Lipoxin A4 attenuates hyperoxia‑induced lung epithelial cell injury via the upregulation of heme oxygenase‑1 and inhibition of proinflammatory cytokines

  • Authors:
    • Yan‑Yan Luo
    • Sheng‑Hua Wu
    • Hong‑Yan Lu
    • Bing‑Jie Li
    • Shu‑Jun Li
    • Zhong‑Yi Sun
    • Rui Jin
    • Xiao‑Qing Chen
  • View Affiliations

  • Published online on: November 15, 2019     https://doi.org/10.3892/mmr.2019.10821
  • Pages: 429-437
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study examined whether lipoxin A4 (LXA4) increases the expression of HO‑1, and inhibits the production of interleukin 6 (IL‑6) and monocyte chemotactic protein 1 (MCP‑1) in LXA4‑induced protection during hyperoxia‑induced injury in murine lung epithelial cells (MLE‑12) and what signal pathway may participate in the actions of LXA4 inhibiting IL‑6 and MCP‑1. MLE‑12 cells were exposed to air or hyperoxia with or without pretreatment with LXA4, Zinc protoporphyrin IX (ZnPP‑IX), IL‑6, anti‑IL‑6, MCP‑1, anti‑MCP‑1, inhibitors of p38 mitogen‑activated protein kinase (p38 MAPK), protein kinase B (Akt) and extracellular signal‑regulated kinase 1/2 (ERK1/2) signaling pathways. The cell survival rates, cell viability, apoptosis rates, expression of superoxide dismutase (SOD), heme oxygenase‑1 (HO‑1), IL‑6 and MCP‑1, and the activations of p38 MAPK, ERK1/2 and Akt were measured. LXA4 significantly increased the cell survival rates, cell viability, SOD levels and HO‑1 expression, reduced the apoptosis rates, and inhibited the MCP‑1 and IL‑6 levels induced by hyperoxia in cells. ZnPP‑IX, an inhibitor of HO‑1, blocked LXA4‑induced protection on cell viability in cells exposed to hyperoxia. Anti‑IL‑6 and anti‑MCP‑1 improved the cell viability of cells exposed to hyperoxia. Inhibition of p38 MAPK and ERK1/2 blocked the expression of MCP‑1 and IL‑6 induced by hyperoxia. LXA4 inhibited the activation of p38 MAPK and ERK1/2 induced by hyperoxia, and increased the activation of the Akt signaling pathway, which was inhibited by hyperoxia. Therefore, LXA4 attenuated hyperoxia‑induced injury in MLE‑12 cells via the upregulation of HO‑1 expression. The protection of LXA4 in hyperoxia‑induced cell injury may be associated with the downregulation IL‑6 and MCP‑1 levels via the inhibition of the p38 MAPK and ERK1/2 signaling pathways.

Introduction

Bronchopulmonary dysplasia (BPD) is an acquired form of chronic lung disease mainly induced by hyperoxia. The general pathogenesis of BPD is attributed to oxidative stress and inflammation injury, and the oxygen free radicals and inflammatory factors are important reasons for hyperoxia-induced lung injury (1). The mechanisms of hyperoxia-induced lung injury in BPD involved pulmonary endothelial cell barrier disruption, increased vascular permeability, neutrophil invasion, alveolar hypoplasia and reduced capillary development (2). Previous study reported that overexpression of heme oxygenase-1 (HO-1) in lung epithelial cells of neonatal mice model of BPD attenuated lung inflammation, pulmonary arterial remodeling and vascular leak, markedly reduced thickening of alveolar septa and reserved vessel density, attenuated the histological injury of BPD (3). In a rat model of BPD by lipopolysaccharide (LPS), the expression of interleukin 6 (IL-6) in lung tissues increased (4). Inflammatory marker monocyte chemotactic protein 1 (MCP-1) in newborn mouse exposed to 85% O2 was higher than those exposed to room air (5). Although pathogenesis of BPD is widely discussed, but the effective treatment is still intractable. Application of glucocorticoids, especially inhaled glucocorticoids, showed some therapeutic effect for BPD, but was not recommended as the preferred treatment due to its potential systemic damage (68).

Lipoxin A4 (LXA4) is a metabolites of arachidonic acid and has dual powerful anti-inflammatory and proresolution activities (9). Lipoxins have protective effects on many inflammatory organ models, such as colitis (10), brain ischemia reperfusion injury (11), asthma (12), acute pancreatitis (13). BML-111, an agonist of LXA4 receptor, attenuated LPS-induced lung injury via inhibition of expression of IL-6 and of activation of the protein kinase B (Akt), extracellular signal-regulated kinase 1/2 (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK) signaling pathways (14). LXA4-imparted inhibition of IL-6 and IL-1β was related to blockage of p38 MAPK and ERK1/2 (15). HO-1 is an important component of the cellular defense enzyme that is induced by and acts against oxidant-induced tissue injury (16). HO-1 overexpression mainly preserved vascular growth and barrier function via antioxidant, anti-inflammatory and iron-independent pathways to meliorate the histological injury of BPD (3). Our previous studies confirmed that LXA4 may protect oxidative stress-induced injury of cardiomyocytes via HO-1 overexpression (17). LXA4 amplified HO-1 gene expression in human corneal epithelial cells (18). LXA4 has glucocorticoids-like anti-inflammatory and anti-oxidant effects without the side effects like glucocorticoids, therefore becomes a promising therapeutic for BPD (19), since studies have shown that the main causes of BPD were hyperoxia injury and inflammation (20). Up to now, it remains unclear whether LXA4-imparted therapeutic effect for BPD is mediated by upregulation of HO-1 and downregulation of IL-6 and MCP-1. Given this background of above mentioned studies, we hypothesized that LXA4 may protect hyperoxia-induced lung epithelial cells injury via regulation of HO-1, IL-6 and MCP-1, and LXA4-imparted regulation of IL-6 and MCP-1 may be related to modulation of p38 MAPK, ERK1/2 and Akt signaling pathways. In the present studies, a classical hyperoxia-induced cellular model of BPD in vitro was used to investigate the protective effect of LXA4 on murine lung epithelial cells against hyperoxia-induced injury.

Materials and methods

Reagents

Fetal bovine serum (FBS) was purchased from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA. TRIzol reagents were purchased from Invitrogen; Thermo Fisher Scientific, Inc.. LXA4 was obtained from Calbiochem (San Diego, CA, USA). Rabbit anti-mouse threonine/tyrosine-dephosphorylated ERK1/2 (P-ERK1/2), total-ERK1/2, threonine/tyrosine-diphosphorylated p38 MAPK (P-p38 MAPK), total-P38 MAPK, serine-phosphorylated Akt (P-Akt), total-Akt, β-actin antibodies were purchased from Cell Signaling Technologies (Danvers, MA, USA). LY294002, an inhibitor of the phosphotransferase activity of Akt, PD98059, an inhibitor of ERK1/2 phosphorylation and SB203580, an inhibitor of p38 MAPK phosphorylation, were obtained from Selleck Chemicals (Boston, MA, USA). IL-6, MCP-1, rabbit anti-mouse IL-6 and anti-mouse MCP-1 antibodies were purchased from Pepro Tech (Rocky Hill, NJ, USA). Rabbit anti-mouse HO-1 antibodies were obtained from Santa Cruz Biotechnology (Dallas, TX, USA). Enzyme-linked immunosorbent assay kits (ELISA) for IL-6, MCP-1 assessment were purchased from Assay Designs (Ann Arbor, MI, USA). Prime Script™ RT reagent kit and SYBR premix Ex Taq™ were obtained from Takara Bio Inc (Shiga, Japan). Trypan blue, cell counting kit-8 (CCK-8) and superoxide dismutase (SOD) kits were purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). Zinc protoporphyrin-IX (ZnPP-IX), a specific inhibitor of HO-1 activity, was obtained from Sigma-Aldrich; Merck KGaA, Darmstadt, Germany.

Cell culture

Murine lung epithelial cells (MLE-12) cells were obtained from Shanghai Institutes of Biological Sciences, Chinese Academy of Medical Sciences. MLE-12 cells were cultured in RPMI-1640 supplemented with 10% FBS, 100 µg/ml streptomycin and 100 U/ml penicillin in a 5% CO2 incubator at 37°C, The medium was changed every 2–3 days. The cells were seeded into 6, 12, or 96-well cell culture plates for different experiments. Logarithmic growth phase cells were prepared for experiment. The cells were then cultured under 21% O2 or 85% O2 for 12 h after pretreatment with or without LXA4 (10 nmol/l), ZnPP-IX (10 µmol/l), IL-6 (10 ng/ml), MCP-1 (10 ng/ml), anti-IL-6 (10 ng/ml) and anti-MCP-1 (10 ng/ml) for 12 h, SB203580 (30 µmol/l), LY294002 (10 µmol/l) or PD98059 (40 µmol/l) for 30 min. For all cell stimulations, three independent experiments were performed.

Measurement of cell survival rates and viability

The cells were seeded into 12 and 96-well plates for cell stimulation experiment, and then collected for determination of cell survival rates and viability by Trypan blue exclusion and CCK-8 respectively following the manufacturer's instructions.

SOD assay

MLE-12 cells were seeded into 12-well plates for 24 h, the supernatants were collected for determination of SOD using SOD kits following the manufacturer's instructions.

Flow cytometry assay for apoptosis

Identification of apoptotic cells was performed by using allophycocyanin conjugated Annexin V labeled with fluorescein isothiocyanate (FITC), following the recommendations of the manufacturer. Necrotic cells were excluded by counter-staining with 2 µg/ml propidium iodide. Data were collected by using a fluorescence activated cell sorter (FACS) Canto flow cytometer and analyzed by using a FACS Diva software package.

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

Expression of lipoxin receptor (ALX) mRNA was determined by semi-quantitative PCR analysis using 10% gels. Expressions of HO-1, IL-6 and MCP-1 mRNA were determined by RT-qPCR analysis. Total RNA was isolated by using TRIzol reagent. The RNA was reverse transcribed by the Prime Script™ RT reagent kit following the manufacturer's instructions. The sets of ALX, HO-1, IL-6, MCP-1 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) primers were selected by software-aided analysis (Primer Premier 5.0). The following sets of primers were used for ALX sense, 5′-GTTGAACACAGCTATCACGTTTGT-3′ and antisense, 5′-ACAACTCCTGTAAGAACTCGGAAA-3′ generating a 171-bp fragment, for HO-1 sense, 5′-ACAGATGGCGTCACTTCG-3′ and antisense, 5′-TGAGGACCCACTGGAGGA-3′ generating a 128-bp fragment, for IL-6 sense, 5′-CGGAGAGGAGACTTCACAGAG-3′ and antisense, 5′-CATTTCCACGATTTCCCAGA-3′ amplifying a 105-bp fragment, for MCP-1 sense, 5′-CAACGAGATGCTCTGGGTAGA-3′ and antisense, 5′-TACCTCTTGGGACCCTCCT-3′ amplifying a 585-bp fragment, for GAPDH sense, 5′-TGACAAACGGGACCTAAT-3′ and antisense, 5′-CTGGCACTGCACAAGAAG-3′ generating a 101-bp fragment. RT-qPCR was performed by using StepOne™ Real-Time PCR System machine (Applied Biosystems, Foster City, CA, USA). A typical cycling protocol was consisted of three stages: 15 sec at 95°C for denaturation, 1 min at 60°C for annealing, 15 sec at 95°C for extension, and an additional 20 s for fluorescent signal acquisition. A total of 40 cycles were performed. The results were analyzed by computing the Cq values for target gene in samples using the 2−∆∆Cq method (21).

Immunofluorescence assay

Cellular HO-1 was determined by using immunofluorescence assay. The cells were grown on glass coverslips over 24-well plates, and then fixed with 4% paraformaldehyde, washed and incubated with 5% BSA for 30 min at 37°C. The cells were then incubated with the antibodies against HO-1 at 1:100 dilution over night at 4°C. Subsequently, the cells were washed and incubated with biotin-conjugated anti-rabbit IgG at 1:500 dilution, followed by incubation with FITC-conjugated streptavidin for 1 h at room temperature. Coverslips were flipped on slides, and images of labeled cells were visualized by fluorescence microscopy (Axiovert 200 M; Carl Zeiss, Jena, Germany).

Western blot analysis

MLE-12 cells were collected, total proteins of the cells were abstracted by using protein extraction kits following the manufacturer instructions. Protein concentration was estimated by using the BCA kit. The 50 µg of the protein was loaded for SDS-polyacrylamide gel for 2 h before transferred onto PVDF membranes. Nonspecific sites on the membranes were blocked for 1 h in Tris buffered saline with Tween-20 (TBST) containing 5% nonfat milk. The membranes were incubated with antibodies against HO-1 at 1:1,000 dilution, p38 MAPK, P-p38 MAPK, Akt, P-Akt, ERK1/2, P-EKR1/2 at 1:1,000 dilution at 4°C overnight and washed with TBST. The membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies for 1 h at 37°C. After washing with TBST, signals were visualized by chemiluminescent horseradish peroxidase substrate and normalized to β-actin.

ELISA of IL-6 and MCP-1

The levels of IL-6 and MCP-1 in cellular supernatants were determined by using ELISA kits according to the manufacturer's instructions.

Statistical analysis

Results are expressed as the mean ± standard error of the mean. Experimental data were analyzed using one-way analysis of variance followed by the Least Significant Difference post hoc test and SPSS version 19.0 software (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

LXA4 alleviated hyperoxia-induced cell injury

Normal MLE-12 cells were showed as polygonal-shaped with full and integral appearance accompanied by good adhesion, hyperoxia exposure led to alterations in cell morphology including cell shrinkage, nonfullness and pyknosis/necrosis accompanied by adhesion disability, however, pretreatment with LXA4 significantly protected the cells from the morphological changes induced by hyperoxia exposure (Fig. 1A). Above results are consistent with the cell viability analysis assessed by using CCK-8 assay (Fig. 1E) and the cell survival rates (Fig. 1D) assessed by using trypan blue exclusion (Fig. 1B). As shown in Fig. 1F, the SOD levels were decreased in the cells exposed to hyperoxia as compared to the cells treated with air alone. However, pretreatment of the cells exposed to hyperoxia with LXA4 increased the SOD levels as compared to the cells undergoing hyperoxia alone. As elucidated in Fig. 1C, the ALX was existed in MLE-12 cells.

LXA4 reduced apoptosis caused by hyperoxia

As presented in Fig. 2A and B, the cell apoptosis rates were increased in the cells exposed to hyperoxia as compared to the cells treated with air alone. LXA4 reduced the hyperoxia-induced cell apoptosis rates as compared to the cells undergoing hyperoxia alone.

ZnPP-IX reversed LXA4-imparted protection

As indicated in Fig. 3, pretreatment of the cells with ZnPP-IX abolished the LXA4-imparted protection cell viability which was reduced by hyperoxia.

LXA4 induced HO-1 expression

HO-1 mRNA and protein expressions were measured in the cells exposed to hyperoxia and pretreated with 0, 1, 10 and 50 nmol/l LXA4 for 1, 6, 12, 24 h. LXA4 upregulated the levels of HO-1 in a dose-dependent manner, and the peak expression of HO-1 was induced by 10 nmol/l LXA4 at 12 h (Fig. 4A and B). Accordingly, 10 nmol/l LXA4 was used for measurement of HO-1 expressions in the cells after treatment with LXA4 for 12 h. LXA4 (10 nmol/l) slightly increased the expressions of HO-1 mRNA and protein in the cells exposed to air alone (Fig. 4C and D) but not reach to statistical significance. Consistently, LXA4 upregulated the levels of HO-1 in the cells exposed to hyperoxia (Fig. 4C and D). The localization of HO-1 in the MLE-12 cells in response to LXA4 and hyperoxia was also assessed by using fluorescence microscope (Fig. 4E). There was no HO-1 expression in the cells exposed to air alone. LXA4 slightly promotes HO-1 expression in the cells exposed to air alone. A stronger expression of HO-1 in the cytoplasm of the cells undergoing hyperoxia was induced at 12 h after the stimulation with LXA4 as compared to the cells exposed to hyperoxia alone.

IL-6 and MCP-1 increased cell damage

As depicted in Fig. 5, treatment of the cells with IL-6 and MCP-1 decreased the cell viability in the cells exposed to air alone, similar to the effect of hyperoxia on cell viability. On the contrary, pretreatment of cells with anti-IL-6 and anti-MCP-1 antibodies reversed the hyperoxia-induced inhibition on cell vitality as compared to cells exposed to hyperoxia alone.

LXA4 inhibited IL-6 and MCP-1 induced by hyperoxia

There were higher levels of IL-6 and MCP-1 in the cells undergoing hyperoxia as compared to cells treated with air alone. However, pretreatment of the cells with LXA4 inhibited the IL-6 and MCP-1 levels induced by hyperoxia as compared to cells exposed to hyperoxia alone (Fig. 6).

Role of p38 MAPK, ERK1/2 and Akt in IL-6 and MCP-1 expression

The effects of p38 MAPK, ERK1/2 and Akt inhibition on expression of IL-6 and MCP-1 were illustrated in Fig. 7A and B. The p38 MAPK pathway inhibitor SB203580 and ERK1/2 pathway inhibitors PD98059 significantly inhibited the hyperoxia-induced IL-6 and MCP-1 expressions respectively, and LXA4 also significantly inhibited the hyperoxia-induced IL-6 and MCP-1 expressions, whereas the Akt inhibitor LY294002 did not. As revealed in Fig. 7C-E, LXA4 alone slightly decreased P-p38 MAPK and P-ERK1/2 expressions but increased P-Akt expression in the cells treated with air alone. Hyperoxia exposure significantly increased the P-p38 MAPK and P-ERK1/2 expressions but decreased the P-Akt expression in the cells without LXA4 pretreatment. LXA4 significantly decreased P-p38 MAPK and P-ERK1/2 expressions but increased P-Akt expression in the cells treated with hyperoxia. LXA4 upregulated Akt signaling pathways which was inhibited by hyperoxia.

Discussion

BPD is one of the most serious lung complication in premature infant caused by hyperoxia and inflammation. Unfortunately, few effective therapies are known for BPD until now. LXA4 play a unique protection on endothelial cells, epithelial cells, lung and other tissues, has anti-oxidative stress, anti-inflammation and anti-proliferation effect (2224), may be the good candidate therapy for BPD (19). In the present study, we identified that LXA4 play a protective role on hyperoxia-induced injury in murine lung epithelial cell model of BPD. First, treatment of MLE-12 cells with LXA4 ameliorated the morphological injury induced by hyperoxia. Second, treatment of MLE-12 cells with LXA4 improved the cell survival rates, cell viability, and the SOD level in the cells exposed to hyperoxia. Furthermore, treatment of MLE-12 cells with LXA4 suppressed apoptosis rates induced by hyperoxia. Our results are in accordance with previous reports which demonstrated that LXA4 improved alveolarization, reduced mucosal inflammation, promoted resolution in a neonatal murine BPD model induced by hyperoxia (19).

Oxygen radicals are direct and important causes of oxidative stress injury, and HO-1 is the powerful antioxidant enzyme to reduce the oxygen free radicals in vivo (25,26). HO-1 and its metabolites carbon monoxide, biliverdin and bilirubin have anti-inflammatory, antioxidant, and cytoprotective functions (27,28). HO-1 can be induced by a variety of factors including inflammatory cytokines, oxidative stress, LPS and endotoxin. LPS induces HO-1 overexpression in monocytes to modulate the expressions of inflammatory cytokines including IL-6 and MCP-1 (29,30). In the present study, we offered the evidence for the first time that overexpression of HO-1 induced by LXA4 attenuated hyperoxia-induced cell injury. First, LXA4 upregulated the expressions of HO-1 in the cells exposed to air alone or hyperoxia. Moreover, ZnPP-IX, a HO-1 inhibitor, reversed the LXA4-imparted protection on cell viability which was reduced by hyperoxia. These results are supported by previous investigations which demonstrated that LXA4 and LXA4 receptor agonist can protect the heart and renal oxidative stress damage via HO-1 overexpression (17,31), and HO-1 play an important role in protecting the developing pulmonary vasculature from hyperoxia-induced injury in a murine model of BPD (3).

In the developing lung, extreme hyperoxia exposure produces a sustained inflammation (32,33), and inflammatory cytokines lead to structural abnormalities and remodeling of the vessels of the neonatal lung (3436). However, it remains unclear whether LXA4 inhibits IL-6 and MCP-1 expressions in hyperoxia-induced cell injury. In the present study, our results offered the evidence for the first time that LXA4-imparted suppression on hyperoxia-induced inflammatory injury is related to downregulation of IL-6 and MCP-1. First, treatment of the cells with LXA4 inhibited the expressions of IL-6 and MCP-1 induced by hyperoxia in parallel to the inhibition on hyperoxia-induced cell injury. Second, IL-6 and MCP-1 decreased cell viability, increased cell damage in cells exposed to air alone. MLE-12 is murine Type II alveolar epithelial cells, can secrete alveolar surfactant to maintain the normal morphology of alveoli, but also can secrete IL-6, MCP-1, MIP-2 and other chemokines involved in inflammation. These inflammatory mediators are involved in the process of alveolar cell injury, which involves a number of signaling pathways that I have already supplemented. IL-6 is a multi-directional cytokine of inflammatory response and immune system. It is involved in the pathological process of many diseases such as immune response and acute phase reaction. IL-6 regulates cell proliferation, differentiation and apoptosis in different tissues through complex cell signaling pathways Death and so on (37,38). Inflammatory reaction can activate many kinds of different stress signal pathways and change the balance between them. JAK-STAT signal pathway is the main signal transduction pathway downstream of IL-6 family and is involved in the induction of various cytokines Of signal transduction, involving a variety of neurological functions such as cell growth, differentiation, inflammation, mutation and apoptosis. Inflammatory reaction can activate many kinds of different stress signal pathways and change the balance between them. JAK-STAT signal pathway is the main signal transduction pathway downstream of IL-6 family and is involved in the induction of various cytokines Of signal transduction, involving a variety of neurological functions such as cell growth, differentiation, inflammation, mutation and apoptosis. Upregulation of IL-6 activates phosphorylation of STAT3 (39). Among the predisposing factors of apoptosis, inflammatory factors play an important role (40,41). MAPKs are intracellular serine/threonine protein kinases. MAPKs signal transduction pathway exists in most cells, and plays an important role in cell proliferation, differentiation, transformation and apoptosis. At present, four parallel MAPKs signaling pathways have been found in higher mammalian cells. Among them, P38MAPK and JNK/SAPK can be stimulated by inflammatory stimuli (IL-6, IL-1 and TNF-α). Growth factor (EGF) and some G protein-coupled receptors are activated by three-layer activated enzyme systems, MAPKs, MAP2Ks, and MAP3Ks, which act through cascade of protein phosphorylation-induced cascades effect. Activated P38MAPK and JNK/SAPK Pathways Regulate Gene Transcription by Phosphorylation of Transcription Factors and Other Targets thereby Inducing Apoptosis (4244). Monocyte chemoattractant protein-1 (MCP-1) is the first human CC chemokine that has been discovered and is currently a member of more research in the MCP family. It is associated with CC chemokines CCR2 can exert different physiological functions, such as inducing homing, migration, activation, differentiation and development of lymphocytes and natural killer cells, inflammation, angiogenesis and the like. The main mechanisms CCR2 and MCP 1 can induce calcium influx. NF-κB is the main regulator of inflammatory response, NF-κB can activate the transcription of MCP-1 gene, upregulated MCP-1 level expression (45). The signaling pathways involved in the damage of alveolar epithelial cells by these inflammatory factors need to be further studied. Furthermore, anti-IL-6 and anti-MCP-1 antibodies reversed the hyperoxia-reduced cell viability, decreased cell injury induced by hyperoxia. These results suggest that effect of LXA4 on hyperoxia-induced cell injury is similar to that of anti-IL-6 and anti-MCP-1. Our results are consistent with previous reports which demonstrated that hyperoxia induced long-term airway reactivity with persistent lung inflammation associated with a marked increase in inflammatory cytokines in newborn mice (5), and LXA4 receptor agonist BML-111 and LXA4 attenuated LPS-induced IL-6 expression (14,15), and LXA4 inhibited IL-6 expression in rat mesangial cells (46), and LXA4 attenuated MCP-1 release in human intestinal mucosa (47).

The phosphorylation of p38 MAPK and ERK1/2 is necessary for IL-6 secretion in endothelial cells exposed to LPS (48,49). In the present study, we offered the evidence that LXA4-imparted suppression of IL-6 and MCP-1 is mediated by p38 MAPK and ERK1/2-dependent signaling pathways in the cells exposed to hyperoxia. First, hyperoxia activated the phosphorylation of p38 MAPK and ERK1/2 in parallel to stimulation of IL-6 and MCP-1 expressions, and LXA4 inhibited the phosphorylation of p38 MAPK and ERK1/2 induced by hyperoxia in parallel to inhibition of IL-6 and MCP-1 expressions induced by hyperoxia. Moreover, treatment of the cells with SB203580 and PD98059 reduced IL-6 and MCP-1 levels induced by hyperoxia, and SB203580, PD98059 plus LXA4 further reduced IL-6 and MCP-1 levels in cells exposed to hyperoxia. These results are supported by previous reports which showed that LXA4 receptor agonist inhibited the phosphorylation of ERK1/2, p38 MAPK in endothelial cells exposed to LPS (14), and LXA4-imparted inhibition of IL-6 and IL-1β was related to blockage of p38 MAPK, p42/44 MAPK (ERK1/2) (15). In addition, we found that the phosphorylation of Akt was inhibited by hyperoxia exposure in MLE-12 cells, and LXA4 reversed the expression of phosphorylation of Akt inhibited by hyperoxia, this result is consistent with the previous study which showed that activation of Akt protected alveoli from neonatal oxygen-induced lung injury (50). However, the precise mechanism needs to be explored in further study.

In conclusion, our present studies demonstrate following findings, first, LXA4 attenuates hyperoxia-induced cell injury via upregulation of HO-1, second, LXA4 decreased the expressions of IL-6 and MCP-1 induced by hyperoxia injury, third, LXA4-imparted inhibition of IL-6 and MCP-1 may be mediated by downregulation of p38 MAPK and ERK1/2 signaling pathways. Overall, our data indicate that LXA4 could be a useful chemical for the choice of a new therapy in treatment of BPD.

Acknowledgements

The authors would like to thank Dr Chao Gao and Dr Yu-Gui Cui for their technical assistance (First Affiliated Hospital with Nanjing Medical University, Jiangsu, China). The abstract was submitted to the American Academy of Pediatrics to be presented on 15th September 2018 at the Marriott Marquis Chicago, Great Lakes, Chicago, IL, USA; however, the abstract was not presented, only published in Pediatrics, Neonatal-Perinatal Medicine Program: Day 1, 142 (1 Meeting Abstract), 191–191: 2018.

Funding

The present study was supported by National Natural Scientific Fund of China (grant no. 81871195 and 81741052) and Construction Program of Jiangsu Provincial Clinical Research Center Support System (grant no. BL2014084).

Availability of data and materials

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

Authors' contributions

SHW, XQC, RJ and HYL conceived and designed the experiments. YYL, BJL, SJL and ZYS performed the experiments, and analyzed the data. RJ and HYL revised the manuscript critically for important intellectual content. YYL, XQC and SHW wrote the paper.

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 

Balany J and Bhandari V: Understanding the impact of infection, inflammation, and their persistence in the pathogenesis of bronchopulmonary dysplasia. Front Med (Lausanne). 2:902015.PubMed/NCBI

2 

Bhandari V: Hyperoxia-derived lung damage in preterm infants. Semin Fetal Neonatal Med. 15:223–229. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Fernandez-Gonzalez A, Alex Mitsialis S, Liu X and Kourembanas S: Vasculoprotective effects of heme oxygenase-1 in a murine model of hyperoxia-induced bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 302:L775–L784. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Muramatsu Y, Ito M, Oshima T, Kojima S and Ohno K: Hydrogen-rich water ameliorates bronchopulmonary dysplasia (BPD) in newborn rats. Pediatr Pulmonol. 51:928–935. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Kumar VH, Lakshminrusimha S, Kishkurno S, Paturi BS, Gugino SF, Nielsen L, Wang H and Ryan RM: Neonatal hyperoxia increases airway reactivity and inflammation in adult mice. Pediatr Pulmonol. 51:1131–1141. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Schreiner C, Schreiner F, Härtel C, Heckmann M, Heep A, Bartmann P, Woelfle J, Müller A, Herting E and Göpel W; German Neonatal Network GNN, : Glucocorticoid receptor gene variants and neonatal outcome in very-low-birth-weight preterm infants. Neonatol. 111:22–29. 2016. View Article : Google Scholar

7 

Lakshminrusimha S, Mathew B and Leach CL: Pharmacologic strategies in neonatal pulmonary hypertension other than nitric oxide. Semin Perinatol. 40:160–173. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Slaughter JL, Stenger MR, Reagan PB and Jadcherla SR: Utilization of inhaled corticosteroids for infants with bronchopulmonary dysplasia. PLoS One. 9:e1068382014. View Article : Google Scholar : PubMed/NCBI

9 

Serhan CN, Chiang N and Van Dyke TE: Resolving inflammation: Dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 8:349–361. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Fiorucci S, Wallace JL, Mencarelli A, Distrutti E, Rizzo G, Farneti S, Morelli A, Tseng JL, Suramanyam B, Guilford WJ and Parkinson JF: A beta-oxidation-resistant lipoxin A4 analog treats hapten-induced colitis by attenuating inflammation and immune dysfunction. Proc Natl Acad Sci USA. 101:15736–15741. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y and Yao SL: Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res. 1323:174–183. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Levy BD, De Sanctis GT, Devchand PR, Kim E, Ackerman K, Schmidt BA, Szczeklik W, Drazen JM and Serhan CN: Multi-pronged inhibition of airway hyper-responsiveness and inflammation by lipoxin A(4). Nat Med. 8:1018–1023. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Yang F, Xie J, Wang W, Xie Y, Sun H, Jin Y, Xu D, Chen B, Andersson R and Zhou M: Regional arterial infusion with lipoxin A4 attenuates experimental severe acute pancreatitis. PLoS One. 9:e1085252014. View Article : Google Scholar : PubMed/NCBI

14 

Tang M, Chen L, Li B, Wang Y, Li S, Wen A, Yao S and Shang Y: BML-111 attenuates acute lung injury in endotoxemic mice. J Surg Res. 200:619–630. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Wu SH, Liao PY, Dong L and Chen ZQ: Signal pathway involved in inhibition by lipoxin A(4) of production of interleukins induced in endothelial cells by lipopolysaccharide. Inflamm Res. 57:430–437. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Hangaishi M, Ishizaka N, Aizawa T, Kurihara Y, Taguchi J, Nagai R, Kimura S and Ohno M: Induction of heme oxygenase-1 can act protectively against cardiac ischemia/reperfusion in vivo. Biochem Biophys Res Commun. 279:582–588. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Chen XQ, Wu SH, Zhou Y and Tang YR: Lipoxin A4-induced heme oxygenase-1 protects cardiomyocytes against hypoxia/reoxygenation injury via p38 MAPK activation and Nrf2/ARE complex. PLoS One. 8:e671202013. View Article : Google Scholar : PubMed/NCBI

18 

Biteman B, Hassan IR, Walker E, Leedom AJ, Dunn M, Seta F, Laniado-Schwartzman M and Gronert K: Interdependence of lipoxin A4 and heme-oxygenase in counter-regulating inflammation during corneal wound healing. FASEB J. 21:2257–2266. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Martin CR, Zaman MM, Gilkey C, Salguero MV, Hasturk H, Kantarci A, Van Dyke TE and Freedman SD: Resolvin D1 and lipoxin A4 improve alveolarization and normalize septal wall thickness in a neonatal murine model of hyperoxia-induced lung injury. PLoS One. 9:e987732014. View Article : Google Scholar : PubMed/NCBI

20 

Kair LR, Leonard DT and Anderson JM: Bronchopulmonary dysplasia. Pediatr Rev. 33:255–264. 2012. View Article : Google Scholar : PubMed/NCBI

21 

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 : PubMed/NCBI

22 

Wu L, Liu ZJ, Miao S, Zou LB, Cai L, Wu P, Ye du Y, Wu Q and Li HH: Lipoxin A4 ameliorates cerebral ischaemia/reperfusion injury through upregulation of nuclear factor erythroid 2-related factor 2. Neurol Res. 35:968–975. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Chen XQ, Wu SH, Zhou Y and Tang YR: Involvement of K+ channel-dependant pathways in lipoxin A4-induced protective effects on hypoxia/reoxygenation injury of cardiomyocytes. Prostaglandins Leukot Essent Fatty Acids. 88:391–397. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Yang F, Xie J, Wang W, Xie Y, Sun H, Jin Y, Xu D, Chen B, Andersson R and Zhou M: Regional arterial infusion with lipoxin A4 attenuates experimental severe acute pancreatitis. PLoS One. 9:e108525. 2014.

25 

Masini E, Vannacci A, Marzocca C, Pierpaoli S, Giannini L, Fantappié O, Mazzanti R and Mannaioni PF: Heme oxygenase-1 and the ischemia-reperfusion injury in the rat heart. Exp Biol Med (Maywood). 228:546–549. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Chok MK, Ferlicot S, Conti M, Almolki A, Dürrbach A, Loric S, Benoît G, Droupy S and Eschwège P: Renoprotective potency of heme oxygenase-1 induction in rat renal ischemia-reperfusion. Inflamm Allergy Drug Targets. 8:252–259. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Paine A, Eiz-Vesper B, Blasczyk R and Immenschuh S: Signaling to heme oxygenase-1and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 80:1895–1903. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Ryter SW and Choi AM: Heme oxygenase-1/carbon monoxide: From metabolism to molecular therapy. Am J Respir Cell Mol Biol. 41:251–260. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Rushworth SA, MacEwan DJ and O'Connell MA: Lipopolysaccharide-induced expression of NAD (P)H: Quinone oxidoreductase 1 and heme oxygenase-1 protects against excessive inflammatory responses in human monocytes. J Immunol. 181:6730–6737. 2008. View Article : Google Scholar : PubMed/NCBI

30 

So Y, Lee SY, Han AR, Kim JB, Jeong HG and Jin CH: Rosmarinic acid methyl ester inhibits LPS-induced NO production via suppression of MyD88-dependent and -independent pathways and induction of HO-1 in RAW 264.7 cells. Molecules. 21(pii): E10832016. View Article : Google Scholar : PubMed/NCBI

31 

Wu SH, Chen XQ, Lü J and Wang MJ: BML-111 attenuates renal ischemia/reperfusion injury via peroxisome proliferator-activated receptor-α-regulated heme oxygenase-1. Inflammation. 39:611–624. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Wu J, Hafner C, Schramel JP, Kaun C, Krychtiuk KA, Wojta J, Boehme S, Ullrich R, Tretter EV, Markstaller K and Klein KU: Cyclic and constant hyperoxia cause inflammation, apoptosis and cell death in human umbilical vein endothelial cells. Acta Anaesthesiol Scand. 60:492–501. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Deng H, Mason SN and Auten RL Jr: Lung inflammation in hyperoxia can be prevented by antichemokine treatment in newborn rats. Am J Respir Crit Care Med. 162:2316–2323. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Aslam M, Baveja R, Liang OD, Fernandez-Gonzalez A, Lee C, Mitsialis SA and Kourembanas S: Bone marrow stromal cells attenuate lung injury in a murine model of neonatal chronic lung disease. Am J Respir Crit Care Med. 180:1122–1130. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Bry K, Hogmalm A and Bäckström E: Mechanisms of inflammatory lung injury in the neonate: Lessons from a transgenic mouse model of bronchopulmonary dysplasia. Semin Perinatol. 34:211–221. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Ryan RM, Ahmed Q and Lakshminrusimha S: Inflammatory mediators in the immunobiology of bronchopulmonary dysplasia. Clin Rev Allergy Immunol. 34:174–190. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Ishihara K and Hirano T: IL-6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine Growth Factor Rev. 13:357–368. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Donegan JJ, Girotti M, Weinberg MS and Morilak DA: A novel role for brain interleukin-6: Facilitation of cognitive flexibility in rat orbitofrontal cortex. J Neurosci. 34:953–962. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Hao Y, Jing H, Bi Q, Zhang J, Qin L and Yang P: Intra-amygdala microinfusion of IL-6 impairs the auditory fear conditioning of rats via JAK/STAT activation. Behav Brain Res. 275:88–95. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Wong LM, Myers SJ, Tsou CL, Gosling J, Arai H and Charo IF: Organization and differential expression of the human monocyte chemoattractant protein 1 receptor gene. Evidence for the role of the carboxyl-terminal tail in receptor trafficking. J Biol Chem. 272:1038–1045. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Cho ML, Yoon BY, Ju JH, Jung YO, Jhun JY, Park MK, Park SH, Cho CS and Kim HY: Expression of CCR2A, an isoform of MCP-1 receptor, is increased by MCP-1, CD40 ligand and TGF-beta in fibroblast like synoviocytes of patients with RA. Exp Mol Med. 39:499–507. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Han J and Sun P: The pathways to tumor suppression via route p38. Trends Biochem Sci. 32:364–371. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Hui L, Bakiri L, Stepniak E and Wagner EF: p38alpha: A suppressor of cell proliferation and tumorigenesis. Cell Cycle. 6:2429–2433. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Mishra S, Mishra JP and Kumar A: Activation of JNK-dependent pathway is required for HIV viral protein R-induced apoptosis in human monocytic cells: Involvement of antiapoptotic BCL2 and c-IAP1 genes. J Biol Chem. 282:4288–4300. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Zhao Y, Wang CL, Li RM, Hui TQ, Su YY, Yuan Q, Zhou XD and Ye L: Wnt5a promotes inflammatory responses via nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in human dental pulp cells. J Biol Chem. 292:43582017. View Article : Google Scholar : PubMed/NCBI

46 

Wu SH, Lu C, Dong L, Zhou GP, He ZG and Chen ZQ: Lipoxin A4 inhibits TNF-α-induced production of interleukins and proliferation of rat mesangial cells. Kidney Int. 68:35–46. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Goh J, Baird AW, O'Keane C, Watson RW, Cottell D, Bernasconi G, Petasis NA, Godson C, Brady HR and MacMathuna P: Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 antagonize TNF-alpha-stimulated neutrophil-enterocyte interactions in vitro and attenuate TNF-alpha-induced chemokine release and colonocyte apoptosis in human intestinal mucosa ex vivo. J Immunol. 167:2772–2780. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Arditi M, Zhou J, Torres M, Durden DL, Stins M and Kim KS: Lipopolysaccharide stimulates the tyrosine phosphorylation of mitogen-activated protein kinases p44, p42, and p41 in vascular endothelial cells in a soluble CD14-dependent manner. Role of protein tyrosine phosphorylation in lipopolysaccharide-induced stimulation of endothelial cells. J Immunol. 155:3994–4003. 1995.PubMed/NCBI

49 

Matsuda T, Omori K, Vuong T, Pascual M, Valiente L, Ferreri K, Todorov I, Kuroda Y, Smith CV, Kandeel F and Mullen Y: Inhibition of p38 pathway suppresses human islet production of pro-inflammatory cytokines and improves islet graft function. Am J Transplant. 5:484–493. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Alphonse RS, Vadivel A, Coltan L, Eaton F, Barr AJ, Dyck JR and Thébaud B: Activation of Akt protects alveoli from neonatal oxygen-induced lung injury. Am J Respir Cell Mol Biol. 44:146–154. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 21 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo YY, Wu SH, Lu HY, Li BJ, Li SJ, Sun ZY, Jin R and Chen XQ: Lipoxin A4 attenuates hyperoxia‑induced lung epithelial cell injury via the upregulation of heme oxygenase‑1 and inhibition of proinflammatory cytokines. Mol Med Rep 21: 429-437, 2020
APA
Luo, Y., Wu, S., Lu, H., Li, B., Li, S., Sun, Z. ... Chen, X. (2020). Lipoxin A4 attenuates hyperoxia‑induced lung epithelial cell injury via the upregulation of heme oxygenase‑1 and inhibition of proinflammatory cytokines. Molecular Medicine Reports, 21, 429-437. https://doi.org/10.3892/mmr.2019.10821
MLA
Luo, Y., Wu, S., Lu, H., Li, B., Li, S., Sun, Z., Jin, R., Chen, X."Lipoxin A4 attenuates hyperoxia‑induced lung epithelial cell injury via the upregulation of heme oxygenase‑1 and inhibition of proinflammatory cytokines". Molecular Medicine Reports 21.1 (2020): 429-437.
Chicago
Luo, Y., Wu, S., Lu, H., Li, B., Li, S., Sun, Z., Jin, R., Chen, X."Lipoxin A4 attenuates hyperoxia‑induced lung epithelial cell injury via the upregulation of heme oxygenase‑1 and inhibition of proinflammatory cytokines". Molecular Medicine Reports 21, no. 1 (2020): 429-437. https://doi.org/10.3892/mmr.2019.10821