Open Access

Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase

  • Authors:
    • Jian Lu
    • Yongbing Qian
    • Wei Jin
    • Rui Tian
    • Yong Zhu
    • Jinfeng Wang
    • Xiaoxiao Meng
    • Ruilan Wang
  • View Affiliations

  • Published online on: December 22, 2017     https://doi.org/10.3892/etm.2017.5677
  • Pages: 2287-2294
  • Copyright: © Lu 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

Pulmonary fibrosis (PF) is one of the most prevalent causes of death following paraquat (PQ) poisoning. As demonstrated in previous studies by the present authors, epithelial-to-mesenchymal transition (EMT) is associated with PQ‑induced PF. In addition, hypoxia‑inducible factor‑1α (HIF‑1α) and lysyl oxidase (LOX) promote EMT following PQ poisoning. However, the association between HIF‑1α‑ and LOX‑mediated regulation of EMT remains unclear. The present study investigated the association between HIF‑1α and LOX with regard to PQ‑induced EMT. A549 and RLE‑6TN cells were treated with PQ, and HIF‑1α and LOX expression was silenced with short interfering RNAs. Changes in the expression of HIF‑1α, LOX, β‑catenin and EMT‑related makers were detected using real‑time quantitative polymerase chain reaction, immunofluorescence, and western blotting. HIF‑1α and LOX were associated with PQ‑induced EMT, and their expression levels were significantly increased (P<0.05). LOX expression was significantly decreased following PQ poisoning when HIF‑1α expression was inhibited (P<0.05). However, the level of HIF‑1α did not change significantly when LOX was silenced. The expression level of β‑catenin and the degree of EMT were significantly decreased following HIF‑1α and LOX silencing in both cell lines (P<0.05). The association between HIF‑1α and LOX in regulating EMT during PQ‑induced PF may be unidirectional. HIF‑1α may regulate PQ‑induced EMT through the LOX/β‑catenin pathway.

Introduction

Paraquat (PQ) has been one of the most effective and widely used herbicides over the last few decades, particularly in rural areas of developing countries; however, PQ poisoning has become a serious problem, with reports of mortality >90% (1,2). The primary pathological effects of PQ are observed in the lung, where pulmonary concentrations are 6–10 times higher than in plasma following PQ ingestion (3). Furthermore, PQ accumulates in the lungs as blood levels begin to decrease (3). The rapid accumulation of PQ damages the parenchymal cells in the lung and induces the excessive repair of lung tissues, which results in irreversible and extensive pulmonary fibrosis (PF) (3) and eventually leads to high mortality rates. However, the exact mechanism that leads to toxicity remains unclear, and no specific therapy has been recommended.

Epithelial-to-mesenchymal transition (EMT) occurs in multiple contexts, including embryonic development, tissue fibrosis, and cancer. EMT is defined as the process by which stationary epithelial cells (identified by high levels of E-cadherin and zonula occludens-1, which are markers of epithelial cells) undergo phenotypic changes, including the loss of cell-cell adhesion and apical-basal polarity, and acquire mesenchymal characteristics, including high levels of α-smooth muscle actin (α-SMA) and N-cadherin (markers of mesenchymal cells), that confer migratory capacity (4,5). According to previous findings, EMT has an important role in the development of PF. Alveolar epithelial cells could acquire mesenchyme cell phenotypes through EMT, these cells could then increase the deposition of extracellular matrix and further promote the development of PF (57). Furthermore, EMT has been demonstrated to serve an important role in PQ-induced PF in recent studies by the present authors (8,9).

Hypoxia-inducible factor-1α (HIF-1α) has roles in tumorigenesis, inflammation, and cell metabolism in hypoxia, and its expression is correlated with a variety of fibrotic diseases (10,11). HIF-1α has also been demonstrated to induce EMT and contribute to PF (12,13). Previous studies have detected an early increase in HIF-1α expression following PQ poisoning and revealed that HIF-1α modulates EMT in cases of PF (9,14).

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that is important for growth, stabilization, remodeling and repair. Its primary function is to catalyze the covalent cross-linking of collagens and elastin in the extracellular matrix, although it also has intracellular functions (15). LOX participates in various fibrosis processes, such as lung, myocardial and renal fibrosis (1618). As demonstrated in a previous study by the present authors, LOX promotes EMT in PQ-induced PF (8). LOX was previously considered a critical target of HIF-1α (19); however, HIF-1α and LOX have since been demonstrated to provide bidirectional regulation of colon and ovarian carcinomas (20,21). The potential for dual regulation via HIF-1α and LOX remains controversial, particularly in PQ-induced PF. The present study investigated the association between HIF-1α and LOX with regard to PQ-induced PF.

Materials and methods

Reagents

PQ powder was obtained from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Anti-HIF-1α antibodies (cat. no. BS3514) were purchased from Bioworld Technology, Inc. (St. Louis Park, MN, USA). Anti-LOX (cat. no. ab174316), anti-E-cadherin (cat. no. ab184633) and anti-α-SMA (cat. no. ab7817) primary antibodies were obtained from Abcam (Cambridge, MA, USA). Anti-β-catenin (cat. no. 8480) and anti-GAPDH (cat. no. 5174) antibodies were purchased from Cell Signaling Technology, Inc. (Boston, MA, USA). Horseradish peroxidase-conjugated anti-rabbit immunoglobulin (Ig)G (cat. no. A0208), anti-mouse IgG secondary antibodies (cat. no. A0216), immunofluorescence staining kits with Alexa Fluor 647-labeled goat anti-rabbit immunoglobulin G (cat. no. A0468) and kits with Alexa Fluor 488-labeled goat anti-rabbit IgG (cat. no. A0423) were obtained from Beyotime Institute of Biotechnology (Shanghai, China).

Cell culture

Human lung adenocarcinoma epithelial cells (A549) and rat alveolar type II cells (RLE-6TN) were obtained from the American Type Culture Collection (Manassas, VA, USA). A549 cells were cultured in Dulbecco's modified Eagle's medium (DMEM) (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and a 1% antibiotic solution (100 U/ml penicillin and 0.1 mg/ml streptomycin). RLE-6TN cells were cultured in DMEM/nutrient mixture F-12 supplemented with 10% FBS and 1% antibiotic solution. Both cell lines were cultured at 37°C in an atmosphere containing 5% CO2. Cells were subsequently treated with PQ (at a concentration of 800 µmol/l for A549 cells and 160 µmol/l for RLE-6TN cells) for 24 h at 37°C. These concentrations were used in accordance with a recent study by the present authors (9). The effect of HIF-1α or LOX silencing on cells was detected and the expression of other proteins was subsequently assessed using western blotting.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from cells using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.). The total RNA concentration was determined using an ultraviolet spectrophotometer. Reverse transcription was performed using a PrimeScript RT Master Mix kit (Takara Biotechnology Co., Ltd., Dalian, China), according to the manufacturer's instructions. Real-time quantitative PCR was performed using a SYBR Premix Ex Taq kit (Takara Biotechnology Co., Ltd.) in a ViiA 7 PCR system. Sangon Biotech Co., Ltd. (Shanghai, China) generated the primers for HIF-1α, LOX, β-catenin and β-actin. Primer sequences are listed in Table I. The thermocycling conditions were as follows: 2 min at 95°C for initial denaturation, followed by 40 amplification cycles consisting of 95°C for 10 sec (denaturation), 60°C for 30 sec (anneal) and 72°C for 30 sec (extension). The method of quantification used was the 2−ΔΔCq method (22). Each assay was performed in triplicate, and β-actin served as a loading control.

Table I.

Primer sequences used in reverse transcription-quantitative polymerase chain reaction.

Table I.

Primer sequences used in reverse transcription-quantitative polymerase chain reaction.

SpeciesGene (direction)Sequence (5′-3′)
HumanHIF-1α (F)GTC TGA GGG GAC AGG AGG AT
HIF-1α (R)CTC CTC AGG TGG CTT GTC AG
LOX (F)CAA CCT GAG ATG CGC GG
LOX (R)GGT CGG CTG GGT AAG AAA TC
β-catenin (F)CGT TTC GCC TTC ATT ATG GAC TAC CT
β-catenin (R)GCC GCT GGG TGT CCT GAT GT
β-actin (F)CTG GAA CGG TGA AGG TGA CA
β-actin (R)AAG GGA CTT CCT GTA ACA ATG CA
RatHIF-1α (F)AAG TCT AGG GAT GCA GCA CG
HIF-1α (R)AGA TGG GAG CTC ACG TTG TG
LOX (F)CCT ACT ACA TCC AGG CAT CCA
LOX (R)AGT CTC TGA CAT CCG CCC TA
β-catenin (F)GTG CAA TTC CTG AGC TGA CC
β-catenin (R)CGG GCT GTT TCT ACG TCA TT
β-actin (F)CCT CTA TGC CAC ACA GT
β-actin (R)AGC CAC CAA TCC ACA CAG

[i] HIF-1α, hypoxia-inducible factor-1α; LOX, lysyl oxidase; F, forward; R, reverse.

Western blotting

Total proteins were harvested from both cell lines in each group and lysed using radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology). Protein concentrations were determined using a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology). Total protein samples (~30 µg per lane) were separated via 8% SDS-PAGE (Beyotime Institute of Biotechnology), transferred to polyvinylidene difluoride membranes (Bio-Rad Laboratories, Inc., Hercules, CA, USA), blocked with 5% skimmed milk in Tris-buffered saline containing Tween-20 (TBST) for 90 min at room temperature (RT), and incubated with antibodies against HIF-1α (1:500), LOX (1:1,000), E-cadherin (1:500), α-SMA (1:500), β-catenin (1:1,000) or GAPDH (1:500) overnight at 4°C. Membranes were subsequently incubated with horseradish peroxidase-conjugated anti-rabbit IgG or anti-mouse IgG secondary antibodies (1:2,000; Beyotime Institute of Biotechnology) at RT. Following three washes with TBST, proteins were observed using a highly sensitive enhanced chemiluminescent agent (Thermo Fisher Scientific, Inc.). The band intensity was determined using ImageJ software (version 10.2; National Institutes of Health, Bethesda, MD, USA).

Immunofluorescence staining

Both cell lines were cultured in confocal dishes for 24 h at 37°C and incubated with PQ for 24 h at 37°C. Cells were washed with PBS, fixed with 4% paraformaldehyde (Sigma-Aldrich; Merck KGaA) for 10 min at RT, permeabilized with 0.5% Triton X100 (Sigma-Aldrich; Merck KgaA) for 10 min and blocked with 5% bovine serum albumin (1 g bovine serum albumin powder and 20 ml Tris-buffered saline; Beyotime Institute of Biotechnology) for 1 h at RT. Subsequently, cells were incubated with anti-LOX (1:100) or anti-HIF-1α (1:50) primary antibodies overnight at 4°C. Following three washes with TBST, cells were incubated with immunofluorescence staining kits with Alexa Fluor 647-labeled goat anti-rabbit IgG (1:200) and kits with Alexa Fluor 488-labeled goat anti-rabbit IgG (1:200) for 1.5 h at RT. Nuclei were stained with DAPI (Beyotime Institute of Biotechnology) for 3 min at RT. Fluorescent signals were detected with a laser confocal scanning microscope (Leica TCS SP8; Leica Microsystems GmbH, Wetzlar, Germany) and the cellular morphology was observed with a phase contrast microscope (AMEX1200, Thermo Fisher Scientific, Inc.) was used to observe the change of cellular morphology.

Transient transfection

A549 and RLE-6TN cells were cultured in 6-well culture plates as described above and divided into dimethyl sulfoxide groups (including the control, sicontrol, siHIF-1α and siLOX groups) and PQ groups (including the control + PQ, sicontrol + PQ, siHIF-1α + PQ and siLOX + PQ groups). HIF-1α and LOX short interfering (si)RNAs and negative control sequences were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China) and are listed in Table II. For transfection of each siRNA, 4 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) were incubated with 100 pmol siRNA or negative control sequences in 500 µl Opti-MEM medium (Gino Biomedical Technology Co., Ltd., Hangzhou, China) for 20 min at RT. Cells were transfected by replacing the medium with 2 ml Opti-MEM medium containing the siRNA or negative control sequences and Lipofectamine® 2000, and then incubating them at 37°C in a humidified atmosphere of 5% CO2 for 6 h. The Opti-MEM medium was then replaced with 2 ml fresh culture medium. Subsequently, the cells in the PQ groups were incubated with PQ for 24 h and the other cells were treated with phosphate buffered saline. The total time from the start of transfection to subsequent experimentation was 48 h.

Table II.

Sequences of siRNAs used for transfection.

Table II.

Sequences of siRNAs used for transfection.

SpeciessiRNASequence (5′-3′)
HumanHIF-1αF: GCC GAG GAA GAA CUA UGA ATT
R: UUC AUA GUU CUU CCU CGG CTT
LOXF: CAG GCG AUU UGC AUG UAC UTT
R: AGU ACA UGC AAA UCG CCU GTT
RatHIF-1αF: GGG CCG UUC AAU UUA UGA ATT
R: UUC AUA AAU UGA ACG GCC CTT
LOXF: CCG GAU GUU AUG AUA CUU ATT
R: UAA GUA UCA UAA CAU CCG GTT

[i] Si, silencing; HIF, hypoxia-inducible factor; LOX, lysyl oxidase; F, forward; R, reverse.

Statistical analyses

Data were analyzed using SPSS (version 16.0; SPSS, Inc., Chicago, IL, USA) and expressed as the mean + standard deviation of triplicate experiments. Comparisons between two groups were performed using a Student's t-test and comparisons of multiple groups were performed using one-way analysis of variance and Dunnett's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

HIF-1α and LOX may regulate PQ-induced EMT

PQ treatment induced a significant decrease in E-cadherin expression and significantly increased α-SMA expression as determined by western blotting (Fig. 1A), which confirmed that EMT participated in PQ-induced PF. Protein levels of HIF-1α and LOX were significantly increased in the PQ groups compared with the control groups (Fig. 1B). Based on the immunofluorescence staining, the levels of HIF-1α and LOX were markedly increased with 24 h of treatment with PQ compared with control groups (Fig. 1C). These results suggested that EMT served an important role in PQ-induced PF, and that HIF-1α and LOX may regulate EMT following PQ poisoning.

HIF-1α may promote EMT by upregulating LOX expression

The levels of HIF-1α, LOX- and EMT-related markers in PQ-poisoned A549 and RLE-6TN cells were measured following HIF-1α silencing to determine the potential roles of HIF-1α and LOX in PQ-induced EMT. HIF-1α mRNA expression was significantly decreased in the siHIF-1α + PQ group compared with the sicontrol + PQ group (Fig. 2A). The expression of EMT markers was reversed following HIF-1α silencing, as α-SMA expression decreased and E-cadherin expression increased (Fig. 2B). In addition, phase-contrast microscopy revealed that the morphology of cells in the PQ groups changed from a polygon to fusiform morphology compared with the control group. However, these changes were alleviated following HIF-1α silencing (Fig. 2C). The level of LOX mRNA was significantly decreased in the siHIF-1α + PQ group compared with the sicontrol + PQ group (Fig. 2D). The protein expression of LOX was reduced in the siHIF-1α + PQ group compared with the sicontrol + PQ group (Fig. 2E). Therefore, HIF-1α may have an important function in modulating PQ-induced EMT by inducing LOX expression.

LOX promotes PQ-induced EMT independently from HIF-1α

Levels of HIF-1α, LOX and EMT markers in PQ-poisoned cells following LOX silencing were subsequently determined. The level of LOX mRNA was significantly decreased in the siLOX group compared with the sicontrol group and in the siLOX + PQ group compared with the sicontrol + PQ group (Fig. 3A). The expression of EMT markers was also reversed following LOX silencing, as α-SMA expression decreased, and E-cadherin increased (Fig. 3B). In addition, phase-contrast microscopy revealed that the morphological changes (the degree of fusiformity was reduced) observed in cells in the PQ groups were alleviated following LOX silencing (Fig. 3C). However, the expression of HIF-1α mRNA was not significantly changed in the siLOX + PQ group compared with the sicontrol + PQ group (Fig. 3D). Levels of HIF-1α protein were also not significantly decreased following LOX expression inhibition (Fig. 3E). These findings suggest that LOX may promote PQ-induced EMT, but it does not regulate HIF-1α expression.

HIF-1α may regulate PQ-induced EMT through the LOX/β-catenin pathway

Changes in the levels of β-catenin were detected following HIF-1α (LOX) inhibition in vitro to further reveal the interactions between HIF-1α, LOX and β-catenin. β-catenin mRNA levels in the PQ groups were significantly decreased following HIF-1α (LOX) silencing (Fig. 4A and B). Similar results were observed for the protein expression of β-catenin (Fig. 4C and D). These findings suggest that HIF-1α may modulate PQ-induced EMT via the LOX/β-catenin pathway.

Discussion

PQ accumulates in the lungs and eventually leads to PF; however, its molecular mechanisms are complex and remain unclear (24,7). EMT is known to have an important function in PF (4,7,23). As demonstrated in recent studies by the present authors, EMT occurs in PQ-induced PF and may be modulated by HIF-1α or LOX (8,9). However, the interaction between HIF-1α and LOX remains unclear. Therefore, the association between HIF-1α and LOX was investigated, as was the pathway that regulates PQ-induced EMT. It was demonstrated that HIF-1α may modulate PQ-induced EMT via the LOX/β-catenin pathway.

LOX is a downstream target gene of HIF-1α, and a number of previous gene profiling studies have confirmed that LOX expression is upregulated by HIF-1α (2427). LOX is also an important regulator of hypoxia-induced tumor progression in a variety of cancers via a HIF-1α-dependent mechanism (19,28). However, the correlation between HIF-1α and LOX in fibrosis remains unexplored. In the present study, HIF-1α and LOX expression were significantly increased in the model of PQ-induced PF. E-cadherin expression was decreased, and α-SMA expression was significantly increased following PQ treatment, which confirmed that HIF-1α, LOX and EMT are associated with PQ-induced PF. Furthermore, HIF-1α silencing downregulated the expression of LOX mRNA and protein. The expression of EMT markers was also reversed following HIF-1α silencing, as α-SMA expression decreased, and E-cadherin expression increased. In addition, changes in cellular morphology were alleviated following HIF-1α silencing, which indicated that the degree of PQ-induced EMT was alleviated following HIF-1α silencing. Therefore, HIF-1α serves an important role in modulating EMT by activating LOX in PQ-induced PF. This result is consistent with findings from previous studies, which demonstrated that HIF-1α promotes EMT by upregulating LOX expression in ovarian and renal cancers (27,29). It was also confirmed that LOX may be a target of HIF-1α in PQ-induced PF and in tumors by modulating EMT.

In addition to acting as a HIF-1α-responsive gene, LOX may have more complex functions. According to a previous study by Pez et al (21), LOX and HIF-1α act synergistically to promote colon cancer cell proliferation and tumor formation. As previously demonstrated by Ji et al (20), LOX silencing downregulates the protein expression of HIF-1α in epithelial ovarian cancer cells. These findings indicated that LOX and HIF-1α may bidirectionally regulate PQ-induced EMT. However, in the present study, LOX silencing did not induce changes in the protein and mRNA levels of HIF-1α. However, the expression of EMT markers was ameliorated following LOX silencing. In addition, changes in cellular morphology were alleviated following LOX silencing. Therefore, the degree of PQ-induced EMT was alleviated following LOX silencing in vitro. This finding is consistent with other previously published results (27,29) which reported that LOX inhibition did not prevent HIF-1α upregulation. Furthermore, LOX is only an intermediate signaling molecule that mediates HIF-1α-promoted PQ-induced EMT.

β-catenin is a protein located in cytoplasmic plaques that serves a major role in EMT. β-catenin has been used as a marker of EMT in a number of studies of embryonic development, cancer, and fibrosis (3033). According to previous studies, β-catenin is associated with EMT during renal fibrosis (34) and fibrosis in other organs (35,36). In addition, β-catenin participates in the development of PF by transforming A549 cells into fibroblasts (23,37). As demonstrated previously, HIF-1α is positively correlated with β-catenin in rat models, and HIF-1α regulates EMT through the β-catenin pathway (9,38). β-catenin mRNA and protein levels were significantly decreased when HIF-1α and LOX were silenced in the present study, which suggests that HIF-1α regulates PQ-induced EMT through the LOX/β-catenin pathway.

The present study aimed to research the role of EMT in the development of PQ-induced pulmonary fibrosis. A549 cells retain the feature of type II alveolar epithelial cells even though they are a type of cancer cell. RLE-6TN cells were type II rat alveolar epithelial cells. These two cell types are widely used to study the mechanism of pulmonary fibrosis, therefore they were each selected for use within the present study to give a more comprehensive investigation. In the present study it was confirmed that EMT served a role in PQ-induced pulmonary fibrosis and may be modulated by HIF-1α or LOX. HIF-1α may modulate PQ-induced EMT via the LOX/β-catenin pathway.

In conclusion, HIF-1α unidirectionally upregulates LOX expression in PQ-induced EMT. The mechanism may be associated with HIF-1α-induced LOX expression, which subsequently increases β-catenin levels, induces EMT and ultimately leads to the development of PQ-induced PF. Therefore, HIF-1α may be a potential target for restraining the development and exacerbation of PF induced by PQ.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81602873 and 81502829) and the Key and Weak Subject Construction Project of the Shanghai Health and Family Planning System (grant no. 2016ZB0205).

Glossary

Abbreviations

Abbreviations:

EMT

epithelial-to-mesenchymal transition

HIF-1α

hypoxia-inducible factor-1α

LOX

lysyl oxidase

PF

pulmonary fibrosis

PQ

paraquat

α-SMA

α-smooth muscle actin

References

1 

Gil HW, Hong JR, Jang SH and Hong SY: Diagnostic and therapeutic approach for acute paraquat intoxication. J Korean Med Sci. 29:1441–1449. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Xu L, Xu J and Wang Z: Molecular mechanisms of paraquat-induced acute lung injury: A current review. Drug Chem Toxicol. 37:130–134. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Dinis-Oliveira RJ, Duarte JA, Sanchez-Navarro A, Remiao F, Bastos ML and Carvalho F: Paraquat poisonings: Mechanisms of lung toxicity, clinical features, and treatment. Crit Rev Toxicol. 38:13–71. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Nieto MA, Huang RY, Jackson RA and Thiery JP: EMT: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Stone RC, Pastar I, Ojeh N, Chen V, Liu S, Garzon KI and Tomic-Canic M: Epithelial-mesenchymal transition in tissue repair and fibrosis. Cell Tissue Res. 365:495–506. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Bartis D, Mise N, Mahida RY, Eickelberg O and Thickett DR: Epithelial-mesenchymal transition in lung development and disease: Does it exist and is it important? Thorax. 69:760–765. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Kage H and Borok Z: EMT and interstitial lung disease: A mysterious relationship. Curr Opin Pulm Med. 18:517–523. 2012.PubMed/NCBI

8 

Wang J, Zhu Y, Tan J, Meng X, Xie H and Wang R: Lysyl oxidase promotes epithelial-to-mesenchymal transition during paraquat-induced pulmonary fibrosis. Mol Biosyst. 12:499–507. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zhu Y, Tan J, Xie H, Wang J, Meng X and Wang R: HIF-1alpha regulates EMT via the Snail and beta-catenin pathways in paraquat poisoning-induced early pulmonary fibrosis. J Cell Mol Med. 20:688–697. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Masoud GN and Li W: HIF-1alpha pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 5:378–389. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Balamurugan K: HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer. 138:1058–1066. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Zhou G, Dada LA, Wu M, Kelly A, Trejo H, Zhou Q, Varga J and Sznajder JI: Hypoxia-induced alveolar epithelial-mesenchymal transition requires mitochondrial ROS and hypoxia-inducible factor 1. Am J Physiol Lung Cell Mol Physiol. 297:L1120–1130. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Darby IA and Hewitson TD: Hypoxia in tissue repair and fibrosis. Cell Tissue Res. 365:553–562. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Wang RL, Tang X, Wu X, Xu R, Yu KL and Xu K: The relationship between HIF-1α expression and the early lung fibrosis in rats with acute paraquat poisoning. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 30:273–277. 2012.(In Chinese). PubMed/NCBI

15 

Cox TR, Bird D, Baker AM, Barker HE, Ho MW, Lang G and Erler JT: LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res. 73:1721–1732. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Lopez B, Gonzalez A, Hermida N, Valencia F, de Teresa E and Diez J: Role of lysyl oxidase in myocardial fibrosis: From basic science to clinical aspects. Am J Physiol Heart Circ Physiol. 299:H1–9. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Deng S, Jin T, Zhang L, Bu H and Zhang P: Mechanism of tacrolimus-induced chronic renal fibrosis following transplantation is regulated by ox-LDL and its receptor, LOX-1. Mol Med Rep. 14:4124–4134. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Cheng T, Liu Q, Zhang R, Zhang Y, Chen J, Yu R and Ge G: Lysyl oxidase promotes bleomycin-induced lung fibrosis through modulating inflammation. J Mol Cell Biol. 6:506–515. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Erler JT, Bennewith KL, Nicolau M, Dornhöfer N, Kong C, Le QT, Chi JT, Jeffrey SS and Giaccia AJ: Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 440:1222–1226. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Ji F, Wang Y, Qiu L, Li S, Zhu J, Liang Z, Wan Y and Di W: Hypoxia inducible factor 1α-mediated LOX expression correlates with migration and invasion in epithelial ovarian cancer. Int J Oncol. 42:1578–1588. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Pez F, Dayan F, Durivault J, Kaniewski B, Aimond G, Le Provost GS, Deux B, Clézardin P, Sommer P, Pouysségur J and Reynaud C: The HIF-1-inducible lysyl oxidase activates HIF-1 via the Akt pathway in a positive regulation loop and synergizes with HIF-1 in promoting tumor cell growth. Cancer Res. 71:1647–1657. 2011. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Elvidge GP, Glenny L, Appelhoff RJ, Ratcliffe PJ, Ragoussis J and Gleadle JM: Concordant regulation of gene expression by hypoxia and 2-oxoglutarate-dependent dioxygenase inhibition: The role of HIF-1alpha, HIF-2alpha, and other pathways. J Biol Chem. 281:15215–15226. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Goto TM, Arima Y, Nagano O and Saya H: Lysyl oxidase is induced by cell density-mediated cell cycle suppression via RB-E2F1-HIF-1α axis. Cell Struct Funct. 38:9–14. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Yang X, Li S, Li W, Chen J, Xiao X, Wang Y, Yan G and Chen L: Inactivation of lysyl oxidase by β-aminopropionitrile inhibits hypoxia-induced invasion and migration of cervical cancer cells. Oncol Rep. 29:541–548. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Wang Y, Ma J, Shen H, Wang C, Sun Y, Howell SB and Lin X: Reactive oxygen species promote ovarian cancer progression via the HIF-1α/LOX/E-cadherin pathway. Oncol Rep. 32:2150–2158. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Reynaud C, Ferreras L, Di Mauro P, Kan C, Croset M, Bonnelye E, Pez F, Thomas C, Aimond G, Karnoub AE, et al: Lysyl oxidase is a strong determinant of tumor cell colonization in bone. Cancer Res. 77:268–278. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Schietke R, Warnecke C, Wacker I, Schödel J, Mole DR, Campean V, Amann K, Goppelt-Struebe M, Behrens J, Eckardt KU and Wiesener MS: The lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: Insights into cellular transformation processes mediated by HIF-1. J Biol Chem. 285:6658–6669. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Zeisberg M and Neilson EG: Biomarkers for epithelial-mesenchymal transitions. J Clin Invest. 119:1429–1437. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Xu L, Cui WH, Zhou WC, Li DL, Li LC, Zhao P, Mo XT, Zhang Z and Gao J: Activation of Wnt/β-catenin signalling is required for TGF-β/Smad2/3 signalling during myofibroblast proliferation. J Cell Mol Med. 21:1545–1554. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Ji S, Deng H, Jin W, Yan P, Wang R, Pang L, Zhou J, Zhang J, Chen X, Zhao X and Shen J: Beta-catenin participates in dialysate-induced peritoneal fibrosis via enhanced peritoneal cell epithelial-to-mesenchymal transition. FEBS Open Bio. 7:265–273. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Wang X, Dai W, Wang Y, Gu Q, Yang D and Zhang M: Blocking the Wnt/β-catenin pathway by lentivirus-mediated short hairpin RNA targeting β-catenin gene suppresses silica-induced lung fibrosis in mice. Int J Environ Res Public Health. 12:10739–10754. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Martinez-Martinez E, Ibarrola J, Calvier L, Fernandez-Celis A, Leroy C, Cachofeiro V, Rossignol P and Lopez-Andres N: Galectin-3 blockade reduces renal fibrosis in two normotensive experimental models of renal damage. PLoS One. 11:e01662722016. View Article : Google Scholar : PubMed/NCBI

35 

Hu BL, Shi C, Lei RE, Lu DH, Luo W, Qin SY, Zhou Y and Jianga HX: Interleukin-22 ameliorates liver fibrosis through miR-200a/beta-catenin. Sci Rep. 6:364362016. View Article : Google Scholar : PubMed/NCBI

36 

Lin JC, Kuo WW, Baskaran R, Chen MC, Ho TJ, Chen RJ, Chen YF, Vijaya Padma V, Lay IS and Huang CY: Enhancement of beta-catenin in cardiomyocytes suppresses survival protein expression but promotes apoptosis and fibrosis. Cardiol J. 24:195–205. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Anastas JN and Moon RT: WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer. 13:11–26. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Xie H, Tan JT, Wang RL, Meng XX, Tang X and Gao S: Expression and significance of HIF-1alpha in pulmonary fibrosis induced by paraquat. Exp Biol Med (Maywood). 238:1062–1068. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 15 Issue 3

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
Lu J, Qian Y, Jin W, Tian R, Zhu Y, Wang J, Meng X and Wang R: Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase. Exp Ther Med 15: 2287-2294, 2018
APA
Lu, J., Qian, Y., Jin, W., Tian, R., Zhu, Y., Wang, J. ... Wang, R. (2018). Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase. Experimental and Therapeutic Medicine, 15, 2287-2294. https://doi.org/10.3892/etm.2017.5677
MLA
Lu, J., Qian, Y., Jin, W., Tian, R., Zhu, Y., Wang, J., Meng, X., Wang, R."Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase". Experimental and Therapeutic Medicine 15.3 (2018): 2287-2294.
Chicago
Lu, J., Qian, Y., Jin, W., Tian, R., Zhu, Y., Wang, J., Meng, X., Wang, R."Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase". Experimental and Therapeutic Medicine 15, no. 3 (2018): 2287-2294. https://doi.org/10.3892/etm.2017.5677