Skip to main content
Erschienen in: Respiratory Research 1/2014

Open Access 01.12.2014 | Research

Impact of ozone exposure on the response to glucocorticoid in a mouse model of asthma: involvements of p38 MAPK and MKP-1

verfasst von: Aihua Bao, Feng Li, Min Zhang, Yuqing Chen, Pengyu Zhang, Xin Zhou

Erschienen in: Respiratory Research | Ausgabe 1/2014

Abstract

Background

Molecular mechanisms involved in the oxidative stress induced glucocorticoids insensitivity remain elusive. The mitogen-activated protein kinase phosphatase (MKP) 1 mediates a part of glucocorticoids action and can be modified by exogenous oxidants. Whether oxidant ozone (O3) can affect the function of MKP-1 and hence blunt the response to corticotherapy is not clear.

Methods

Here we employed a murine model of asthma established with ovalbumin (OVA) sensitization and challenge to evaluate the influence of O3 on the inhibitory effect of dexamethasone on AHR and airway inflammation, and by administration of SB239063, a selective p38 MAPK inhibitor, to explore the underlying involvements of the activation of p38 MAPK and the expression of MKP-1.

Results

Ozone exposure not only aggravated the pulmonary inflammation and AHR, but also decreased the inhibitory effects of dexamethasone, accompanied by the elevated oxidative stress, airway neutrophilia, enhanced phosphorylation of p38 MAPK, and upregulated expression of IL-17. Administration of SB239063 caused significant inhibition of the p38 MAPK phosphorylation, alleviation of the airway neutrophilia, and decrement of the ozone-induced IL-17 expression, and partly restored the ozone-impaired effects of dexamethasone. Ozone exposure not only decreased the protein expression of MKP-1, but also diminished the dexamethasone-mediated induction process of MKP-1 mRNA and protein expression.

Conclusions

The glucocorticoids insensitivity elicited by ozone exposure on current asthma model may involve the enhanced phosphorylation of p38 MAPK and disturbed expression of MKP-1.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12931-014-0126-x) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Background

Bronchial asthma is a common airway disease characterized by airway hyperresponsiveness (AHR), airway inflammation, and airway remodeling. Up to date, the most effective treatment of asthma is performed through glucocorticoids (GC), taken systemically or by inhalation. However, up to 10% of the asthmatic patients are clinically insensitive to corticotherapy [1], while the molecular mechanisms contributing to this lack of sensitivity remain unclear. The classical model of the glucocorticoid anti-inflammatory action is established as that the binding of glucocorticoid to its receptor (GR/NR3C1) may transrepress the inflammatory gene transcription or transactivate multiple anti-inflammatory/repressive factors [2]. However, an alternative mechanism of the GC action has gained an increasing body of concern focusing attention on the role of the mitogen-activated protein kinase phosphatase 1 (MKP-1), a phosphatase, induced by glucocorticoids, which inactivates the mitogen-activated protein kinase (MAPKs) [3],[4]. It was reported that the genetic inhibition of MKP-1 aggravated the inflammation of an allergic animal model [5] and attenuated the suppressive effect of dexamethasone (Dex) on several pro-inflammatory gene expressions in macrophages [6]. Previously, we observed that the inhibitory effect of dexamethasone on the acetylcholine-induced airway smooth muscle contraction was dependent on the function of MKP-1 [7]. Maneechotesuwan et al. found that the inhibitory effect of fluticasone on the Th2 cytokines production in cultured human T lymphocytes in vitro was mediated by its induction of MKP-1 expression, which further suppressed the p38 MAPK activation and the GATA-3 nuclear translocation [8]. It was revealed by Furst et al. that the intracellular reactive oxygen species (ROS), which are synthesized under the stimulation of corticosteroids, initiate the induction of the MKP-1 expression by GC, through the c-Jun N-terminal kinase (JNK)-activator protein (AP)-1 pathway [9]. Since the intracellular redox status is vulnerable to the exogenous oxidative stress, it sheds light on the hypothesis that the extrinsic oxidants could influence the inducing process of MKP-1 expression by corticosteroids, subsequently interrupting the anti-inflammatory actions of the drugs.
Mounting evidences support an opinion that oxidative stress plays crucial roles in the pathogenesis of glucocorticoids insensitivity [10], such as causing defective histone acetylation and GR modification, etc. [1]. However, few investigations have been conducted to determine whether the oxidants-mediated disruption of MKP-1 also contributes to the resistance to GCs.
O3 is an exogenous oxidant which adversely affects human health by irritating the mucosa and harming the respiratory system [11]; it is particularly deleterious when encountered by the lungs of asthmatics. We have reported that acute ozone exposure on a mouse model of allergic asthma caused the aggravation of the pulmonary inflammation, AHR, and mucus hyperproduction [12]. Using a chronic allergic asthma model, we found that repeated O3 exposures reduced the response to systemically administrated dexamethasone of the experimental animals [13]. Furthermore, in a human study researchers reported that transient exposure to ozone rendered the stable asthmatic patients more insensitive to the same dosage of inhaled glucocorticoids, compared with those air-exposed patients [14]. O3 is a potent stressor, and the exposure of mice to its influence often causes significant activation of p38 MAPK, which plays a crucial role in the ozone-induced pulmonary inflammation and AHR [15]. Taken into account that the over-phosphorylation of p38 MAPK might be a reflection of impaired dephosphorylation, it is reasonable to infer that ozone might inhibit the function of MKP-1. Generally, we hypothesized that the oxidant ozone might exert some negative effects on the function of MKP-1, and hence interfere with the anti-inflammatory action of GCs.
To verify this hypothesis, we employed a murine asthma model established with ovalbumin (OVA )-induced asthma model mice, in which the animals were exposed to filtered air or 1.0 ppm O3 and administered with dexamethasone in combination with or without a selective p38 MAPK inhibitor, SB239063. Their evaluation was performed thereafter, including the observation and estimation of parameters, such as the pulmonary inflammation, AHR, the phosphorylation of p38 MAPK, and the expression of MKP-1. The results of the current study indicated that the inhibitory effect of dexamethasone was compromised after the O3 exposure, which was associated with the O3-enhanced phosphorylation of p38 MAPK and O3-decreased expression of MKP-1 protein.

Methods

Animals

Six-week-old female Balb/c mice weighting 18 ~ 20 g were purchased from SLAC Laboratory Animal Co. Ltd.(Shanghai, China) and bred under specific-pathogen-free conditions and kept on an ovalbumin (OVA)-free diet in our own facility.

Ethics statement

This study was carried out in strict accordance with the recommendations of the Shanghai Committee for Accreditation of Laboratory Animal. The protocol was approved by the Shanghai First People's Hospital Institutional Review Board (Permit Number: 2010KY047). All surgery was performed under sodium pentobarbital anesthesia, and all efforts were made to minimize suffering.

OVA sensitization and challenge

The protocol of experimental model establishing was adopted as previously described [12]. Mice were sensitized IP with 20 μg chicken OVA (Grade V, Sigma Aldrich Co., St. Louis, MO) emulsified in 2.0 mg of alum (Shanghai No.4 Reagent & H.V. Chemical Industries Ltd., Shanghai, China) in a total volume of 100 μl of 0.9% sterile saline on days 1 and 14. Non-sensitized mice just received 2.0 mg of alum in 0.9% saline. On day 24, 25, and 26, mice received three aerosol challenges with 5% OVA (wt/vol) (Grade II, Sigma Aldrich Co., St. Louis, MO) in 0.9% endotoxin-free saline (non-sensitized mice received saline only) for 30 min daily. Using a plastic box (50 × 30 × 40 cm3) as exposure chamber, where the air was kept at 20 - 22°C and relative humidity 50 - 60%, the aerosol was generated by a sidestream jet nebulizer (PARI BOY, Starnberg, Germany) into the chamber and mice were put into stainless cages inside the chamber with irradiated food and acidified water provided ad libitum. One hour before each challenge, mice received dexamethasone (2 mg/kg) IP in 0.1 ml endotoxin-free PBS or PBS alone. The dosage of dexamethasone has been adopted by multiple publications [16].

Ozone exposure and SB239063 delivery

On days 23, 25 and 27, conscious mice were exposed to ozone [1.0 parts/million (ppm) for 3 h] or filtered air. Ozone was generated by directing an air stream with a micro air pump (SC3601PM, Shenzhen, China) into a AQUA MEDIC O3 generator (model 300, AB Aqua Medic GmbH, Bissendorf, Germany). Ozone concentration in the chamber was monitored continuously by an Ozone Switch (OS-4, Newark, USA), which also controlled the operation of the ozone generator in response to the instantaneous value. The target O3 concentration range was set as 0.77 ppm ~ 1.32 ppm, which has been proved by our preliminary experiment to be able to maintain a comparatively stable O3 concentration averaged as 1.01 ± 0.02 ppm (data not shown). This methodology of O3 exposure has been adapted by previous study which had documented that, such level of ozone inhalation can induced the airway inflammation but not influence the body weight of mice [17], which means less toxic effect. One hour before and 4 hrs after the O3 exposure, mice were injected through tail vein SB239063 (Sigma Aldrich, St. Louis, MO) dissolved in 3% DMSO (0.1 ml, Sigma Aldrich, St. Louis, MO) or DMSO only, respectively. The dosages of SB239063 were titrated in a preliminary experiment, where the final dosage of 4.0 mg/kg was selected. The present experimental protocol was outlined in Figure 1.

Airway responsiveness testing

Airway responsiveness was assessed noninvasively using a whole-body plethysmography (Buxco., NY, USA) in conscious, unrestrained mice as described previously [18]. Briefly, in day 28, mice were placed in individual chambers. After ten minutes adaptation they were exposed to aerosolized PBS (to establish baseline), followed by increasing concentrations of aerosolized methacholine (Mch, Sigma Aldrich, St. Louis, MO) (1.5625, 3.125, 6.25, 12.5, 25, 50 mg/ml). Each dose of Mch was delivered for 0.5 minute, and respiratory measurements were recorded and averaged over a 7-minute period from the beginning of nebulization. Airflow obstruction was expressed as enhanced pause (Penh), which correlates well with airway resistance, and calculated as: Penh = {[Te (expiratory time)/Tr (relaxation time)]-1} × [Pef (peak expiratory flow)/Pif (peak inspiratory flow)]. Penh after each concentration was expressed as percentage change from baseline. The log concentrations of MCh required to increase Penh by 100% from baseline was calculated (LogPC100Penh).

Bronchoalveolar lavage and measurements of BALF cytokines

Immediately after the assessment of airway reactivity, mice were sacrificed by an overdose injection of pentobarbital (500 mg/kg i.p). The tracheal was exposed and intubated with PE-60 tubing (0.72-mm inner diameter, 1.22-mm outer diameter). Three aliquots of 0.3 ml sterilized saline were instilled and retrieved as the bronchoalveolar lavage fluid (BALF). Return volume was recorded and was consistently > 60% of the instilled volume. The BALF was then centrifuged at 1000 × g for 10 min at 4°C. The supernatant was aliquoted and stored at -80°C until assay. The remaining cell pellet was resuspended in 1 ml PBS solution. Total cell counts were determined using a haemocytometry, by adding 100 μl of the cell suspension to 100 μl trypan blue stain. Differential cell counts were performed on cytocentrifuge preparations (Cytospin 2; Shandon, UK) stained with Wright-Giemsa by counting 200 cells under × 400 magnification from each individual by the same inspector in a blinded manner. Cells were identified by standard morphology and differentiated into neutrophils, eosinophils, lymphocytes, and macrophages.
Concentrations of soluble mediators in BALF supernatants were determined by enzyme-linked immunosorbent assay (ELISA), as previously described [19]. Measurements of IFN-γ, IL-6 and IL-13 were performed by using commercial kits (R&D Systems China Co. Ltd., Shanghai, China), following the manufacturer's protocol. The limits of detection were 3 pg/ml for IL-6 and IL-13, and 10 pg/ml for IFN-γ.

Processing of lung tissues and histological scoring of lung inflammation

After BAL, the left lung lobe was removed and placed in 10% neutral-buffered formalin solution and embedded into paraffin. The remains were microdissected and placed into liquid nitrogen and then kept in -80°C until further process. Four-micrometer paraffin sections were placed onto Fisher PLUS slides. After deparaffinization and rehydration, some paraffin sections were stained with hematoxylin and eosin (H&E), dehydrated, and mounted.
Slides were coded and graded by two independent investigators in a blinded fashion using a reproducible scoring system described elsewhere [20]. A value from 0 to 3 per criterion was adjudged to each tissue section scored. Two criteria were scored to document the pulmonary inflammation: peribronchial inflammation and perivascular inflammation. A value of 0 was adjudged when no inflammation was detectable, a value of 1 for occasional cuffing with inflammatory cells, a value of 2 for most bronchi or vessels surrounded by thin layer (one to five cells) of inflammatory cells and a value of 3 when most bronchi or vessels were surrounded by a thick layer (more than five cells) of inflammatory cells. Total lung inflammation was defined as the average of the peribronchial and perivascular inflammation scores. 2 ~ 3 tissue sections per mouse were scored, and the inflammation scores was expressed as a mean value of 15 ~ 20 sections per group.

Immunoblotting

Mouse lung tissues were first incubated in ice-cold 1 × cell lysis buffer(Cell signaling technology, Mass., US) plus 1 mM PMSF and a protease inhibitor cocktail (Sigma Aldrich, St. Louis, MO), as well as a serine/threonine phosphatase inhibitor cocktail2 (Sigma-Aldrich, St. Louis, MO4) for 30 minutes and lysed using Fastprep system (MP Biomedicals, Germany). The Homogenates were centrifuged at 15,000 × g for 15 minutes at 4°C and the concentration of proteins in the supernatants fractions was determined by BCA Protein Assay Kit (Pierce, Thermo Scientific Co, US). Equal amounts of protein (75 μg) from blindly selected samples were renumbered and subjected to 10% SDS-PAGE. Separated proteins were transferred to PVDF membranes (iBlot™ Dry Blotting System, Invitrogen) after electrophoresis at 90 V for 7 min. Membranes were blocked with 5% nonfat dry milk dissolved in TBST buffer (10 mM Tris hCl (pH 7.5), 150 mM NaCl, 0.1% Tween-20) overnight at 4°C. Binding of the primary antibody was detected by infrared dye-conjugated secondary antibodies and Odyssey system (Li-Cor, USA), and the bands were quantified with densitometry. The source of the antibodies and dilutions used were as follows: mouse anti-p38 MAPK (1:5000 dilution), mouse anti-phospho-p38 MAPK (1:5000 dilution) (Cell Signaling Technology, Mass., US), mouse anti-phospho-MKP-1 (1:4000 dilution) and mouse anti-α-actin (1:1000 dilution) (Sigma Aldrich, St. Louis, MO).

Real-time reverse transcription-polymerase chain reaction

Total RNA was extracted using the TRIzol® Reagent (Invitrogen, Calif. US) according to the manufacturer's instruction. RNA (2 μg per sample) was used to synthesise single-stranded complimentary DNA (cDNA) using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, CA, USA) in a PTC-200 Peltier Thermal Cycler (MJ Research, Watertown, Mass., USA). The cDNA generated was used as a template in subsequent real-time PCR analyses. Transcript levels were determined by Applied Biosystems 7500 Real Time PCR System (Applied Biosystems, CA, USA) using SYBR Green PCR Master Mix Reagent (Qiagen, Germany). The sequences of murine forward and reverse primers used are displayed in Table 1 (Devised by Invitrogen Shanghai Ltd. China).
Table 1
Sequences of target genes
primer
Forward primer
Reverse primer
IL-5
5′-CCATGCAGAGTCCTCAGAAC AA-3′
5′- TTACTGGAA AGGCCCAAG CA-3′
IL-17
5′- CCTGGCGGCTACAGTGAAG-3′
5′- TTTGGACACGCTGAGCTTTG-3′
TNF-α
5′-AGCCGATGGGTTGTACCTTGTC TA-3′
5′-TGAGATAGCAAATCGGCTGACGGT-3′
CXCL1
5′-TGGCTGGGATTCACCTCAAGAACA-3′
5′-TGTGGCTATGACTTCGGTTTGGGT-3′
MKP-1
5′-CTGCCTTGATCAACGTCTCA-3′
5′-ACCCTTCCTCCAGCATTCTT-3′
The thermal cycling program of qPCR was as follows: Step 1, 15 min at 95°C; step 2, 15 s at 95°C; step 3: 30 s at 60°C; step 4: 30 s at 72°C, with step 2 to step 4 repeated for 40°Cycles. Relative gene expression was normalized to β-actin. Data are expressed as fold-increase in RNA expression compared with control animals, which are set at a value of 1.

Measurement of bio-markers of oxidative stress

Malondialdehyde (MDA) level was utilized to quantify the lipid peroxidation in tissues as previously described [21]. Briefly, following lung homogenate preparation, MDA content was based on the reaction of MDA with thiobarbituric acid (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China) at 95°C. Samples were then heated for 1 h at 95°C and centrifuged at 3000 g for 10 min. The absorbance of the supernatant was measured by spectrophotometry at 532 nm using 1,1,3,3-tetramethoxypropane as an external standard. MDA was expressed in micromole per g weight of wet tissue.
Glutathione peroxidase (GSH-Px) activity in lung supernatants was determined spectrophotometrically with minor modification of methods previously described [22]. The rate of GSSG formation was assessed by measuring the reduction of GSH (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China). The unit of GSH-Px activity was defined as μmpl/L reduced GSH · g protein-1 · min-1.

Statistical analysis

Data are expressed as mean ± standard error of the mean (SEM). One-way ANOVA was used to determine the statistical significance of differences among groups. S-N-K test was used to correct for multiple comparisons when statistical significances were identified in the ANOVA. Comparisons within each group are made by unpaired t-test. Criterion for significance was taken to be p ≤ 0.05. For statistical analysis, SPSS program (SPSS Inc. Chicago, IL, USA) was used. For statistical charting, GraphPad Prism 5.0 (GraphPad Software Inc., San Diego, CA) was used.

Results

Ozone impaired the inhibitory effects of dexamethasone on lung inflammation and AHR

As expected, compared with the normal controls, mice in the asthma model exhibited AHR (decreased LogPC100Penh), higher BAL number of total cells, lymphocytes, eosinophils and neutrophils, elevated BAL concentration of IFN-γ, IL-6 and IL-13, as well as higher pathological inflammatory score and mRNA expressions of inflammatory cytokines (TNF-α, IL-5, CXCL-1 and IL-17) in lung tissues (Figure 2). Subsequent intraperitoneal administration of dexamethasone (2 mg/kg), but not saline, significantly decreased the AHR, lowered the number of total cells and eosinophils, decreased the BAL concentration of IFN-γ and IL-6, attenuated the pathological inflammation and the mRNA expression of IL-5 in lung tissues (Figure 2).
In comparison with their air-exposed counterparts, O3-exposed asthmatic mice had significantly enhanced AHR, increased BAL number of neutrophils and BAL concentration of IL-13, aggravation of lung inflammation, and prominent elevations in lung mRNA expressions of TNF-α, CXCL-1 and IL-17(Figure 2).
Combined administration of dexamethasone on O3-exposed asthmatic mice failed to elicit any significant variation on AHR or pulmonary inflammation (Figure 2).

Ozone increased oxidative stress and p38 MAPK phosphorylation and differentially influenced MKP-1 gene and protein expression

As a potent exotic oxidant, O3 might exert influence on the intrinsic oxidative metabolism. We measured the MDA concentration and the GSH-Px activity in lung tissues and found that the asthmatic mice exhibited higher level of MDA concentration and lower extent of GSH-Px activity than their normal controls. Ozone exposure further significantly intensified the trends of both biomarkers. Further administration of dexamethasone changed neither of them (Figure 3A and B).
To find out whether the p38 MAPK and its phosphatase, MKP-1, get involved in the O3 interference with the dexamethasone effect, we evaluated the phosphorylation ratio of the p38 MAPK and the expression of MKP-1. Data showed that the activation of p38 MAPK was prominent in asthma model, in contrast with their normal controls. Ozone exposure on asthma model further enhanced the phosphorylation of p38 MAPK. Administration of dexamethasone on these model animals slightly decreased the phosphorylation ratio of p38 MAPK (saline 0.762 ± 0.232 vs dexamethasone 0.273 ± 0.081, p = 0.070). This trend was obscured when the experimental animals received the O3 exposure simultaneously (saline 2.571 ± 0.308 vs dexamethasone 2.378 ± 0.302, p = 0.663) (Figure 3C).
To evaluate the MKP-1 protein expression level, we measured the phosphorylated protein of this phosphatase, for the phosphorylation promotes its stability and otherwise it is quickly degraded and hence transient [23],[24]. Our data showed that both the protein and the mRNA expression of MKP-1 were elevated in allergic mice exposed to air, while the identical group of mice exposed to O3 exhibited higher expression of MKP-1 at mRNA level but not at protein level. Administration of dexamethasone on the air-exposed experimental animals increased the expression of MKP-1 at both levels, while this effect was diminished when these asthmatic mice received combined ozone inhalation (Figure 3D and E).

SB239063 restored partly the ozone-impaired effects of dexamethasone

To further demonstrate the involvements of p38 MAPK in the ozone-induced inhibition of glucocorticoids effects, an inhibitor of p38 MAPK, SB239063, was administrated in mice before and after each O3 exposure. The dosage of SB239063 were titrated in a preliminary experiment, in which three dosages of SB239063 (0.4 mg/kg, 1.2 mg/kg, 4 mg/kg; diluted with DMSO) and DMSO alone were randomly injected through tail vein in mice exposed 1.0 ppm ozone for 3 hours (usage referred to the previous study [25]). Some measurements were performed including BAL cells and AHR to evaluate the dose-dependent inhibitory effects of SB239063. The data showed that, compared with DMSO alone and other two dosages, 4 mg/kg SB239063 significantly decreased the number of BAL total cells (O3: 400.00 ± 75.86 vs O3 + SB2390634mg/kg: 170.00 ± 24.06, p = 0.028) and macrophages (O3: 367.41 ± 69.22 vs O3 + SB2390634mg/kg: 156.44 ± 46.40, p = 0.028), and slightly inhibited the ozone-induced AHR (O3: 0.315 ± 0.118 vs O3 + SB2390634mg/kg: 0.696 ± 0.118, p = 0.062) (Additional file 1: Figure S1.). Accordingly, 4 mg/kg was adopted as the dosage of SB239063 during the subsequent experiment.
Data showed that SB239063 in O3-exposed asthmatic mice significantly inhibited the AHR, decreased the number of BAL neutrophils and concentrations of IL-6 and IL-13, alleviated the perivascular lung inflammation and down-regulated the lung IL-17 mRNA expression. Furthermore, upon the delivery of SB239063 but not DMSO only, the administration of dexamethasone elicited some inhibitory effects on the AHR, the number of BAL total and eosinophil cells, the peribronchial lung inflammation, and the lung expression of IL-5 mRNA in O3-exposed asthmatic mice (Figure 4).

SB239063-induced changes in oxidative stress, the p38 MAPK phosphorylation and the expression of MKP-1

SB239063 affected neither the concentration of MDA nor the activity of GSH-Px in O3-exposed asthmatic mice (Figure 5A and B).
Compared with DMSO only, SB239063 significantly decreased the phosphorylation ratio of p38 MAPK, but not influenced the mRNA or the protein expression of MKP-1, in lung tissues of O3-exposed asthmatic mice (Figure 5C, D and E). However, upon the delivery of SB239063, further administration of dexamethasone caused a slight non-significant increase in the protein (SB2390634mg/kg: 0.656 ± 0.088 vs DMSO: 0.423 ± 0.084, p = 0.079) and mRNA (SB2390634mg/kg: 7.922 ± 1.427 vs DMSO: 5.093 ± 0.591, p = 0.092) expression of MKP-1, in O3-exposed asthmatic mice (Figure 5D and E).

Discussion

As expected, the mice of the present allergic asthma model exhibited AHR, eosinophilic airway inflammation, and elevated BAL and lung immunological cytokines. Consistently with other reports, the enhanced phosphorylation of p38 MAPK was observed in the present animal model, which may indicate the participation of this kinase in the pathophysiological process of anaphylaxis [26],[27]. Interestingly, the protein and gene expression levels of MKP-1 in the lungs of the experimental animals were also elevated. This phenomenon, which has not been reported heretofore, provides a clue that the p38 MAPK and MKP-1 might balance each other at a comparatively elevated baseline level within such an asthma model. Exposing these experimental animals to ozone enhanced the AHR, induced the neutrophilic airway inflammation, promoted the expressions of Th1 and Th17 cytokines in BAL and lung tissues, and increased the oxidative stress levels. These aggravating effects of ozone on the allergic animals have been demonstrated previously by other researchers and ourselves [12],[28],[29]. The activated p38 MAPK pathway is the cornerstone of the ozone-induced detrimental effects both on the AHR and airway inflammation in mice [7],[15]. In the present study, we illustrated that the inhibition of p38 MAPK significantly eliminated the adverse activities of ozone on the AHR and airway inflammation as well as on most of the inflammatory cytokines, such as IL-6 and IL-13 in BAL, and IL-17 in the lung tissues of the asthmatic mice (Figure 4). It is reasonable to come to a conclusion that the p38 MAPK pathway plays central roles in the influence of ozone on current asthma model. However, the mRNA expressions of several inflammatory cytokines, such as CXCL-1 and TNFα, cannot be inhibited by SB239063, which may indicate that the expressions of these pro-inflammatory cytokines, whether being induced by allergic inflammation or by ozone stimulation, are regulated by an exceedingly complicated crosstalk network in which the p38 MAPK pathway is subtly involved. However, whether the oxidant ozone affects the therapeutic effects of corticosteroids on this allergic experimental model has not been reported yet.
The inhibitory effects of dexamethasone on the current experimental model were evidenced by the decreased AHR (increased LogPC100Penh), attenuated airway inflammatory cellularity and pathologic alleviation of the lung inflammation (peri-bronchial and peri-vascular area) as well as by the lowered mRNA expression of Th2 cytokines (IL-5) in the lungs. Interestingly, these dexamethasone-induced alterations were diminished in the mice of identical background airway inflammation but received the O3 exposure simultaneously with the medication. Accordingly, it can be modestly inferred that the subacute ozone exposure in the present study impaired the inhibitory effects of dexamethasone on this classical allergic asthma model. In other words, the ozone exposure blunted the response to the corticotherapy of these allergic mice.
The underlying mechanisms of the ozone-decreased biological response to glucocorticoids action remain unclear. However, we found in current study that the O3 exposure of the asthmatic mice caused four main specific alterations differing from their air-exposed counterparts. These particular features, namely the airway neutrophilia, enhanced activation of p38 MAPK, upregulated expression of IL-17, and altered expression of MKP-1, may lead to the resistance to the subsequent delivery of dexamethasone.
Neutrophils are generally considered less responsive to GCs compared to other inflammatory cells [30]. The neutrophilic inflammation existing in the context of some chronic conditions, such as the chronic obstructive pulmonary disease (COPD) and chronic asthma, tends to be resistant to corticotherapy, whether in human patients [10],[31] or in experimental animals [32]. However, it does not mean that neutrophils are resistant to GCs by nature, for Hirsch et al. reported that neutrophils are as sensitive as other leukocytes to the inhibitory effects of glucocorticoids on the expressions of pro-inflammatory genes in peripheral blood mononuclear cells (PBMCs) of normal subjects [33]. Since the current study did not measure the direct response of neutrophils to the application of glucocorticoids, our data cannot offer further supporting information on whether a subacute oxidative stress conditions, as created in the present investigation, might render the accumulated neutrophils resistant to corticosteroids. However, some neutrophil-related cytokines, such as TNF-α, were proved to be insensitive to dexamethasone, as shown in this examination (Figure 2G). It has been previously reported by Onda et al. that TNF-α can reduce the GC-induced transactivation of endogenous genes as well as that of a reporter plasmid which contains a GC responsive element (GRE) in cultured human epidermal keratinocyte cells [34]. Furthermore, Van et al. demonstrated that TNF-α not only impaired the GR-dependent gene expression, but also downregulated the levels of both GR mRNA and protein [35]. Given the intimate relationship between TNF-α and neutrophils, it is reasonable to infer that the contribution of neutrophils to the GC insensitivity may involve the function of TNF-α.
In addition to boosting airway neutrophils, ozone exposure on present asthma model caused significant activation of p38 MAPK, a kinase involved in the pathogenesis of both the allergic and the ozone-induced airway inflammation and AHR [15],[26]. In this study, we demonstrated that the inhibition of p38 MAPK not only mitigated the aggravating effects of O3 on pulmonary inflammation and AHR, but also restored partly the O3-impaired inhibitory effects of dexamethasone. Although numerous examinations have indicated that the inhibition of p38 MAPK can promote the cellular response to corticosteroids in vitro [36]-[38], demonstrating the roles of this kinase in the pathogenesis of glucocorticoids insensitivity, the current investigation confirmed this viewpoint in vivo. The phosphorylation of GRα by the activated p38 MAPK in PBMCs is reported to contribute to the glucocorticoids insensitivity of severe asthmatic patients [39]. Besides, p38 MPAK might also exert its roles through its downstream regulatory molecules, such as IL-17.
As reported previously, the expression of IL-17 mRNA can be upregulated by ozone exposure [17],[40],[41]. However, it is by far not known how ozone initiates the upregulation of this Th17 cytokine. The results of the present research showed that the O3-elevated IL-17 mRNA expression was decreased by the SB239063, suggesting the critical functions of the activated p38 MAPK executed in the course between the O3 exposure and IL-17 expression. Besides, the pivotal roles of p38 MAPK in the synthesis of IL-17 by CD4 (+) cells have been validated by previous studies [42],[43]. Overall, we have reasons to infer that the p38 MAPK pathway might contribute to the ozone-induced upregulation of IL-17. This hypothesis is worthwhile and needs to be further substantiated in vitro.
Th17 cells and cytokines play pivotal roles in the ozone-induced airway hyperresponsiveness in mice [17]. Interestingly, the AHR of mice, induced by the transfer of Th17 cytokines or Th17-cells, is susceptible to becoming insensitive to the inhibition effects of dexamethasone [44]. In the current examination, the elevated expression of IL-17 mRNA was associated with the decreased response to dexamethasone in the ozone-exposed asthmatic mice and vice versa after they received the administration of BS239063. Actually, the contribution of Th17 cytokines to the pathogenesis of developing absence of sensitivity to glucocorticoids has already been confirmed by numerous ex vivo investigations [44]-[46]. Based on these established contributions of IL-17 to GC insensitivity, it may be hypothesized that O3 exposure of asthmatic mice may promote the production of IL-17 via the activation of p38 MAPK pathway, and hence it may impair the therapeutic effect of dexamethasone.
It is well known that MKP-1 mediates the anti-inflammatory effects of GCs [4],[47]-[49]. Impairments in the expression of this phosphatase will compromise the action of GCs. In the present study, we measured the phosphorylated MKP-1 protein instead of the total protein, considering the fact that after its synthesis, the MKP-1 protein is quickly degraded and hence is transient, while the phosphorylation by ERK1/2 on its serine residues, Serine 359 and Serine 364, promotes its stability and hence possesses phosphatase activity [23]. Moreover, given the common option that extracellular regulated protein kinases (ERK) 1/2 is involved to a lower extent in the response to extrinsic stress, it is reasonable to assume that the ozone exposure has an insignificant impact on the phosphorylation of the MKP-1 protein. Hence, the p-MKP-1 protein measured in the current investigation can be regarded as the response of the change in the total MKP-1 protein rather than the variation of the phosphorylation intensity.
Our results demonstrated that GCs induced the mRNA and protein expression of MKP-1, which is consistent with previous published data [49]. According to the conclusions originating from an earlier examination [9], this induction progress is initiated by the intracellular ROS, of which this course is vulnerable to the outer source of oxidants. In our study, we observed that the ozone exposure diminished the favorable influence of dexamethasone on mRNA and protein expression of MKP-1 in asthmatic mice. This trend was accompanied by the ozone-impaired GC therapeutic effects. Furthermore, the blockade of p38 MAPK by SB239063 not only restored a part of the curative actions of Dex, but also regained the slight conducive trend of GCs on the expression of MKP-1. These observations provide inferences that the impaired GCs induction of MKP-1 expression might contribute to the ozone-induced GC insensitivity.
Numerous studies have placed their focus on the influence of oxidative stress on the expression of MKP-1, mostly in vitro xpeiments [50],[51]. However, the manner in which the oxidative stress influences the expression of MKP-1 in vivo still remains elusive. In the present experiments, we observed that the O3 exposure in allergic mice caused higher expression of MKP-1 at the mRNA level, but, unexpectedly, not at the protein level. Although previous research illustrated that p38 MAPK mediates the oxidative stress-induced MKP-1 mRNA expression in vitro [50], we demonstrated that the inhibition of p38 MAPK did not prevent the transcriptional induction of MKP-1 mRNA by O3 in vivo. The substantiate role of p38 MAPK in the ozone induction of MKP-1 needs to be further explored in different systems with diverse stimuli. However, this is the first report, which highlights the ozone-induced discrepant alterations between the mRNA and the protein expression of MKP-1. The biological basis for this discrepancy is by far not clear. According to Kuwano et al., the H2O2 treatment promotes the translation of MKP-1 by increasing the association of the MKP-1 mRNA with its binding protein HuR and NF 90 in cultured HeLa cells [52]. However, the oxidative modification of MKP-1 protein, promoted by oxidative stress, not only inactivates MKP-1 but also targets this phosphatase for proteasomal degradation [53]. Furthermore, though the proper intracellular activation of ERKs contributes to the phosphorylation and the stability of the MKP-1 protein, the sustained activation of ERKs causes the degradation of MKP-1 protein via the ubiquitin-proteasome pathway [54]. Overall, we defined a hypothesis that this differential influence on the MKP-1 protein may result from the O3-enhanced oxidative stress-related post-translational modification, which needs to be further substantiated.

Conclusions

In summary, present study demonstrated that ozone inhalation impaired the therapeutic effects of dexamethasone on AHR and airway inflammation of mice with allergic airway disease, which can be reversed by the inhibition of p38 MAPK. The pathophysiological basis underlying the ozone-induced declining sensitivity to GCs may involve the over phosphorylation of p38 MAPK, which may further mediate the upregulated IL-17 expression, and the elevated oxidative stress, which may disturb the GCs induction of MKP-1 expression and promote the degradation of MKP-1 protein. This study put forward an alternative explanation for the ozone oxidative stress induced insensitivity to glucocorticoids therapy, and provide the identification of new therapeutic targets which may aid in the management of patients with steroids-resistant asthma.

Authors' contributions

AB conceived of the study, carried out the animal exposure studies, participated in the molecular biological experiments and drafted the manuscript. FL measured the AHR. MZ participated in the Westernblot assay. YC participated in the design of the study and performed the statistical analysis. PZ participated in its design and coordination and helped to draft the manuscript. XZ worked as a supervisor and participated in the coordination and manuscript revise. All authors read and approved the final manuscript.

Additional file

Acknowledgements

The authors would like to thank Prof. Kian Fung Chung (National Heart and Lung Institute, Imperial College, London) for the idea inspiration and the manuscript revise. We also thank Dr. Cai Yu, Dr. Wang Lingfan and Dr. He Weigang (Hutchison Medipharma Limited Co.) for their technical help.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.
Literatur
1.
Zurück zum Zitat Barnes PJ, Adcock IM: Glucocorticoid resistance in inflammatory diseases. Lancet. 2009, 373 (9678): 1905-1917. 10.1016/S0140-6736(09)60326-3.PubMedCrossRef Barnes PJ, Adcock IM: Glucocorticoid resistance in inflammatory diseases. Lancet. 2009, 373 (9678): 1905-1917. 10.1016/S0140-6736(09)60326-3.PubMedCrossRef
2.
Zurück zum Zitat King EM, Chivers JE, Rider CF, Minnich A, Giembycz MA, Newton R: Glucocorticoid repression of inflammatory gene expression shows differential responsiveness by transactivation- and transrepression-dependent mechanisms. PLoS One. 2013, 8 (1): e53936-10.1371/journal.pone.0053936.PubMedPubMedCentralCrossRef King EM, Chivers JE, Rider CF, Minnich A, Giembycz MA, Newton R: Glucocorticoid repression of inflammatory gene expression shows differential responsiveness by transactivation- and transrepression-dependent mechanisms. PLoS One. 2013, 8 (1): e53936-10.1371/journal.pone.0053936.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Andrew R, Clark JRSM, Tchen CR: Role of dual specificity phosphatases in biological responses to glucocorticoids. J Biol Chem. 2008, 283 (38): 25765-25769. 10.1074/jbc.R700053200.CrossRef Andrew R, Clark JRSM, Tchen CR: Role of dual specificity phosphatases in biological responses to glucocorticoids. J Biol Chem. 2008, 283 (38): 25765-25769. 10.1074/jbc.R700053200.CrossRef
4.
Zurück zum Zitat Furst R, Schroeder T, Eilken HM, Bubik MF, Kiemer AK, Zahler S, Vollmar AM: MAPK phosphatase-1 represents a novel anti-inflammatory target of glucocorticoids in the human endothelium. FASEB J. 2007, 21 (1): 74-80. 10.1096/fj.06-6752com.PubMedCrossRef Furst R, Schroeder T, Eilken HM, Bubik MF, Kiemer AK, Zahler S, Vollmar AM: MAPK phosphatase-1 represents a novel anti-inflammatory target of glucocorticoids in the human endothelium. FASEB J. 2007, 21 (1): 74-80. 10.1096/fj.06-6752com.PubMedCrossRef
5.
Zurück zum Zitat Maier JV, Brema S, Tuckermann J, Herzer U, Klein M, Stassen M, Moorthy A, Cato AC: Dual specificity phosphatase 1 knockout mice show enhanced susceptibility to anaphylaxis but are sensitive to glucocorticoids. Mol Endocrinol. 2007, 21 (11): 2663-2671. 10.1210/me.2007-0067.PubMedCrossRef Maier JV, Brema S, Tuckermann J, Herzer U, Klein M, Stassen M, Moorthy A, Cato AC: Dual specificity phosphatase 1 knockout mice show enhanced susceptibility to anaphylaxis but are sensitive to glucocorticoids. Mol Endocrinol. 2007, 21 (11): 2663-2671. 10.1210/me.2007-0067.PubMedCrossRef
6.
Zurück zum Zitat Abraham SM, Lawrence T, Kleiman A, Warden P, Medghalchi M, Tuckermann J, Saklatvala J, Clark AR: Antiinflammatory effects of dexamethasone are partly dependent on induction of dual specificity phosphatase 1. J Exp Med. 2006, 203 (8): 1883-1889. 10.1084/jem.20060336.PubMedPubMedCentralCrossRef Abraham SM, Lawrence T, Kleiman A, Warden P, Medghalchi M, Tuckermann J, Saklatvala J, Clark AR: Antiinflammatory effects of dexamethasone are partly dependent on induction of dual specificity phosphatase 1. J Exp Med. 2006, 203 (8): 1883-1889. 10.1084/jem.20060336.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Li F, Zhang M, Hussain F, Triantaphyllopoulos K, Clark AR, Bhavsar PK, Zhou X, Chung KF: Inhibition of p38 MAPK-dependent bronchial contraction after ozone by corticosteroids. Eur Respir J. 2011, 37 (4): 933-942. 10.1183/09031936.00021110.PubMedPubMedCentralCrossRef Li F, Zhang M, Hussain F, Triantaphyllopoulos K, Clark AR, Bhavsar PK, Zhou X, Chung KF: Inhibition of p38 MAPK-dependent bronchial contraction after ozone by corticosteroids. Eur Respir J. 2011, 37 (4): 933-942. 10.1183/09031936.00021110.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Maneechotesuwan K, Yao X, Ito K, Jazrawi E, Usmani OS, Adcock IM, Barnes PJ: Suppression of GATA-3 nuclear import and phosphorylation: a novel mechanism of corticosteroid action in allergic disease. PLoS Med. 2009, 6 (5): e1000076-10.1371/journal.pmed.1000076.PubMedPubMedCentralCrossRef Maneechotesuwan K, Yao X, Ito K, Jazrawi E, Usmani OS, Adcock IM, Barnes PJ: Suppression of GATA-3 nuclear import and phosphorylation: a novel mechanism of corticosteroid action in allergic disease. PLoS Med. 2009, 6 (5): e1000076-10.1371/journal.pmed.1000076.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Furst R, Zahler S, Vollmar AM: Dexamethasone-induced expression of endothelial mitogen-activated protein kinase phosphatase-1 involves activation of the transcription factors activator protein-1 and 3′,5′-cyclic adenosine 5′-monophosphate response element-binding protein and the generation of reactive oxygen species. Endocrinology. 2008, 149 (7): 3635-3642. 10.1210/en.2007-1524.PubMedCrossRef Furst R, Zahler S, Vollmar AM: Dexamethasone-induced expression of endothelial mitogen-activated protein kinase phosphatase-1 involves activation of the transcription factors activator protein-1 and 3′,5′-cyclic adenosine 5′-monophosphate response element-binding protein and the generation of reactive oxygen species. Endocrinology. 2008, 149 (7): 3635-3642. 10.1210/en.2007-1524.PubMedCrossRef
10.
Zurück zum Zitat Barnes PJ: Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2013, 131 (3): 636-645. 10.1016/j.jaci.2012.12.1564.PubMedCrossRef Barnes PJ: Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2013, 131 (3): 636-645. 10.1016/j.jaci.2012.12.1564.PubMedCrossRef
11.
Zurück zum Zitat Committee of the Environmental and Occupational Health Assembly of the American Thoracic Society. Am J Respir Crit Care Med. 1996, 153 (1): 3-50. 10.1164/ajrccm.153.1.8542133. Committee of the Environmental and Occupational Health Assembly of the American Thoracic Society. Am J Respir Crit Care Med. 1996, 153 (1): 3-50. 10.1164/ajrccm.153.1.8542133.
12.
Zurück zum Zitat Bao A, Liang L, Li F, Min Z, Zhou X: Effects of acute ozone exposure on lung peak allergic inflammation of mice. Front Biosci. 2013, 18: 838-851. 10.2741/4147.CrossRef Bao A, Liang L, Li F, Min Z, Zhou X: Effects of acute ozone exposure on lung peak allergic inflammation of mice. Front Biosci. 2013, 18: 838-851. 10.2741/4147.CrossRef
13.
Zurück zum Zitat Liang L, Li F, Bao A, Zhang M, Chung KF, Zhou X: Activation of p38 mitogen-activated protein kinase in ovalbumin and ozone-induced mouse model of asthma. Respirology. 2013, 18 (Supplement S3): 20-29. 10.1111/resp.12189.PubMedCrossRef Liang L, Li F, Bao A, Zhang M, Chung KF, Zhou X: Activation of p38 mitogen-activated protein kinase in ovalbumin and ozone-induced mouse model of asthma. Respirology. 2013, 18 (Supplement S3): 20-29. 10.1111/resp.12189.PubMedCrossRef
14.
Zurück zum Zitat Stenfors N, Bosson J, Helleday R, Behndig AF, Pourazar J, Tornqvist H, Kelly FJ, Frew AJ, Sandstrom T, Mudway IS, Blomberg A: Ozone exposure enhances mast-cell inflammation in asthmatic airways despite inhaled corticosteroid therapy. Inhal Toxicol. 2010, 22 (2): 133-139. 10.3109/08958370903005736.PubMedCrossRef Stenfors N, Bosson J, Helleday R, Behndig AF, Pourazar J, Tornqvist H, Kelly FJ, Frew AJ, Sandstrom T, Mudway IS, Blomberg A: Ozone exposure enhances mast-cell inflammation in asthmatic airways despite inhaled corticosteroid therapy. Inhal Toxicol. 2010, 22 (2): 133-139. 10.3109/08958370903005736.PubMedCrossRef
15.
Zurück zum Zitat Williams AS, Issa R, Durham A, Leung SY, Kapoun A, Medicherla S, Higgins LS, Adcock IM, Chung KF: Role of p38 mitogen-activated protein kinase in ozone-induced airway hyperresponsiveness and inflammation. Eur J Pharmacol. 2008, 600 (1-3): 117-122. 10.1016/j.ejphar.2008.09.031.PubMedCrossRef Williams AS, Issa R, Durham A, Leung SY, Kapoun A, Medicherla S, Higgins LS, Adcock IM, Chung KF: Role of p38 mitogen-activated protein kinase in ozone-induced airway hyperresponsiveness and inflammation. Eur J Pharmacol. 2008, 600 (1-3): 117-122. 10.1016/j.ejphar.2008.09.031.PubMedCrossRef
16.
Zurück zum Zitat Wang JM, Denis M, Fournier M, Laviolette M: Cyclosporin A increases the pulmonary eosinophilia induced by inhaled Aspergillus antigen in mice. Clin Exp Immunol. 1993, 93 (3): 323-330. 10.1111/j.1365-2249.1993.tb08180.x.PubMedPubMedCentralCrossRef Wang JM, Denis M, Fournier M, Laviolette M: Cyclosporin A increases the pulmonary eosinophilia induced by inhaled Aspergillus antigen in mice. Clin Exp Immunol. 1993, 93 (3): 323-330. 10.1111/j.1365-2249.1993.tb08180.x.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, Matangkasombut P, Zhu M, Iwakura Y, Savage PB, DeKruyff RH, Shore SA, Umetsu DT: Ozone exposure in a mouse model induces airway hyperreactivity that requires the presence of natural killer T cells and IL-17. J Exp Med. 2008, 205 (2): 385-393. 10.1084/jem.20071507.PubMedPubMedCentralCrossRef Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, Matangkasombut P, Zhu M, Iwakura Y, Savage PB, DeKruyff RH, Shore SA, Umetsu DT: Ozone exposure in a mouse model induces airway hyperreactivity that requires the presence of natural killer T cells and IL-17. J Exp Med. 2008, 205 (2): 385-393. 10.1084/jem.20071507.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Hamelmann E, Schwarze J, Takeda K, Oshiba A, Larsen GL, Irvin CG, Gelfand EW: Noninvasive measurement of airway responsiveness in allergic mice using barometric plethysmography. Am J Respir Crit Care Med. 1997, 156 (3 Pt 1): 766-775. 10.1164/ajrccm.156.3.9606031.PubMedCrossRef Hamelmann E, Schwarze J, Takeda K, Oshiba A, Larsen GL, Irvin CG, Gelfand EW: Noninvasive measurement of airway responsiveness in allergic mice using barometric plethysmography. Am J Respir Crit Care Med. 1997, 156 (3 Pt 1): 766-775. 10.1164/ajrccm.156.3.9606031.PubMedCrossRef
19.
Zurück zum Zitat Zhou LF, Zhu Y, Cui XF, Xie WP, Hu AH, Yin KS: Arsenic trioxide, a potent inhibitor of NF-kappaB, abrogates allergen-induced airway hyperresponsiveness and inflammation. Respir Res. 2006, 7: 146-10.1186/1465-9921-7-146.PubMedPubMedCentralCrossRef Zhou LF, Zhu Y, Cui XF, Xie WP, Hu AH, Yin KS: Arsenic trioxide, a potent inhibitor of NF-kappaB, abrogates allergen-induced airway hyperresponsiveness and inflammation. Respir Res. 2006, 7: 146-10.1186/1465-9921-7-146.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Lee KS, Lee HK, Hayflick JS, Lee YC, Puri KD: Inhibition of phosphoinositide 3-kinase delta attenuates allergic airway inflammation and hyperresponsiveness in murine asthma model. FASEB J. 2006, 20 (3): 455-465. 10.1096/fj.05-5045com.PubMedCrossRef Lee KS, Lee HK, Hayflick JS, Lee YC, Puri KD: Inhibition of phosphoinositide 3-kinase delta attenuates allergic airway inflammation and hyperresponsiveness in murine asthma model. FASEB J. 2006, 20 (3): 455-465. 10.1096/fj.05-5045com.PubMedCrossRef
21.
Zurück zum Zitat Hulo S, Tiesset H, Lancel S, Edme JL, Viollet B, Sobaszek A, Neviere R: AMP-activated protein kinase deficiency reduces ozone-induced lung injury and oxidative stress in mice. Respir Res. 2011, 12: 64-10.1186/1465-9921-12-64.PubMedPubMedCentralCrossRef Hulo S, Tiesset H, Lancel S, Edme JL, Viollet B, Sobaszek A, Neviere R: AMP-activated protein kinase deficiency reduces ozone-induced lung injury and oxidative stress in mice. Respir Res. 2011, 12: 64-10.1186/1465-9921-12-64.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Dolezych B, Szulinska E: Selenium modifies glutathione peroxidase activity and glutathione concentration in mice exposed to ozone-provoked oxidative stress. J Trace Elem Med Biol. 2003, 17 (2): 133-137. 10.1016/S0946-672X(03)80009-3.PubMedCrossRef Dolezych B, Szulinska E: Selenium modifies glutathione peroxidase activity and glutathione concentration in mice exposed to ozone-provoked oxidative stress. J Trace Elem Med Biol. 2003, 17 (2): 133-137. 10.1016/S0946-672X(03)80009-3.PubMedCrossRef
23.
Zurück zum Zitat Brondello JM, Pouyssegur J, McKenzie FR: Reduced MAP kinase phosphatase-1 degradation after p42/p44MAPK-dependent phosphorylation. Science. 1999, 286 (5449): 2514-2517. 10.1126/science.286.5449.2514.PubMedCrossRef Brondello JM, Pouyssegur J, McKenzie FR: Reduced MAP kinase phosphatase-1 degradation after p42/p44MAPK-dependent phosphorylation. Science. 1999, 286 (5449): 2514-2517. 10.1126/science.286.5449.2514.PubMedCrossRef
24.
Zurück zum Zitat Sohaskey ML, Ferrell JE: Activation of p42 mitogen-activated protein kinase (MAPK), but not c-Jun NH(2)-terminal kinase, induces phosphorylation and stabilization of MAPK phosphatase XCL100 in Xenopus oocytes. Mol Biol Cell. 2002, 13 (2): 454-468. 10.1091/mbc.01-11-0553.PubMedPubMedCentralCrossRef Sohaskey ML, Ferrell JE: Activation of p42 mitogen-activated protein kinase (MAPK), but not c-Jun NH(2)-terminal kinase, induces phosphorylation and stabilization of MAPK phosphatase XCL100 in Xenopus oocytes. Mol Biol Cell. 2002, 13 (2): 454-468. 10.1091/mbc.01-11-0553.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Mihaescu A, Santen S, Jeppsson B, Thorlacius H: p38 Mitogen-activated protein kinase signalling regulates vascular inflammation and epithelial barrier dysfunction in an experimental model of radiation-induced colitis. Br J Surg. 2010, 97 (2): 226-234. 10.1002/bjs.6811.PubMedCrossRef Mihaescu A, Santen S, Jeppsson B, Thorlacius H: p38 Mitogen-activated protein kinase signalling regulates vascular inflammation and epithelial barrier dysfunction in an experimental model of radiation-induced colitis. Br J Surg. 2010, 97 (2): 226-234. 10.1002/bjs.6811.PubMedCrossRef
26.
Zurück zum Zitat Kim SR, Lee KS, Park SJ, Jeon MS, Lee YC: Inhibition of p38 MAPK reduces expression of vascular endothelial growth factor in allergic airway disease. J Clin Immunol. 2012, 32 (3): 574-586. 10.1007/s10875-012-9672-5.PubMedCrossRef Kim SR, Lee KS, Park SJ, Jeon MS, Lee YC: Inhibition of p38 MAPK reduces expression of vascular endothelial growth factor in allergic airway disease. J Clin Immunol. 2012, 32 (3): 574-586. 10.1007/s10875-012-9672-5.PubMedCrossRef
27.
Zurück zum Zitat Kim SR, Lee KS, Park SJ, Min KH, Lee MH, Lee KA, Bartov O, Atlas D, Lee YC: A novel dithiol amide CB3 attenuates allergic airway disease through negative regulation of p38 mitogen-activated protein kinase. Am J Respir Crit Care Med. 2011, 183 (8): 1015-1024. 10.1164/rccm.200906-0902OC.PubMedCrossRef Kim SR, Lee KS, Park SJ, Min KH, Lee MH, Lee KA, Bartov O, Atlas D, Lee YC: A novel dithiol amide CB3 attenuates allergic airway disease through negative regulation of p38 mitogen-activated protein kinase. Am J Respir Crit Care Med. 2011, 183 (8): 1015-1024. 10.1164/rccm.200906-0902OC.PubMedCrossRef
28.
Zurück zum Zitat Larsen ST, Matsubara S, McConville G, Poulsen SS, Gelfand EW: Ozone increases airway hyperreactivity and mucus hyperproduction in mice previously exposed to allergen. J Toxicol Environ Health A. 2010, 73 (11): 738-747. 10.1080/15287391003614034.PubMedCrossRef Larsen ST, Matsubara S, McConville G, Poulsen SS, Gelfand EW: Ozone increases airway hyperreactivity and mucus hyperproduction in mice previously exposed to allergen. J Toxicol Environ Health A. 2010, 73 (11): 738-747. 10.1080/15287391003614034.PubMedCrossRef
29.
Zurück zum Zitat Kierstein S, Krytska K, Sharma S, Amrani Y, Salmon M, Panettieri RA, Zangrilli J, Haczku A: Ozone inhalation induces exacerbation of eosinophilic airway inflammation and hyperresponsiveness in allergen-sensitized mice. Allergy. 2008, 63 (4): 438-446. 10.1111/j.1398-9995.2007.01587.x.PubMedCrossRef Kierstein S, Krytska K, Sharma S, Amrani Y, Salmon M, Panettieri RA, Zangrilli J, Haczku A: Ozone inhalation induces exacerbation of eosinophilic airway inflammation and hyperresponsiveness in allergen-sensitized mice. Allergy. 2008, 63 (4): 438-446. 10.1111/j.1398-9995.2007.01587.x.PubMedCrossRef
30.
Zurück zum Zitat Schleimer RP: Glucocorticoids suppress inflammation but spare innate immune responses in airway epithelium. Proc Am Thorac Soc. 2004, 1 (3): 222-230. 10.1513/pats.200402-018MS.PubMedCrossRef Schleimer RP: Glucocorticoids suppress inflammation but spare innate immune responses in airway epithelium. Proc Am Thorac Soc. 2004, 1 (3): 222-230. 10.1513/pats.200402-018MS.PubMedCrossRef
31.
Zurück zum Zitat Ordonez CL, Shaughnessy TE, Matthay MA, Fahy JV: Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: Clinical and biologic significance. Am J Respir Crit Care Med. 2000, 161 (4 Pt 1): 1185-1190. 10.1164/ajrccm.161.4.9812061.PubMedCrossRef Ordonez CL, Shaughnessy TE, Matthay MA, Fahy JV: Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: Clinical and biologic significance. Am J Respir Crit Care Med. 2000, 161 (4 Pt 1): 1185-1190. 10.1164/ajrccm.161.4.9812061.PubMedCrossRef
32.
Zurück zum Zitat Ito K, Herbert C, Siegle JS, Vuppusetty C, Hansbro N, Thomas PS, Foster PS, Barnes PJ, Kumar RK: Steroid-resistant neutrophilic inflammation in a mouse model of an acute exacerbation of asthma. Am J Respir Cell Mol Biol. 2008, 39 (5): 543-550. 10.1165/rcmb.2008-0028OC.PubMedPubMedCentralCrossRef Ito K, Herbert C, Siegle JS, Vuppusetty C, Hansbro N, Thomas PS, Foster PS, Barnes PJ, Kumar RK: Steroid-resistant neutrophilic inflammation in a mouse model of an acute exacerbation of asthma. Am J Respir Cell Mol Biol. 2008, 39 (5): 543-550. 10.1165/rcmb.2008-0028OC.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Hirsch G, Lavoie-Lamoureux A, Beauchamp G, Lavoie JP: Neutrophils are not less sensitive than other blood leukocytes to the genomic effects of glucocorticoids. PLoS One. 2012, 7 (9): e44606-10.1371/journal.pone.0044606.PubMedPubMedCentralCrossRef Hirsch G, Lavoie-Lamoureux A, Beauchamp G, Lavoie JP: Neutrophils are not less sensitive than other blood leukocytes to the genomic effects of glucocorticoids. PLoS One. 2012, 7 (9): e44606-10.1371/journal.pone.0044606.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Onda K, Nagashima M, Kawakubo Y, Inoue S, Hirano T, Oka K: Mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase (MEK-1/ERK) inhibitors sensitize reduced glucocorticoid response mediated by TNFalpha in human epidermal keratinocytes (HaCaT). Biochem Biophys Res Commun. 2006, 351 (1): 266-272. 10.1016/j.bbrc.2006.10.032.PubMedCrossRef Onda K, Nagashima M, Kawakubo Y, Inoue S, Hirano T, Oka K: Mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase (MEK-1/ERK) inhibitors sensitize reduced glucocorticoid response mediated by TNFalpha in human epidermal keratinocytes (HaCaT). Biochem Biophys Res Commun. 2006, 351 (1): 266-272. 10.1016/j.bbrc.2006.10.032.PubMedCrossRef
35.
Zurück zum Zitat Van Bogaert T, Vandevyver S, Dejager L, Van Hauwermeiren F, Pinheiro I, Petta I, Engblom D, Kleyman A, Schutz G, Tuckermann J, Libert C: Tumor necrosis factor inhibits glucocorticoid receptor function in mice: a strong signal toward lethal shock. J Biol Chem. 2011, 286 (30): 26555-26567. 10.1074/jbc.M110.212365.PubMedPubMedCentralCrossRef Van Bogaert T, Vandevyver S, Dejager L, Van Hauwermeiren F, Pinheiro I, Petta I, Engblom D, Kleyman A, Schutz G, Tuckermann J, Libert C: Tumor necrosis factor inhibits glucocorticoid receptor function in mice: a strong signal toward lethal shock. J Biol Chem. 2011, 286 (30): 26555-26567. 10.1074/jbc.M110.212365.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Goleva E, Li LB, Leung DY: IFN-gamma reverses IL-2- and IL-4-mediated T-cell steroid resistance. Am J Respir Cell Mol Biol. 2009, 40: 223-230. 10.1165/rcmb.2007-0327OC.PubMedPubMedCentralCrossRef Goleva E, Li LB, Leung DY: IFN-gamma reverses IL-2- and IL-4-mediated T-cell steroid resistance. Am J Respir Cell Mol Biol. 2009, 40: 223-230. 10.1165/rcmb.2007-0327OC.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Bhavsar P, Khorasani N, Hew M, Johnson M, Chung KF: Effect of p38 MAPK inhibition on corticosteroid suppression of cytokine release in severe asthma. Eur Respir J. 2010, 35: 750-756. 10.1183/09031936.00071309.PubMedCrossRef Bhavsar P, Khorasani N, Hew M, Johnson M, Chung KF: Effect of p38 MAPK inhibition on corticosteroid suppression of cytokine release in severe asthma. Eur Respir J. 2010, 35: 750-756. 10.1183/09031936.00071309.PubMedCrossRef
38.
Zurück zum Zitat Mercado N, Hakim A, Kobayashi Y, Meah S, Usmani OS, Chung KF, Barnes PJ, Ito K: Restoration of corticosteroid sensitivity by p38 mitogen activated protein kinase inhibition in peripheral blood mononuclear cells from severe asthma. PLoS One. 2012, 7: e41582-10.1371/journal.pone.0041582.PubMedPubMedCentralCrossRef Mercado N, Hakim A, Kobayashi Y, Meah S, Usmani OS, Chung KF, Barnes PJ, Ito K: Restoration of corticosteroid sensitivity by p38 mitogen activated protein kinase inhibition in peripheral blood mononuclear cells from severe asthma. PLoS One. 2012, 7: e41582-10.1371/journal.pone.0041582.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF, Adcock IM: p38 Mitogen-activated protein kinase-induced glucocorticoid receptor phosphorylation reduces its activity: role in steroid-insensitive asthma. J Allergy Clin Immunol. 2002, 109: 649-657. 10.1067/mai.2002.122465.PubMedCrossRef Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF, Adcock IM: p38 Mitogen-activated protein kinase-induced glucocorticoid receptor phosphorylation reduces its activity: role in steroid-insensitive asthma. J Allergy Clin Immunol. 2002, 109: 649-657. 10.1067/mai.2002.122465.PubMedCrossRef
40.
Zurück zum Zitat Kasahara DI, Kim HY, Williams AS, Verbout NG, Tran J, Si H, Wurmbrand AP, Jastrab J, Hug C, Umetsu DT, Shore SA: Pulmonary inflammation induced by subacute ozone is augmented in adiponectin-deficient mice: role of IL-17A. J Immunol. 2012, 188 (9): 4558-4567. 10.4049/jimmunol.1102363.PubMedPubMedCentralCrossRef Kasahara DI, Kim HY, Williams AS, Verbout NG, Tran J, Si H, Wurmbrand AP, Jastrab J, Hug C, Umetsu DT, Shore SA: Pulmonary inflammation induced by subacute ozone is augmented in adiponectin-deficient mice: role of IL-17A. J Immunol. 2012, 188 (9): 4558-4567. 10.4049/jimmunol.1102363.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Zhou S, Potts EN, Cuttitta F, Foster WM, Sunday ME: Gastrin-releasing peptide blockade as a broad-spectrum anti-inflammatory therapy for asthma. Proc Natl Acad Sci U S A. 2011, 108: 2100-2105. 10.1073/pnas.1014792108.PubMedPubMedCentralCrossRef Zhou S, Potts EN, Cuttitta F, Foster WM, Sunday ME: Gastrin-releasing peptide blockade as a broad-spectrum anti-inflammatory therapy for asthma. Proc Natl Acad Sci U S A. 2011, 108: 2100-2105. 10.1073/pnas.1014792108.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Gulen MF, Kang Z, Bulek K, Youzhong W, Kim TW, Chen Y, Altuntas CZ, Sass Bak-Jensen K, McGeachy MJ, Do JS, Xiao H, Delgoffe GM, Min B, Powell JD, Tuohy VK, Cua DJ, Li X: The receptor SIGIRR suppresses Th17 cell proliferation via inhibition of the interleukin-1 receptor pathway and mTOR kinase activation. Immunity. 2010, 32 (1): 54-66. 10.1016/j.immuni.2009.12.003.PubMedPubMedCentralCrossRef Gulen MF, Kang Z, Bulek K, Youzhong W, Kim TW, Chen Y, Altuntas CZ, Sass Bak-Jensen K, McGeachy MJ, Do JS, Xiao H, Delgoffe GM, Min B, Powell JD, Tuohy VK, Cua DJ, Li X: The receptor SIGIRR suppresses Th17 cell proliferation via inhibition of the interleukin-1 receptor pathway and mTOR kinase activation. Immunity. 2010, 32 (1): 54-66. 10.1016/j.immuni.2009.12.003.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Commodaro AG, Bombardieri CR, Peron JP, Saito KC, Guedes PM, Hamassaki DE, Belfort RN, Rizzo LV, Belfort R, de Camargo MM: p38{alpha} MAP kinase controls IL-17 synthesis in vogt-koyanagi-harada syndrome and experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2010, 51: 3567-3574. 10.1167/iovs.09-4393.PubMedCrossRef Commodaro AG, Bombardieri CR, Peron JP, Saito KC, Guedes PM, Hamassaki DE, Belfort RN, Rizzo LV, Belfort R, de Camargo MM: p38{alpha} MAP kinase controls IL-17 synthesis in vogt-koyanagi-harada syndrome and experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2010, 51: 3567-3574. 10.1167/iovs.09-4393.PubMedCrossRef
44.
Zurück zum Zitat McKinley L, Alcorn JF, Peterson A, Dupont RB, Kapadia S, Logar A, Henry A, Irvin CG, Piganelli JD, Ray A, Kolls JK: TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J Immunol. 2008, 181 (6): 4089-4097. 10.4049/jimmunol.181.6.4089.PubMedPubMedCentralCrossRef McKinley L, Alcorn JF, Peterson A, Dupont RB, Kapadia S, Logar A, Henry A, Irvin CG, Piganelli JD, Ray A, Kolls JK: TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J Immunol. 2008, 181 (6): 4089-4097. 10.4049/jimmunol.181.6.4089.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Zijlstra GJ, Ten Hacken NH, Hoffmann RF, van Oosterhout AJ, Heijink IH: Interleukin-17A induces glucocorticoid insensitivity in human bronchial epithelial cells. Eur Respir J. 2012, 39 (2): 439-445. 10.1183/09031936.00017911.PubMedCrossRef Zijlstra GJ, Ten Hacken NH, Hoffmann RF, van Oosterhout AJ, Heijink IH: Interleukin-17A induces glucocorticoid insensitivity in human bronchial epithelial cells. Eur Respir J. 2012, 39 (2): 439-445. 10.1183/09031936.00017911.PubMedCrossRef
46.
Zurück zum Zitat Vazquez-Tello A, Halwani R, Hamid Q, Al-Muhsen S: Glucocorticoid receptor-beta up-regulation and steroid resistance induction by IL-17 and IL-23 cytokine stimulation in peripheral mononuclear cells. J Clin Immunol. 2013, 33 (2): 466-478. 10.1007/s10875-012-9828-3.PubMedCrossRef Vazquez-Tello A, Halwani R, Hamid Q, Al-Muhsen S: Glucocorticoid receptor-beta up-regulation and steroid resistance induction by IL-17 and IL-23 cytokine stimulation in peripheral mononuclear cells. J Clin Immunol. 2013, 33 (2): 466-478. 10.1007/s10875-012-9828-3.PubMedCrossRef
47.
Zurück zum Zitat Newton R, King EM, Gong W, Rider CF, Staples KJ, Holden NS, Bergmann MW: Glucocorticoids inhibit IL-1beta-induced GM-CSF expression at multiple levels: roles for the ERK pathway and repression by MKP-1. Biochem J. 2010, 427 (1): 113-124. 10.1042/BJ20091038.PubMedCrossRef Newton R, King EM, Gong W, Rider CF, Staples KJ, Holden NS, Bergmann MW: Glucocorticoids inhibit IL-1beta-induced GM-CSF expression at multiple levels: roles for the ERK pathway and repression by MKP-1. Biochem J. 2010, 427 (1): 113-124. 10.1042/BJ20091038.PubMedCrossRef
48.
Zurück zum Zitat Lasa M, Abraham SM, Boucheron C, Saklatvala J, Clark AR: Dexamethasone causes sustained expression of mitogen-activated protein kinase (MAPK) phosphatase 1 and phosphatase-mediated inhibition of MAPK p38. Mol Cell Biol. 2002, 22 (22): 7802-7811. 10.1128/MCB.22.22.7802-7811.2002.PubMedPubMedCentralCrossRef Lasa M, Abraham SM, Boucheron C, Saklatvala J, Clark AR: Dexamethasone causes sustained expression of mitogen-activated protein kinase (MAPK) phosphatase 1 and phosphatase-mediated inhibition of MAPK p38. Mol Cell Biol. 2002, 22 (22): 7802-7811. 10.1128/MCB.22.22.7802-7811.2002.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Quante T, Ng YC, Ramsay EE, Henness S, Allen JC, Parmentier J, Ge Q, Ammit AJ: Corticosteroids reduce IL-6 in ASM cells via up-regulation of MKP-1. Am J Respir Cell Mol Biol. 2008, 39 (2): 208-217. 10.1165/rcmb.2007-0014OC.PubMedCrossRef Quante T, Ng YC, Ramsay EE, Henness S, Allen JC, Parmentier J, Ge Q, Ammit AJ: Corticosteroids reduce IL-6 in ASM cells via up-regulation of MKP-1. Am J Respir Cell Mol Biol. 2008, 39 (2): 208-217. 10.1165/rcmb.2007-0014OC.PubMedCrossRef
50.
Zurück zum Zitat Li J, Gorospe M, Hutter D, Barnes J, Keyse SM, Liu Y: Transcriptional induction of MKP-1 in response to stress is associated with histone H3 phosphorylation-acetylation. Mol Cell Biol. 2001, 21 (23): 8213-8224. 10.1128/MCB.21.23.8213-8224.2001.PubMedPubMedCentralCrossRef Li J, Gorospe M, Hutter D, Barnes J, Keyse SM, Liu Y: Transcriptional induction of MKP-1 in response to stress is associated with histone H3 phosphorylation-acetylation. Mol Cell Biol. 2001, 21 (23): 8213-8224. 10.1128/MCB.21.23.8213-8224.2001.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Xu Q, Konta T, Nakayama K, Furusu A, Moreno-Manzano V, Lucio-Cazana J, Ishikawa Y, Fine LG, Yao J, Kitamura M: Cellular defense against H2O2-induced apoptosis via MAP kinase-MKP-1 pathway. Free Radic Biol Med. 2004, 36 (8): 985-993. 10.1016/j.freeradbiomed.2004.01.009.PubMedCrossRef Xu Q, Konta T, Nakayama K, Furusu A, Moreno-Manzano V, Lucio-Cazana J, Ishikawa Y, Fine LG, Yao J, Kitamura M: Cellular defense against H2O2-induced apoptosis via MAP kinase-MKP-1 pathway. Free Radic Biol Med. 2004, 36 (8): 985-993. 10.1016/j.freeradbiomed.2004.01.009.PubMedCrossRef
52.
Zurück zum Zitat Kuwano Y, Kim HH, Abdelmohsen K, Pullmann R, Martindale JL, Yang X, Gorospe M: MKP-1 mRNA stabilization and translational control by RNA-binding proteins HuR and NF90. Mol Cell Biol. 2008, 28 (14): 4562-4575. 10.1128/MCB.00165-08.PubMedPubMedCentralCrossRef Kuwano Y, Kim HH, Abdelmohsen K, Pullmann R, Martindale JL, Yang X, Gorospe M: MKP-1 mRNA stabilization and translational control by RNA-binding proteins HuR and NF90. Mol Cell Biol. 2008, 28 (14): 4562-4575. 10.1128/MCB.00165-08.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Kim HS, Ullevig SL, Zamora D, Lee CF, Asmis R: Redox regulation of MAPK phosphatase 1 controls monocyte migration and macrophage recruitment. Proc Natl Acad Sci U S A. 2012, 109 (41): E2803-E2812. 10.1073/pnas.1212596109.PubMedPubMedCentralCrossRef Kim HS, Ullevig SL, Zamora D, Lee CF, Asmis R: Redox regulation of MAPK phosphatase 1 controls monocyte migration and macrophage recruitment. Proc Natl Acad Sci U S A. 2012, 109 (41): E2803-E2812. 10.1073/pnas.1212596109.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Lin YW, Chuang SM, Yang JL: ERK1/2 achieves sustained activation by stimulating MAPK phosphatase-1 degradation via the ubiquitin-proteasome pathway. J Biol Chem. 2003, 278 (24): 21534-21541. 10.1074/jbc.M301854200.PubMedCrossRef Lin YW, Chuang SM, Yang JL: ERK1/2 achieves sustained activation by stimulating MAPK phosphatase-1 degradation via the ubiquitin-proteasome pathway. J Biol Chem. 2003, 278 (24): 21534-21541. 10.1074/jbc.M301854200.PubMedCrossRef
Metadaten
Titel
Impact of ozone exposure on the response to glucocorticoid in a mouse model of asthma: involvements of p38 MAPK and MKP-1
verfasst von
Aihua Bao
Feng Li
Min Zhang
Yuqing Chen
Pengyu Zhang
Xin Zhou
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2014
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-014-0126-x

Weitere Artikel der Ausgabe 1/2014

Respiratory Research 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.