Chan L, Smolyaninova LV, Akomova OA, Mutlu GM, Orlov SN and Dulin NO: Downregulation of TGF-β Receptor-2 Expression and Signaling through Inhibition of Na/K-ATPase. PLoS One. 11:e01683632016. View Article : Google Scholar

46 

Li B, Huang X, Liu Z, Xu X, Xiao H, Zhang X, Dai H and Wang C: Ouabain ameliorates bleomycin induced pulmonary fibrosis by inhibiting proliferation and promoting apoptosis of lung fibroblasts. Am J Transl Res. 10:2967–2974. 2018.PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 44 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li B, Huang X, Xu X, Ning W, Dai H and Wang C: The profibrotic effect of downregulated Na,K‑ATPase β1 subunit in alveolar epithelial cells during lung fibrosis. Int J Mol Med 44: 273-280, 2019
APA
Li, B., Huang, X., Xu, X., Ning, W., Dai, H., & Wang, C. (2019). The profibrotic effect of downregulated Na,K‑ATPase β1 subunit in alveolar epithelial cells during lung fibrosis. International Journal of Molecular Medicine, 44, 273-280. https://doi.org/10.3892/ijmm.2019.4201
MLA
Li, B., Huang, X., Xu, X., Ning, W., Dai, H., Wang, C."The profibrotic effect of downregulated Na,K‑ATPase β1 subunit in alveolar epithelial cells during lung fibrosis". International Journal of Molecular Medicine 44.1 (2019): 273-280.
Chicago
Li, B., Huang, X., Xu, X., Ning, W., Dai, H., Wang, C."The profibrotic effect of downregulated Na,K‑ATPase β1 subunit in alveolar epithelial cells during lung fibrosis". International Journal of Molecular Medicine 44, no. 1 (2019): 273-280. https://doi.org/10.3892/ijmm.2019.4201