SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide‑induced acute lung injury

  • Authors:
    • Guirong Li
    • Youai Dai
    • Jianxin Tan
    • Jian Zou
    • Xiaowei Nie
    • Zhenkun Yang
    • Jingjing Zhao
    • Xusheng Yang
    • Jingyu Chen
  • View Affiliations

  • Published online on: June 4, 2020     https://doi.org/10.3892/mmr.2020.11214
  • Pages: 1656-1662
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute lung injury (ALI) is characterized by acute hypoxic respiratory failure, pulmonary edema and inflammatory infiltration. ALI has a high mortality rate (~30%) in the clinical setting; therefore, focusing on the treatment of lung edema and inflammatory responses in ALI is of significance. The present study investigated the effect of the p38 mitogen‑activated protein kinase (p38MAPK) inhibitor, SB203580, on lung edema and inflammatory responses in ALI in vivo. A mouse model of ALI was established to assess the effect of SB203580 on edema, proinflammatory cytokine production, and the expression of interferon regulatory factor 5 (IRF5) and inducible nitric oxide synthase (iNOS) in lung tissues using immunoblotting, immunohistochemistry, immunofluorescence, hematoxylin and eosin staining, and ELISA. SB203580 inhibited LPS‑induced lung injury and proinflammatory cytokine expression, including tumor necrosis factor‑α and interleukin‑1β. SB203580 also downregulated LPS‑induced IRF5 and iNOS expression, which are widely used as markers of proinflammatory macrophages. Collectively, the present study demonstrated that SB203580 protected against inflammatory responses and lung injury by inhibiting lung edema and downregulating proinflammatory mediators in LPS‑induced lung injury.

Introduction

Acute lung injury (ALI) is a clinical syndrome characterized by acute hypoxemic respiratory failure, pulmonary infiltration and edema in the lung tissues, of which the most severe manifestation is acute respiratory distress syndrome (1). The mortality rate of patients with ALI is ~30%, which has not changed over the past several years (2,3). The increased permeability of the alveolocapillary membrane is an important physiological alteration that is observed in patients with ALI, which is induced by alveolocapillary barrier dysfunction and epithelial injury. Increased permeability of the alveolocapillary membrane causes lung edema due to infiltration of large amounts of intravascular protein-rich fluid into the pulmonary interstitial and alveolar space (4). Moreover, overwhelming inflammation is also a critical pathophysiological process observed in patients with ALI (5). Previous studies suggested that the extensive infiltration of neutrophils and the presence of resident and recruited macrophages could mediate robust inflammatory responses in patients with ALI and animal models of ALI (6,7). Immune cells that accumulate in lung tissues release various proinflammatory mediators, such as interleukin (IL)-6, tumor necrosis factor (TNF)-α and interferon (IFN)-γ, which exacerbate inflammation. Moreover, immune cells, including proinflammatory macrophages, produce nitric oxide (NO) (8,9).

Interferon regulatory factor 5 (IRF5) is widely expressed in immune cells, including macrophages, B-cells, monocytes and dendritic cells (1012). Previous studies demonstrated that IRF5 mediated the activation of inflammatory macrophages in vivo and classical macrophages in vitro (1012). Activation of proinflammatory macrophages leads to excessive secretion of TNF-α, IL-6, IL-12 and IL-23, and downregulation of IL-10 and transforming growth factor-β (13). Therefore, the expression of IRF5 may also promote inflammatory macrophage activation and inflammatory responses in lung tissue.

It has been reported that p38 mitogen-activated protein kinase (p38MAPK) serves a crucial role in intracellular inflammatory signaling pathways, including the TNF and TLR4 signaling pathways (14). However, the effects of SB203580, an inhibitor of p38MAPK, on lung tissue injury and the inflammatory response are not completely understood. In the present study, a mouse model of ALI was established by 24-h lipopolysaccharide (LPS) stimulation. The aim of the present study was to investigate the effect of SB203580 on inflammatory response and lung injury in ALI.

Materials and methods

Animals and treatment

In the present study, a total of 45 healthy male C57BL/6 mice (age, 8 weeks; weight, 20–25 g) were purchased from Changzhou Cavans Experimental Animal Co., Ltd. The animals were kept under standardized conditions with a mean room temperature of 22–24°C, a 12/12h light/dark cycle and a humidity of 50–60%. The mice were fed a standard animal diet with food and tap water ad libitum. The mice were randomly divided into three groups (Ctrl, ALI and SB203580 groups), each containing 15 mice. All experiments were approved by The Institutional Animal Care and Use Committee at Nanjing Medical University.

LPS causes ALI after intratracheal administration and reduces associated non-pulmonary organ dysfunction, whereas intravenous administration does not lead to tissue-specific lung injury (15,16). Therefore, in the present study, mice were treated with 50 µl (5 mg/kg) LPS (Escherichia coli O111:B4; cat. no. L2630; Sigma-Aldrich; Merck KGaA) by intratracheal injection under anesthesia with 40 mg/kg pentobarbital sodium by intraperitoneal injection. In the control (Ctrl) group, mice were treated with an equal volume of PBS by intratracheal injection. The absorption rate of intraperitoneal injection is lower compared with intratracheal injection, therefore, SB203580 injection was performed prior to LPS treatment. Thus, in the SB203580 group, mice received 20 mg/kg SB203580 (cat. no. S1863; Beyotime Institute of Biotechnology) by intraperitoneal injection 1 h before LPS stimulation. A total of 30 mice were anesthetized by isoflurane inhalation (5%) and then sacrificed by cervical dislocation at 24 h post-LPS treatment. For the histological examination, immunohistochemistry and immunofluorescence assays, a further 15 mice were anesthetized by the intraperitoneal injection of pentobarbital sodium (40 mg/kg) and sacrificed by myocardial perfusion fixation after LPS stimulation for 24 h. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected for subsequent analysis at 24 h post-LPS treatment, since the features of ALI in the model are close to the diagnostic criteria of patients with ALI at this time point (15,17).

Histological examination of lung tissue

At 24 h post-LPS treatment, tissue from the middle lobe of the right lung was collected, fixed with 10% formaldehyde solution at room temperature for 24 h, dehydrated using a graded ethanol series, embedded in paraffin and cut into 4-µm sections. The sections were stained with hematoxylin for 10 min, followed by eosin for 3 min, at room temperature. Sections were observed under a BX53 light microscope (magnification, ×400; Olympus Corporation). Lung tissue injuries were scored according to a previous study (18).

Evaluation of pulmonary edema

Native tissue from the right lower lobe of the lung was weighed to obtain the ‘wet weight’ (W). Subsequently, to obtain the ‘dry weight’ (D), the tissues was dried at 65°C for 72 h and then weighed. Pulmonary edema was evaluated by calculating the W/D ratio.

BALF collection and cell counting

The pulmonary lavage was performed three times with 1 ml PBS. The collected BALF was centrifuged at 845 × g for 10 min at 4°C. Subsequently, the cell pellet was resuspended with 1 ml PBS. The total number of BALF cells was counted using a hematocytometer under a light microscope (magnification, ×200).

Immunohistochemistry

To assess the expression and distribution of IRF5 in lung tissue, the middle lobe of the right lung was fixed in 10% formaldehyde at room temperature for 24 h, then dehydrated and embedded in paraffin. All samples in paraffin were cut into 4-µm sections. Subsequently, 4-µm sections were dewaxed and rehydrated through graded alcohol at room temperature and then subjected to antigen retrieval by high pressure in 10 mmol/l citrate acid (pH 6.0) for 8 min. The sections were blocked with 10% goat serum (cat. no. AR0009; Boster Biological Technology) at room temperature for 1 h. The sections were incubated with an IRF5 primary antibody (cat. no. ab181553; 1:600; Abcam) at 4°C overnight. The sections were washed using PBS and then were incubated with a goat anti-rabbit biotinylated secondary antibody (cat. no. ab6720; 1:1,000; Abcam) at room temperature for 1.5 h, and streptavidin-horseradish peroxidase (HRP; 1:10,000; cat. no. ab7403; Abcam) at room temperature for 45 min, respectively. Chromagen detection was performed with the DAB substrate kit (cat. no. ab64238; Abcam), according to the manufacturer's instructions. Lung tissue sections were counterstained with hematoxylin for 8 min at room temperature and rinsed with distilled water, dehydrated through gradient ethanol and xylene, then mounted with mounting medium (cat. no. 14177; Cell Signaling Technology, Inc.). Images were captured using a BX53 light microscope (magnification, ×400; Olympus Corporation). Quantification was performed by calculating the percentage of IRF5-positive cells. IRF5-positive cells and total cells were counted in five sections and five different fields/section.

Western blot analysis of the lung tissues

Native tissue from the left lobe of the lung was homogenized and protein samples were extracted using RIPA buffer (cat. no. P0013B; Beyotime Institute of Biotechnology). Protein content was determined using BCA kit (cat. no. P0012S; Beyotime Institute of Biotechnology). Proteins (30 µg/line) were separated via SDS-PAGE on 10% gel and transferred to PVDF membranes. PVDF membranes were blocked using 5% non-fat milk at room temperature for 1 h and then incubated with primary antibodies targeted against IRF5 (cat. no. ab181553; 1:1,000; Abcam), inducible NO synthase (iNOS; cat. no. ab15323; 1:1,000; Abcam), arginase 1 (Arg1; cat. no. 93668; 1:1,000; Cell Signaling Technology, Inc.) and β-actin (cat. no. ab8227; 1:2,000; Abcam) at 4°C overnight. Following primary incubation, membranes were incubated with HRP-conjugated goat anti-rabbit IgG (cat. no. A0208; 1:1000; Beyotime Institute of Biotechnology) secondary antibodies at room temperature for 1.5 h. In addition, enhanced chemiluminescence reagents (cat. no. WBKLS0100; EMD Millipore) were used for imaging and quantification was performed using ImageJ software version 1.47 (National Institutes of Health) with β-actin as the loading control.

Immunofluorescence

Following treatment with LPS for 24 h, mice were subjected to myocardial perfusion fixation to preserve tissue. Tissue from the left lung was fixed with 10% formaldehyde solution at room temperature for 4 h, dehydrated in 30% sucrose, embedded in optimal cutting temperature reagent and cut into 7-µm sections using a freezing microtome. The sections were blocked with 10% goat serum (cat. no. AR0009; Boster Biological Technology) at room temperature for 40 min. The frozen lung sections were stained with F4/80 (1:50; cat. no. ab16911; Abcam) and iNOS (1:50; cat. no. ab15323; Abcam) primary antibodies at 4°C overnight. Subsequently, the sections were incubated with anti-rat Alexa Fluor® 594-conjugated (1:500; cat. no. A-11007; Invitrogen; Thermo Fisher Scientific, Inc.) and anti-rabbit Alexa Fluor® 488-conjugated (1:500; cat. no. A-11034; Invitrogen; Thermo Fisher Scientific, Inc.) secondary antibodies at room temperature for 2 h. The localization of protein was examined by confocal laser scanning microscopy (magnification, ×200; Leica Microsystems GmbH).

Measurements of cytokines

Following LPS treatment for 24 h, lung tissues were ground into protein homogenate. The levels of the proinflammatory cytokines TNF-α (cat. no. MTA00B) and IL-1β (cat. no. MLB00C) were measured using ELISA kits (both R&D Systems, Inc.) according to the manufacturer's instructions.

Statistical analysis

All experiments were repeated at least three times and the data are presented as the mean ± SEM. Statistical analyses were performed using GraphPad Prism software (version 7; GraphPad Software, Inc.). Multiple comparisons were analyzed using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

SB203580 attenuates LPS-induced lung histopathological alterations

Histopathological alterations in the lung were detected using hematoxylin and eosin (H&E) staining (Fig. 1). Lung sections of Ctrl group showed a normal alveolar morphology (Fig. 1A). Histological sections from the lungs of LPS-induced mice displayed edema, as well as basal membrane and alveolar wall thickening (Fig. 1B). By contrast, treatment with SB203580 markedly attenuated LPS-induced lung injury (Fig. 1C). The lung injury score in the ALI group was significantly increased compared with the Ctrl group. However, treatment with SB203580 significantly decreased the lung injury score compared with the ALI group (Fig. 1D). Thus, the intraperitoneal injection of SB203580 significantly decreased lung injury in ALI model mice.

SB203580 reduces LPS-induced lung edema and inflammatory responses in vivo

Lung tissue edema was assessed by calculating the W/D weight ratio. The lung W/D ratio was significantly decreased in the SB203580 group compared with the ALI group (Fig. 2A). In addition, inflammation was evaluated by calculating the total number of cells in the BALF and measuring proinflammatory cytokine levels in lung tissues. Total cell counts in the BALF were higher in the ALI group compared with the Ctrl group. However, treatment with SB203580 significantly decreased cell counts in total BALF compared with the ALI group (Fig. 2B). Similarly, the levels of proinflammatory cytokines TNF-α and IL-1β were significantly elevated in the ALI group compared with the Ctrl group, and treatment with SB203580 significantly decreased the levels of TNF-α and IL-1β compared with the ALI group (Fig. 2C and D).

SB203580 inhibits IRF5 expression in the lung tissues of ALI model mice

The effect of SB203580 on IRF5 expression was assessed by western blotting and immunohistochemical staining. The levels of IRF5 were significantly increased in ALI model mice compared with Ctrl mice. Treatment with SB203580 resulted in a significant decrease in IRF5 expression levels compared with the ALI group (Fig. 3A and B). IRF5 expression was further investigated by immunohistochemical staining (Fig. 3C). The frequency of IRF5-postive cells was significantly increased in the lungs in the ALI group compared with the Ctrl group; however, treatment with SB203580 significantly inhibited LPS-induced IRF5 expression (Fig. 3D).

SB203580 decreases the expression of iNOS and F4/80 in the lung tissues of ALI model mice

iNOS is an inflammatory gene marker, which serves a critical role in LPS-induced ALI (19). In the present study, the effect of SB203580 on iNOS expression was evaluated using western blotting and immunofluorescence assays. The expression of iNOS was significantly increased in the lung tissues of ALI model mice compared with Ctrl mice. However, LPS-induced iNOS expression was significantly decreased following SB203580 treatment (Fig. 4A and C). The levels of Arg1, were low in the Ctrl group and remained unchanged in the ALI group (Fig. 4B).

F4/80 is widely used as a macrophage marker, although it is also expressed on monocytes and neutrophils in mice (20,21). The expression of F4/80 was increased in ALI lung tissues compared with Ctrl lung tissues, which was inhibited by SB203580 treatment (Fig. 4C).

Discussion

ALI is a common clinical condition characterized by hypoxia, edema and inflammatory cell infiltration of neutrophils and macrophages into lung tissues, leading to respiratory failure (22,23). Alveolar epithelium and capillary endothelium injuries increase alveolar barrier permeability, resulting in edema, inflammatory leukocyte infiltration and bleeding (4,24). Focusing on the treatment of lung edema and inflammatory injury in patients with ALI is of significance. Mouse models of LPS-induced lung injury have been widely used to study the pathogenesis of ALI (4,18). The mild LPS-induced mouse model of ALI used in the present study displayed edema and inflammation in lung tissues.

Inhibition of proinflammatory cytokine production allows the regulation of inflammatory responses (25). The p38MAPK signaling pathway has a key regulatory role in inflammatory processes taking place in the lung (26). M39, an inhibitor of p38MAPK, inhibits the activation of p38MAPK in neutrophils and macrophages in vitro, which prevents the release of TNF-α and macrophage inflammatory protein 2, limits the activation of p38MAPK in vivo and reduces the accumulation of neutrophils in the airway. Moreover, the novel synthesized flavonoid LFG-500 can alleviate LPS-induced inflammatory responses by inhibiting the expression of IL-6, TNF-α and IL-1β (27).

In the present study, the effects of SB203580, an inhibitor of p38MAPK, on ALI and inflammation in LPS-induced mice were investigated. SB203580 treatment significantly reduced LPS-induced expression of TNF-α and IL-1β in lung tissues, which suggested that SB203580 inhibited ALI-stimulated inflammatory responses by inhibiting the expression of proinflammatory cytokines. Furthermore, SB203580 reduced the W/D weight ratio compared with the ALI group, indicating attenuation of LPS-induced lung edema.

The transcription factor IRF5 is involved in multiple autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and inflammatory bowel disease (28). IRF5 induces the upregulation of proinflammatory genes, repression of anti-inflammatory mediators and polarization of macrophages to a proinflammatory phenotype (2830). In the present study, the expression of IRF5 was significantly upregulated in ALI model mice compared with Ctrl mice, and significantly downregulated in SB203580-treated ALI model mice. The results suggested that SB203580 inhibited LPS-stimulated inflammatory responses by decreasing the expression of IRF5 in lung tissue.

iNOS is an enzyme expressed in macrophages and endothelial cells that synthesizes NO via L-arginase oxidation. iNOS competes with Arg1 for the same substrate and is considered a marker of inflammatory responses (31,32). In the present study, the expression of iNOS was increased in the lungs of ALI mice compared with Ctrl mice, and SB203580 treatment decreased LPS-induced iNOS expression. By contrast, the expression of Arg1 in lung tissues remained almost unchanged among the three groups. Thus, the results suggested that SB203580 attenuated the inflammatory response by decreasing the expression of iNOS in LPS-induced lung tissues.

In conclusion, the inhibitor of the p38MAPK signaling pathway SB203580 attenuated lung injury and inflammatory responses in ALI model mice. Treatment with SB203580 in LPS-induced ALI model mice reduced edema and the expression of proinflammatory cytokines, and alleviated pathological changes in lung tissues. SB203580 also decreased the expression of IRF5 and iNOS in ALI lung tissues. Thus, IRF5 may serve as a key regulatory factor of the inflammatory response in the lungs and may exert its effects by promoting a proinflammatory macrophage phenotype. Although further research is required to fully understand the mechanism of SB203580 attenuating lung edema and inflammatory response in ALI, the present study suggested a protective role of SB203580 in LPS-induced ALI; therefore, SB203580 may serve as a potential preventive agent for ALI.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Jiangsu Province (grants nos. BK20190150 and BK20160196) and the National Natural Science Foundation of China (grant no. 81500039).

Availability of data and materials

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

Authors' contributions

GL designed the study, analyzed the data and wrote the manuscript. YD performed the hematoxylin and eosin staining and immunohistochemistry assay. JT performed immunofluorescence and ELISA assays. JZo and XN analyzed the data and helped to write the paper. ZY, JZh and XY performed animal experiments and western blot assays. JC designed the present study and provided financial support for this work. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experiments in the present study were approved by The Institutional Animal Care and Use Committee at Nanjing Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ashbaugh DG, Bigelow DB, Petty TL and Levine BE: Acute respiratory distress in adults. Lancet. 2:319–323. 1967. View Article : Google Scholar : PubMed/NCBI

2 

Villar J, Blanco J and Kacmarek RM: Current incidence and outcome of the acute respiratory distress syndrome. Curr Opin Crit Care. 22:1–6. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Erickson SE, Martin GS, Davis JL, Matthay MA and Eisner MD; NIH NHLBI ARDS Network, : Recent trends in acute lung injury mortality: 1996–2005. Crit Care Med. 37:1574–1579. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Patel BV, Wilson MR and Takata M: Resolution of acute lung injury and inflammation: A translational mouse model. Eur Respir J. 39:1162–1170. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Gill SE, Yamashita CM and Veldhuizen RA: Lung remodeling associated with recovery from acute lung injury. Cell Tissue Res. 367:495–509. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Matute-Bello G, Frevert CW and Martin TR: Animal models of acute lung injury. Am J Physiol Lung Cell Mol Physiol. 295:L379–L399. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Han S and Mallampalli RK: The acute respiratory distress syndrome: From mechanism to translation. J Immunol. 194:855–860. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Robb CT, Regan KH, Dorward DA and Rossi AG: Key mechanisms governing resolution of lung inflammation. Semin Immunopathol. 38:425–448. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Freire MO and Van Dyke TE: Natural resolution of inflammation. Periodontol 2000. 63:149–164. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, Hussell T, Feldmann M and Udalova IA: IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. 12:231–238. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Lien C, Fang CM, Huso D, Livak F, Lu R and Pitha PM: Critical role of IRF-5 in regulation of B-cell differentiation. Proc Natl Acad Sci USA. 107:4664–4668. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Krausgruber T, Saliba D, Ryzhakov G, Lanfrancotti A, Blazek K and Udalova IA: IRF5 is required for late-phase TNF secretion by human dendritic cells. Blood. 115:4421–4430. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Aggarwal NR, King LS and D'Alessio FR: Diverse macrophage populations mediate acute lung inflammation and resolution. Am J Physiol Lung Cell Mol Physiol. 306:L709–L725. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Schnyder-Candrian S, Quesniaux VF, Di Padova F, Maillet I, Noulin N, Couillin I, Moser R, Erard F, Vargaftig BB, Ryffel B, et al: Dual effects of p38 MAPK on TNF-dependent bronchoconstriction and TNF-independent neutrophil recruitment in lipopolysaccharide-induced acute respiratory distress syndrome. J Immunol. 175:262–269. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Szarka RJ, Wang N, Gordon L, Nation PN and Smith RH: A murine model of pulmonary damage induced by lipopolysaccharide via intranasal instillation. J Immunol Methods. 202:49–57. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Chen H, Bai C and Wang X: The value of the lipopolysaccharide-induced acute lung injury model in respiratory medicine. Expert Rev Respir Med. 4:773–783. 2010. View Article : Google Scholar : PubMed/NCBI

17 

van Helden HP, Kuijpers WC, Steenvoorden D, Go C, Bruijnzeel PL, van Eijk M and Haagsman HP: Intratracheal aerosolization of endotoxin (LPS) in the rat: A comprehensive animal model to study adult (acute) respiratory distress syndrome. Exp Lung Res. 23:297–316. 1997. View Article : Google Scholar : PubMed/NCBI

18 

Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS and Kuebler WM; Acute Lung Injury in Animals Study Group, : An official American Thoracic Society workshop report: Features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 44:725–738. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Mehta S: The effects of nitric oxide in acute lung injury. Vascul Pharmacol. 43:390–403. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD and Gordon S: Macrophage receptors and immune recognition. Annu Rev Immunol. 23:901–944. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Gordon S and Mantovani A: Diversity and plasticity of mononuclear phagocytes. Eur J Immunol. 41:2470–2472. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Hughes KT and Beasley MB: Pulmonary manifestations of acute lung injury: More than just diffuse alveolar damage. Arch Pathol Lab Med. 141:916–922. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Butt Y, Kurdowska A and Allen TC: Acute Lung Injury: A Clinical and Molecular Review. Arch Pathol Lab Med. 140:345–350. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Elicker BM, Jones KT, Naeger DM and Frank JA: Imaging of Acute Lung Injury. Radiol Clin North Am. 54:1119–1132. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Pyee Y, Chung HJ, Choi TJ, Park HJ, Hong JY, Kim JS, Kang SS and Lee SK: Suppression of inflammatory responses by handelin, a guaianolide dimer from Chrysanthemum boreale, via downregulation of NF-κB signaling and pro-inflammatory cytokine production. J Nat Prod. 77:917–924. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Nick JA, Young SK, Brown KK, Avdi NJ, Arndt PG, Suratt BT, Janes MS, Henson PM and Worthen GS: Role of p38 mitogen-activated protein kinase in a murine model of pulmonary inflammation. J Immunol. 164:2151–2159. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Li C, Yang D, Cao X, Wang F, Jiang H, Guo H, Du L, Guo Q and Yin X: LFG-500, a newly synthesized flavonoid, attenuates lipopolysaccharide-induced acute lung injury and inflammation in mice. Biochem Pharmacol. 113:57–69. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Eames HL, Corbin AL and Udalova IA: Interferon regulatory factor 5 in human autoimmunity and murine models of autoimmune disease. Transl Res. 167:167–182. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Khoyratty TE and Udalova IA: Diverse mechanisms of IRF5 action in inflammatory responses. Int J Biochem Cell Biol. 99:38–42. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Weiss M, Blazek K, Byrne AJ, Perocheau DP and Udalova IA: IRF5 is a specific marker of inflammatory macrophages in vivo. Mediators Inflamm. 2013:2458042013. View Article : Google Scholar : PubMed/NCBI

31 

Cook HT, Jansen A, Lewis S, Largen P, O'Donnell M, Reaveley D and Cattell V: Arginine metabolism in experimental glomerulonephritis: Interaction between nitric oxide synthase and arginase. Am J Physiol. 267:F646–F653. 1994.PubMed/NCBI

32 

Li Z, Zhao ZJ, Zhu XQ, Ren QS, Nie FF, Gao JM, Gao XJ, Yang TB, Zhou WL, Shen JL, et al: Differences in iNOS and arginase expression and activity in the macrophages of rats are responsible for the resistance against T. gondii infection. PLoS One. 7:e358342012. View Article : Google Scholar : PubMed/NCBIPubMed/NCBIPubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 22 Issue 2

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
Li G, Dai Y, Tan J, Zou J, Nie X, Yang Z, Zhao J, Yang X and Chen J: SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide‑induced acute lung injury. Mol Med Rep 22: 1656-1662, 2020
APA
Li, G., Dai, Y., Tan, J., Zou, J., Nie, X., Yang, Z. ... Chen, J. (2020). SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide‑induced acute lung injury. Molecular Medicine Reports, 22, 1656-1662. https://doi.org/10.3892/mmr.2020.11214
MLA
Li, G., Dai, Y., Tan, J., Zou, J., Nie, X., Yang, Z., Zhao, J., Yang, X., Chen, J."SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide‑induced acute lung injury". Molecular Medicine Reports 22.2 (2020): 1656-1662.
Chicago
Li, G., Dai, Y., Tan, J., Zou, J., Nie, X., Yang, Z., Zhao, J., Yang, X., Chen, J."SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide‑induced acute lung injury". Molecular Medicine Reports 22, no. 2 (2020): 1656-1662. https://doi.org/10.3892/mmr.2020.11214