Open Access

Nrf2 overexpression protects against paraquat‑induced A549 cell injury primarily by upregulating P‑glycoprotein and reducing intracellular paraquat accumulation

  • Authors:
    • Bin Wu
    • Hai‑Xiao Li
    • Jie Lian
    • Yong‑Jie Guo
    • Ya‑Hui Tang
    • Zi‑Juan Chang
    • Lu‑Feng Hu
    • Guang‑Ju Zhao
    • Guang‑Liang Hong
    • Zhong‑Qiu Lu
  • View Affiliations

  • Published online on: December 4, 2018     https://doi.org/10.3892/etm.2018.7044
  • Pages: 1240-1247
  • Copyright: © Wu 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

Paraquat (PQ) intoxication causes thousands of mortalities every year, worldwide. Its pulmonary‑targeted accumulation and the acute lung injury it subsequently causes, remain a challenge for detoxification treatment. A previous study has demonstrated that the upregulation of nuclear factor erythroid‑2 related factor 2 (Nrf2) prevents PQ toxicity in cell line and murine models. As Nrf2 target genes include a group of membrane transporters, the current study assessed the protective mechanism exerted by Nrf2 against PQ toxicity and intracellular PQ accumulation via its effects on P‑glycoprotein (P‑gp), a downstream transporter of Nrf2. Adenovirus vectors containing the Nrf2 gene were transfected into A549 cells. Cell proliferation was assessed by Cell Counting Kit‑8. The levels of LDH, MDA, SOD, TNF‑α, IL‑6 levels were detected using their respective ELISA kits. In addition, the levels of Nrf2 and P‑gp protein expression were detected by western blot analysis. The concentration of PQ was measured by HPLC. The results revealed that overexpressed Nrf2 significantly increased P‑gp protein levels, decreased the intracellular accumulation of PQ and attenuated PQ‑induced toxicity. However, the protective effects of Nrf2 overexpression on PQ‑challenged A549 cells were abrogated following cyclosporine A treatment, a competitive inhibitor of P‑gp, which also increased intracellular PQ levels. These data indicated that Nrf2 gene overexpression prevented PQ toxicity in A549 cells, potentially via the upregulation of P‑gp activity and the inhibition of intracellular PQ accumulation. Thus, Nrf2 and P‑gp may serve as potential therapeutic targets for the treatment of PQ‑induced injury.

Introduction

Paraquat (N,N′-dimethyl-4,4′-bipyridinium dichloride; PQ) is one of the most commonly used herbicides worldwide (1,2). However, long-term accumulation or acute intoxication of PQ may cause organ injury, resulting in a mortality rate of >60–70% (3,4). Following oral administration, PQ is absorbed through the gastrointestinal tract within 1–2 h and accumulates in various organs, including the lung, liver, kidney and central nervous system (5). PQ primarily amasses within alveolar epithelial and bronchiolar Clara cells of the lung, and results in acute lung injury and acute respiratory distress syndrome, which is the leading cause of mortality in patients with PQ poisoning (6). Although the exact pathogenic mechanism of PQ remains largely unknown, PQ-induced pulmonary-specific accumulation, excessive oxidative stress, inflammatory injury and an imbalance in the deposition of the extracellular matrix, are major contributors to lung injury following PQ intoxication (79). However, pulmonary-targeted accumulation, which results in acute lung injury, remains a challenge for detoxification treatment.

P-glycoprotein (P-gp), a member of the ATP-binding cassette (ABC) transporter family, is vital for many cellular processes that require the transport of chemicals across the cell membrane (10,11). P-gp hyperactivity causes drugs to be pumped out of cells, resulting in chemotherapeutic agent and antimicrobial drug resistance (12,13). Previous studies have demonstrated that P-gp may be closely associated with the removal of PQ from cells (14,15). In corroboration with this association, P-gp levels are significantly increased in rat alveolar type II cells following PQ exposure (16) and a high level of P-gp has been revealed to alleviate PQ toxicity in human epithelial colon cancer cells and to significantly reduce rat lung tissue PQ concentration (17,18). However, these protective effects were reversed following treatment with a specific inhibitor of P-gp, cyclosporine A (CsA) (18). Therefore, P-gp appears to serve an important role in PQ transport across cells and upregulating the activity of P-gp may be an effective measure to attenuate the intracellular accumulation of PQ and thus, its toxicity.

Nuclear factor erythroid-2 related factor 2 (Nrf2) is a transcription factor that belongs to the cap ‘n’ collar family of basic leucine zipper proteins, which is tightly regulated by Kelch-like ECH-associated protein 1 (Keap1) (19). The Keap1-Nrf2 pathway, including its targeted cytoprotective protein expression, is the fundamental mechanism for cellular defence against oxidative and electrophilic stress (20,21). Various natural compounds, including cycloartenyl ferulate and resveratrol, inhibit PQ-induced oxidative stress and apoptosis by activating the Nrf2 pathway (22,23). A previous study demonstrated that Nrf2 overexpression attenuated PQ toxicity in A549 cells and mice by activating heme oxygenase-1 and NAD(P)H: Quinone oxidoreductase 1 (24). The primary focus of research on Nrf2 targets has been on detoxifying/antioxidant enzymes; however, several ABC transporters are Nrf2 targets (25). Furthermore, previous studies have revealed that the activation of Nrf2 is necessary to increase P-gp activity (2628). Therefore, the current study hypothesized that Nrf2 may prevent organ injury in PQ poisoning by increasing P-gp activation and reducing intracellular PQ concentration.

The present study revealed that the Nrf2 gene was overexpressed in the A549 cell line. Subsequent to this result, the role of P-gp activation in PQ-challenged A549 cells was determined. It was hypothesized that Nrf2 and P-gp activity may be potential targets for the prevention of organ injury in PQ poisoning.

Materials and methods

Materials and reagents

PQ and the adenoviral (AD) system, including plasmids containing the predesigned human Nrf2 gene and fetal bovine serum (FBS), were purchased from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). RPMI-1640 medium was obtained from Gibco; Thermo Fisher Scientific, Inc. (Waltham MA, USA). A549 cells were purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). The Cell Counting Kit-8 (CCK-8) was obtained from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) detection kits were supplied by Nanjing Jiancheng Bioengineering Institute (Nanjing, China). Antibodies against Nrf2 and P-gp were obtained from Abcam (Cambridge, UK). GFP was purchased from Thermo Fisher Scientific, Inc. CsA (cat. no. 59865-13-3) was purchased from Sigma-Aldrich; Merck, KGaA.

Transfection and transfection efficiency analysis

A549 cells were grown in a monolayer culture in RPMI-1640 medium supplemented with 10% FBS at 37°C in a humidified atmosphere with 5% CO2. Following this incubation, cells (4×104 cells/well) were inoculated onto a 96-well culture plate and left to grow in the logarithmic stage for 24 h at 37°C. A549 cells were transfected with adenoviral vectors containing either AD-Nrf2 or AD-(GFP) (cat. no. PEP033; Thermo Fisher Scientific, Inc.), at a multiplicity of infection of 50. Control cells were cultured in RPMI-1640 medium only. The cells were cultured in RPMI-1640 medium supplemented with 10% FBS and maintained at 37°C in a 5% CO2-humidified incubator for 24 h. A fluorescence microscope (magnification, ×100; Olympus CKX41SF; Olympus Corporation, Tokyo, Japan) was used to determine the percentage of GFP synthesizing cells. The ratio of cells that emitted green fluorescence in the same field of view was regarded as the transfection efficiency. Nrf2 overexpression was also confirmed via western blotting.

Cell culture and treatments

The current study included five groups: A control group, a PQ group, a PQ + AD-Nrf2 group, a PQ + AD-Nrf2 + CsA group and a control + CsA group. A549 cells were grown in a monolayer culture in RPMI-1640 medium supplemented with 10% FBS and maintained at 37°C in a 5% CO2-humidified incubator. A549 cells (4×104 cells/well) were seeded onto a 96-well culture plate. Following logarithmic stage growth for 24 h at 37°C, A549 cells were transfected with AD-Nrf2 and preincubated with CsA at a concentration of 12 µg/ml or vehicle control (0.1% ethanol). Cells were subsequently incubated in RPMI-1640 medium supplemented with 10% FBS and cultured at 37°C in a 5% CO2-humidified incubator for 24 h and the experimental groups were administered 10 µl PQ (1×103 mol/l). Following further incubation for 24 h at 37°C, cells were harvested and used for subsequent experimentation.

CCK-8 assay

Cell survival was assessed using a CCK-8 kit, according to the manufacturer's protocol and viable cell density was adjusted to 4×104 cells/ml. A549 cells (4×104 cells/well) were seeded onto a 96-well culture plate and cultured in RPMI-1640 medium supplemented with 10% FBS and maintained at 37°C in a 5% CO2-humidified incubator for 24 h. Growth medium was replaced with serum-free medium and cells were cultured for a further 24 h. Subsequently, the supernatant was discarded and 10 µl CCK-8 solution was added to each well. Following incubation for 1 h at 37°C, absorbance was measured at 450 nm using a Benchmark Plus Microplate Spectrophotometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

LDH release assay

The LDH release was examined using a LDH release assay, according to the manufacturer's protocol. The release of LDH was expressed as a percentage of the total LDH quantity in cells treated with 2% Triton X-100.

MDA, SOD, TNF-α and IL-6 detection

The cells were centrifuged at 14,000 × g for 15 min at 4°C and the supernatant was transferred to centrifuge tubes. The levels of MDA (cat. no. A003-1), SOD (cat. no. A001-3), TNF-α (cat. no. H052) and IL-6 (cat. no. H007) in cell supernatant was detected using their respective ELISA kits, according to the manufacturer's protocol. Samples were analyzed using a spectrophotometer. TNF-α, IL-6 and SOD activities were expressed in pg/ml and MDA levels were expressed as nmol/ml.

Western blot analysis

Total protein was extracted from A549 cells using radioimmunoprecipitation assay (RIPA) buffer (Beyotime Institute of Biotechnology, Haimen, China) and incubated on ice for 30 min. Cell lysates were centrifuged for 20 min at 16,000 × g at 4°C. Total protein was quantified using a bicinchoninic acid assay and 20 µg protein/lane was separated via SDS-PAGE on a 10% gel. The separated proteins were transferred onto nitrocellulose membranes (Thermo Fisher Scientific, Inc.) and blocked with 5% non-fat milk for 1 h at room temperature. The membranes were incubated with primary antibodies against Nrf2 (1:1,000; cat. no. ab62352), P-gp (1:1,000; cat. no. ab170904) or GAPDH (1:5,000; cat. no. ab8245; all Abcam, Cambridge, UK) antibodies overnight at 4°C. Membranes were washed with Tris-buffered saline Tween® 20. Following primary incubation, the membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit (1:2,000; cat. no. 14708) or mouse (1:2,000; cat. no. 14709; both Cell Signaling Technology, Inc., Danvers, MA, USA) secondary antibodies for 2 h at room temperature. Protein bands were visualized using the chemiluminescence reagent (Thermo Fisher Scientific, Inc.). Quantitative analysis was performed using Image J software (Version 1.8.0_172; National Institutes of Health, Bethesda, MD, USA). GAPDH was utilized as the loading control.

High performance liquid chromatography (HPLC) analysis

Transfected A549 cells exposed to PQ were harvested. Following repeated freezing and thawing, cells were centrifuged at 16,000 × g for 5 min at 4°C. Cell supernatant (200 µl) was subsequently collected and treated with an equivalent volume of acetonitrile (200 µl; cat. no. A0793; TCI Development Co., Ltd., Shanghai, China). Following centrifugation at 12,000 × g for 15 min at 4°C, 200 µl cell supernatant was filtered through an organic solvent-compatible 0.45-µm syringe filter (cat. no. F512545; Sangon Biotech Co., Ltd, Shanghai, China) and 20 µl was injected into an Agilent 1100 series HPLC system (Agilent Technologies, Inc., Santa Clara, CA, USA) equipped with a diode-array UV detector, on-line degasser, autosampler, thermostat-columned compartment and quaternary pump. Samples were eluted isocratically with an isocratic elution composed of 4% mobile phase A (acetonitrile) and 96% mobile phase B (20 mM sodium dihydrogen phosphate, 0.4 mM sodium heptanesulfonate and adjusted to pH 2.3) at a flow rate of 0.6 ml/min at 30°C. Separation was performed using an Agilent Zorbax-SB-Aq column (internal diameter, 4.6 mm; length, 250 mm; Agilent Technologies, Inc.) and results were detected at a wavelength of 256 nm.

Statistical analysis

Experiments were performed at least three times and data were presented as the mean ± standard deviation. Differences between three or more groups were analyzed using one-way analysis of variance followed by the least significant difference test. P<0.05 was considered to indicate a statistically significant result.

Results

Transfection efficiency analysis

The results of transfection efficacy analysis verified that the control group did not express green fluorescence (Fig. 1A). From the green fluorescence observed under a magnification of ×100, AD vector transfection efficiency was determined to be >90% (data not shown). The results of western blotting revealed that Nrf2 expression in cells transfected with AD-Nrf2 was significantly higher compared with the control group (P<0.001; Fig. 1B), which demonstrated that the cell model that was abundant for Nrf2 expression, and that the use of AD vectors was successful.

Cell viability

The results demonstrated that PQ exposure significantly reduced A549 cell viability (P<0.001; Fig. 2A) and that PQ-induced cytotoxicity was alleviated following Nrf2 treatment (P<0.001; Fig. 2A). However, treatment with CsA significantly reversed the protective effects of Nrf2 on cell viability following PQ exposure (P<0.01; Fig. 2A). No significant differences were identified between the control and the control + CsA group.

LDH activity

The LDH activity of A549 cells was assessed to determine PQ-induced cell injury. The LDH activity of PQ-exposed cells was significantly increased when compared with the control group (P<0.001; Fig. 2B). However, treatment with Nrf2 reversed the PQ induced increase of LDH activity (P<0.01; Fig. 2B). Furthermore, LDH activity significantly increased in cells of the PQ + AD-Nrf2 + CsA-treated group compared with those of the PQ + AD-Nrf2 group (P<0.01; Fig. 2B). No significant differences were identified between the control and the control + CsA group.

SOD and MDA

Compared with the control group, a significant reduction in SOD activity following PQ exposure was observed (P<0.001; Fig. 3A). However, treatment with AD-Nrf2 reversed this effect (P<0.01; Fig. 3A). The SOD activity of the PQ + AD-Nrf2 + CsA-treated group was also significantly decreased compared with those cells exposed to PQ + AD-Nrf2 treatment (P<0.01; Fig. 3A).

Cells of the PQ group exhibited a significant increase in MDA concentration when compared with the control (P<0.001; Fig. 3B). Whereas, the MDA concentration of cells treated with PQ + AD-Nrf2 was significantly reduced when compared with the PQ group (P<0.001; Fig. 3B). However, treatment with CsA reversed the protective effect of AD-Nrf2 (P<0.001; Fig. 3B). No significant differences were identified in SOD activity or MDA concentration between the control and the control + CsA group.

TNF-α and IL-6

When compared with the control group, significant increases in TNF-α and IL-6 were observed in the PQ group (P<0.001; Fig. 3C and D). AD-Nrf2 treatment reversed these increases (TNF-α, P<0.001; IL-6, P<0.01; Fig. 3C and D) and the levels of TNF-α and IL-6 in the PQ + AD-Nrf2 + CsA-treated group were significantly increased compared with cells exposed to PQ + AD-Nrf2 (P<0.01; Fig. 3C and D). No significant differences were identified in the levels of TNF-α or IL-6 expression between the control and the control + CsA group.

P-gp protein expression

Treatment with PQ induced a significant decrease of P-gp protein levels when compared with the control group (Fig. 4A). However, P-gp protein expression increased ~1.9-fold in the AD-Nrf2-treated group compared with the PQ group (P<0.001; Fig. 4B). Treatment with CsA was demonstrated to reverse the increase in P-gp expression in the PQ + AD-Nrf2 + CsA group compared with the PQ + AD-Nrf2 group (P<0.001; Fig. 4B). No significant differences were identified in the level of P-gp protein expression in the control group compared with the control + CsA group.

PQ concentration

The concentration of PQ in the PQ-treated group was 3.14±0.14 mg/l. Cells that received PQ + AD-Nrf2 treatment exhibited a significant decrease in PQ concentration compared with the PQ group (2.53±0.15; P<0.001; Fig. 5). Furthermore, cells of the PQ + AD-Nrf2 + CsA group significantly reversed the effect of AD-Nrf2 treatment, resulting in a PQ concentration of 2.85±0.22 mg/l (P<0.01; Fig. 5).

Discussion

PQ intoxication and the subsequent selective accumulation of PQ molecules results in multi-organ failure and severe pulmonary injury when in the lung (29). The current study successfully constructed Nrf2-overexpressed A549 cells that exhibited resistance to PQ toxicity. Nrf2 treatment was revealed to upregulate P-gp activity and subsequently reduce the intracellular accumulation of PQ and cell injury. However, these results were reversed following treatment with the specific inhibitor of P-gp (CsA). To the best of our knowledge, this is the first study to assess the role of Nrf2 treatment on P-gp expression and PQ concentration in PQ exposed cells, which may serve as potential therapeutic targets for PQ detoxification.

Nrf2 is a key transcription factor that has been demonstrated to be an effective target for the prevention or treatment of various human diseases, which include cardiovascular diseases, neurodegenerative diseases, neuropsychiatric disorders and cancer (3034). Previous studies have revealed that the upregulation of Nrf2 is critical for cytoprotection against various types of cell injury, which include hydrogen peroxide-induced oxidative stress and inflammation (3537). In another previous study, mifepristone-induced Nrf2 gene overexpression in the lungs ameliorated PQ-induced injury by activating the Nrf2-antioxidant response element (ARE) pathway (38). In addition, silent information regulator 2-related enzyme 1 was demonstrated to trigger the Nrf2/ARE antioxidant pathway and protect against lung injury induced by PQ poisoning (39). These studies have revealed the important role of Nrf2 in the prevention of PQ toxicity. Consistent with these results, the Nrf2 gene-transfected A549 cells of the current study presented resistance to PQ toxicity, as evidenced by an elevated cell viability, a decreased LDH activity, an improved oxidative stress response and an attenuation of inflammation, with decreased TNF-α and IL-6 levels following PQ challenge.

In the present study, it was demonstrated that the overexpressed Nrf2 gene also decreased intracellular PQ concentrations, which indicated that certain membrane transporters may be involved in Nrf2-induced cytoprotection against PQ toxicity. Not including phase-II detoxification enzymes, the activities of Nrf2-regulated phase-III drug transporters represent a cellular mechanism that protects against xenobiotics (40). Maher et al (41) revealed that the activation of the Nrf2 pathway may stimulate the coordinated induction of hepatic multi-drug resistance-associated proteins, which are ATP-dependent efflux transporters that serve an important role in cellular defense against various xenobiotics. The activities of other membrane transporters, including organic cation transporters, are also induced by Nrf2 (42). Since P-gp has been extensively studied and demonstrated to be closely associated with PQ excretion (17,43,44), the present study further assessed whether P-gp served a role in the Nrf2-induced inhibition of PQ accumulation and cytoprotection in PQ-challenged A549 cells.

In the current study, P-gp protein levels significantly increased following Nrf2 overexpression in A549 cells. Upregulated P-gp was also demonstrated to reduce intracellular PQ concentrations, which was ameliorated following treatment with the specific inhibitor of P-gp (CsA). Furthermore, Nrf2-induced cytoprotection against oxidative stress and inflammation was inhibited following CsA treatment. These results indicate that P-gp serves a vital role in PQ transport and in the protective effects of Nrf2 against PQ toxicity. The current study corroborated with several previous studies (38,39). It has been demonstrated that an increased P-gp protein level exerts protective effects against PQ-induced toxicity in vitro and in vivo, by inhibiting the accumulation of intracellular PQ, indicating that PQ may be a P-gp substrate (18). Several other studies have revealed that a decrease of P-gp activity at the blood-brain barrier leads to the increased accumulation of neurotoxicants in the brain (4547).

The present study had several limitations. Experiments of the current study were performed in vitro, meaning that the influences of Nrf2 and P-gp in vivo are unknown. In addition, Nrf2 overexpression in PQ poisoning may also activate other various key efflux transporters excluding P-gp, which include multidrug resistance-associated proteins and organic anion transporting polypeptide 2. Thus, additional studies are required to assess Nrf2 and the role of other transporters in vitro and in vivo.

In summary, the current study demonstrated that the Nrf2-mediated increase of P-gp is an important theoretical pathway against PQ toxicity, which reduces intracellular PQ concentrations. Therefore, the present study provides a novel therapeutic target for PQ accumulation and toxicity within the lung.

Acknowledgements

Not applicable.

Funding

The current study was supported by grants from the Natural Science Foundation of Zhejiang Province (grant no. LQ14H150002), the Core Project of Medicine and Health Care Platform in Zhejiang Province (grant no. 2016RCA021), the Traditional Chinese Medical Project of Zhejiang Province (grant no. 2015ZZ015) and the Key Construction Academic Subject (Medical Innovation) of Zhejiang Province (grant no. 11-CX26).

Availability of data and materials

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

Authors' contributions

GLH and ZQL designed the study. BW prepared the manuscript, participated in experimental design and guided students to complete experiments and statistical anlaysis. HXL, JL, YJG, YHT, ZJC and LFH collected the data. GJZ analyzed the data.

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 

Yao R, Cao Y, He YR, Lau WB, Zeng Z and Liang ZA: Adiponectin attenuates lung fibroblasts activation and pulmonary fibrosis induced by paraquat. PLoS One. 10:e01251692015. View Article : Google Scholar : PubMed/NCBI

2 

Cherukuri H, Pramoda K, Rohini D, Thunga G, Vijaynarayana K, Sreedharan N, Varma M and Pandit V: Demographics, clinical characteristics and management of herbicide poisoning in tertiary care hospital. Toxicol Int. 21:209–213. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Seok SJ, Gil HW, Jeong DS, Yang JO, Lee EY and Hong SY: Paraquat intoxication in subjects who attempt suicide: Why they chose paraquat. Korean J Intern Med. 24:247–251. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Senarathna L, Eddleston M, Wilks MF, Woollen BH, Tomenson JA, Roberts DM and Buckley NA: Prediction of outcome after paraquat poisoning by measurement of the plasma paraquat concentration. QJM. 102:251–259. 2009. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Koepsell H: Polyspecific organic cation transporters: Their functions and interactions with drugs. Trends Pharmacol Sci. 25:375–381. 2004. View Article : Google Scholar : PubMed/NCBI

7 

He X, Wang L, Szklarz G, Bi Y and Ma Q: Resveratrol inhibits paraquat-induced oxidative stress and fibrogenic response by activating the nuclear factor erythroid 2-related factor 2 pathway. J Pharmacol Exp Ther. 342:81–90. 2012. View Article : Google Scholar : PubMed/NCBI

8 

She X, Hong G, Tan J, Zhao G, Li M and Lu Z: The protective effect of ulinastatin on paraquat-induced injury in HK-2 cells and the underlying mechanisms. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 33:501–506. 2015.(In Chinese). PubMed/NCBI

9 

Yao R, Zhou Y, He Y, Jiang Y, Liu P, Ye L, Zheng Z, Lau WB, Cao Y and Zeng Z: Adiponectin protects against paraquat-induced lung injury by attenuating oxidative/nitrative stress. Exp Ther Med. 9:131–136. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Kimura M, Yamamoto T, Zhang J, Itoh K, Kyo M, Kamiya T, Aburatani H, Katsuoka F, Kurokawa H, Tanaka T, et al: Molecular basis distinguishing the DNA binding profile of Nrf2-Maf heterodimer from that of Maf homodimer. J Biol Chem. 282:33681–33690. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Aleksunes LM and Manautou JE: Emerging role of Nrf2 in protecting against hepatic and gastrointestinal disease. Toxicol Pathol. 35:459–473. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Zhou SF: Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition. Xenobiotica. 38:802–832. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Bosquillon C: Drug transporters in the lung-do they play a role in the biopharmaceutics of inhaled drugs? J Pharm Sci. 99:2240–2255. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Lacher SE, Gremaud JN, Skagen K, Steed E, Dalton R, Sugden KD, Cardozo-Pelaez F, Sherwin CM and Woodahl EL: Absence of P-glycoprotein transport in the pharmacokinetics and toxicity of the herbicide paraquat. J Pharmacol Exp Ther. 348:336–345. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Huang H, Lu-Bo Y and Haddad GG: A Drosophila ABC transporter regulates lifespan. PLoS Genet. 10:e10048442014. View Article : Google Scholar : PubMed/NCBI

16 

Lehmann T, Kohler C, Weidauer E, Taege C and Foth H: Expression of MRP1 and related transporters in human lung cells in culture. Toxicology. 167:59–72. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Silva R, Palmeira A, Carmo H, Barbosa DJ, Gameiro M, Gomes A, Paiva AM, Sousa E, Pinto M, Bastos Mde L and Remião F: P-glycoprotein induction in Caco-2 cells by newly synthetized thioxanthones prevents paraquat cytotoxicity. Arch Toxicol. 89:1783–1800. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Dinis-Oliveira RJ, Remiao F, Duarte JA, Ferreira R, Sanchez Navarro A, Bastos ML and Carvalho F: P-glycoprotein induction: An antidotal pathway for paraquat-induced lung toxicity. Free Radic Biol Med. 41:1213–1224. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Itoh K, Tong KI and Yamamoto M: Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles. Free Radic Biol Med. 36:1208–1213. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD and Yamamoto M: Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 13:76–86. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K and Yamamoto M: Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol. 24:7130–7139. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Hong GL, Liu JM, Zhao GJ, Wang L, Liang G, Wu B, Li MF, Qiu QM and Lu ZQ: The reversal of paraquat-induced mitochondria-mediated apoptosis by cycloartenyl ferulate, the important role of Nrf2 pathway. Exp Cell Res. 319:2845–2855. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Li S, Zhao G, Chen L, Ding Y, Lian J, Hong G and Lu Z: Resveratrol protects mice from paraquat-induced lung injury: The important role of SIRT1 and NRF2 antioxidant pathways. Mol Med Rep. 13:1833–1838. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Lu H, Chang Z, Han W, Wang L and Hong G: Curcumin reduces paraquat-induced oxidative injury in A549 cells by activation of the Nrf2-ARE pathway. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 32:44–49. 2014.(In Chinese). PubMed/NCBI

25 

Adachi T, Nakagawa H, Chung I, Hagiya Y, Hoshijima K, Noguchi N, Kuo MT and Ishikawa T: Nrf2-dependent and -independent induction of ABC transporters ABCC1, ABCC2, and ABCG2 in HepG2 cells under oxidative stress. J Exp Ther Oncol. 6:335–348. 2007.PubMed/NCBI

26 

Cen J, Zhang L, Liu F, Zhang F and Ji B: Long-term alteration of reactive oxygen species led to multidrug resistance in MCF-7 cells. Oxid Med Cell Longev. 2016:70534512016. View Article : Google Scholar : PubMed/NCBI

27 

Wu J, Zhu Y, Li F, Zhang G, Shi J, Ou R, Tong Y, Liu Y, Liu L, Lu L and Liu Z: Spica prunellae and its marker compound rosmarinic acid induced the expression of efflux transporters through activation of Nrf2-mediated signaling pathway in HepG2 cells. J Ethnopharmacol. 193:1–11. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Ghanem CI, Rudraiah S, Bataille AM, Vigo MB, Goedken MJ and Manautou JE: Role of nuclear factor-erythroid 2-related factor 2 (Nrf2) in the transcriptional regulation of brain ABC transporters during acute acetaminophen (APAP) intoxication in mice. Biochem Pharmacol. 94:203–211. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Gawarammana IB and Buckley NA: Medical management of paraquat ingestion. Br J Clin Pharmacol. 72:745–757. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Bai Y, Wang X, Zhao S, Ma C, Cui J and Zheng Y: Sulforaphane protects against cardiovascular disease via Nrf2 activation. Oxid Med Cell Longev. 2015:4075802015. View Article : Google Scholar : PubMed/NCBI

31 

de Oliveira MR, Nabavi SF, Habtemariam S, Erdogan Orhan I, Daglia M and Nabavi SM: The effects of baicalein and baicalin on mitochondrial function and dynamics: A review. Pharmacol Res. 100:296–308. 2015. View Article : Google Scholar : PubMed/NCBI

32 

de Oliveira MR: Phloretin-induced cytoprotective effects on mammalian cells: A mechanistic view and future directions. Biofactors. 42:13–40. 2016.PubMed/NCBI

33 

de Oliveira MR, Nabavi SF, Manayi A, Daglia M, Hajheydari Z and Nabavi SM: Resveratrol and the mitochondria: From triggering the intrinsic apoptotic pathway to inducing mitochondrial biogenesis, a mechanistic view. Biochim Biophys Acta. 1860:727–745. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Oliveira MR, Nabavi SF, Daglia M, Rastrelli L and Nabavi SM: Epigallocatechin gallate and mitochondria-A story of life and death. Pharmacol Res. 104:70–85. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Hichor M, Sampathkumar NK, Montanaro J, Borderie D, Petit PX, Gorgievski V, Tzavara ET, Eid AA, Charbonnier F, Grenier J and Massaad C: Paraquat induces peripheral myelin disruption and locomotor defects: Crosstalk with LXR and wnt pathways. Antioxid Redox Signal. 27:168–183. 2017. View Article : Google Scholar : PubMed/NCBI

36 

de Oliveira MR, de Souza ICC and Furstenau CR: Carnosic acid induces anti-inflammatory effects in paraquat-treated SH-SY5Y cells through a mechanism involving a crosstalk between the Nrf2/HO-1 axis and NF-kB. Mol Neurobiol. 55:890–897. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Shinjo T, Tanaka T, Okuda H, Kawaguchi AT, Oh-Hashi K, Terada Y, Isonishi A, Morita-Takemura S, Tatsumi K, Kawaguchi M and Wanaka A: Propofol induces nuclear localization of Nrf2 under conditions of oxidative stress in cardiac H9c2 cells. PLoS One. 13:e01961912018. View Article : Google Scholar : PubMed/NCBI

38 

Hong GL, Cai QQ, Tan JP, Jiang XZ, Zhao GJ, Wu B, Li MF, Qiu QM and Lu ZQ: Mifepristone-inducible recombinant adenovirus attenuates paraquat-induced lung injury in rats. Hum Exp Toxicol. 34:32–43. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Ding YW, Zhao GJ, Li XL, Hong GL, Li MF, Qiu QM, Wu B and Lu ZQ: SIRT1 exerts protective effects against paraquat-induced injury in mouse type II alveolar epithelial cells by deacetylating NRF2 in vitro. Int J Mol Med. 37:1049–1058. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Xu C, Li CY and Kong AN: Induction of phase III and III drug metabolism/transport by xenobiotics. Arch Pharm Res. 28:249–268. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Maher JM, Dieter MZ, Aleksunes LM, Slitt AL, Guo G, Tanaka Y, Scheffer GL, Chan JY, Manautou JE, Chen Y, et al: Oxidative and electrophilic stress induces multidrug resistance-associated protein transporters via the nuclear factor-E2-related factor-2 transcriptional pathway. Hepatology. 46:1597–1610. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Jeddi F, Soozangar N, Sadeghi MR, Somi MH, Shirmohamadi M, Eftekhar-Sadat AT and Samadi N: Nrf2 overexpression is associated with P-glycoprotein upregulation in gastric cancer. Biomed Pharmacother. 97:286–292. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Zerin T, Kim YS, Hong SY and Song HY: Protective effect of methylprednisolone on paraquat-induced A549 cell cytotoxicity via induction of efflux transporter, P-glycoprotein expression. Toxicol Lett. 208:101–107. 2012. View Article : Google Scholar : PubMed/NCBI

44 

She XR, Tian X, Fan XK, Hong GL, Zhao GJ, Li MF and Lu ZQ: The effects of P-glycoprotein expression induced by ulinastatin on HK-2 cells damage induced by paraquat. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 34:805–809. 2016.(In Chinese). PubMed/NCBI

45 

Bartels AL, Kortekaas R, Bart J, Willemsen AT, de Klerk OL, de Vries JJ, van Oostrom JC and Leenders KL: Blood-brain barrier P-glycoprotein function decreases in specific brain regions with aging: A possible role in progressive neurodegeneration. Neurobiol Aging. 30:1818–1824. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Lee G and Bendayan R: Functional expression and localization of P-glycoprotein in the central nervous system: Relevance to the pathogenesis and treatment of neurological disorders. Pharm Res. 21:1313–1330. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Furuno T, Landi MT, Ceroni M, Caporaso N, Bernucci I, Nappi G, Martignoni E, Schaeffeler E, Eichelbaum M, Schwab M and Zanger UM: Expression polymorphism of the blood-brain barrier component P-glycoprotein (MDR1) in relation to Parkinson's disease. Pharmacogenetics. 12:529–534. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 17 Issue 2

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
Wu B, Li HX, Lian J, Guo YJ, Tang YH, Chang ZJ, Hu LF, Zhao GJ, Hong GL, Lu ZQ, Lu ZQ, et al: Nrf2 overexpression protects against paraquat‑induced A549 cell injury primarily by upregulating P‑glycoprotein and reducing intracellular paraquat accumulation. Exp Ther Med 17: 1240-1247, 2019
APA
Wu, B., Li, H., Lian, J., Guo, Y., Tang, Y., Chang, Z. ... Lu, Z. (2019). Nrf2 overexpression protects against paraquat‑induced A549 cell injury primarily by upregulating P‑glycoprotein and reducing intracellular paraquat accumulation. Experimental and Therapeutic Medicine, 17, 1240-1247. https://doi.org/10.3892/etm.2018.7044
MLA
Wu, B., Li, H., Lian, J., Guo, Y., Tang, Y., Chang, Z., Hu, L., Zhao, G., Hong, G., Lu, Z."Nrf2 overexpression protects against paraquat‑induced A549 cell injury primarily by upregulating P‑glycoprotein and reducing intracellular paraquat accumulation". Experimental and Therapeutic Medicine 17.2 (2019): 1240-1247.
Chicago
Wu, B., Li, H., Lian, J., Guo, Y., Tang, Y., Chang, Z., Hu, L., Zhao, G., Hong, G., Lu, Z."Nrf2 overexpression protects against paraquat‑induced A549 cell injury primarily by upregulating P‑glycoprotein and reducing intracellular paraquat accumulation". Experimental and Therapeutic Medicine 17, no. 2 (2019): 1240-1247. https://doi.org/10.3892/etm.2018.7044