Anti-inflammatory effects of methanol extract of Canarium lyi C.D. Dai & Yakovlev in RAW 264.7 macrophages and a murine model of lipopolysaccharide-induced lung injury

  • Authors:
    • Ju-Mi Hong
    • Ok-Kyoung Kwon
    • In-Sik Shin
    • Chan-Mi Jeon
    • Na-Rae Shin
    • Joongku Lee
    • Sang-Hong Park
    • Tran The Bach
    • Do Van Hai
    • Sei-Ryang Oh
    • Sang-Bae Han
    • Kyung-Seop Ahn
  • View Affiliations

  • Published online on: March 2, 2015     https://doi.org/10.3892/ijmm.2015.2117
  • Pages: 1403-1410
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Canarium lyi C.D. Dai & Yakovlev (CL) is a member of the Anacardiaceae family. To the best of our knowledge, no studies on its anti-inflammatory effects have yet been reported. In the present study, we investigated the protective effects of CL on inflammation in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and LPS-induced acute lung injury (ALI) mice. CL attenuated the production of LPS-stimulated inflammatory mediators such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and interleukin-6 (IL-6). Furthermore, CL suppressed phosphorylation of the inhibitor κB-α (IκB-α), p38, c-Jun terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), as well as the translocation of the nuclear factor-κB (NF-κB) p65 subunit into the nucleus. For the in vivo efficacy, the effect of CL on a mouse model of LPS-induced acute lung injury was assessed. CL treatment of the mice significantly inhibited the inflammatory cell recruitment and pro-inflammatory cytokine production in bronchoalveolar lavage fluids (BALF). CL-treated mice also showed a marked inhibition of cyclooxygenase-2 (COX-2) and phosphorylation of IκB and p65. In addition, CL attenuated lung histopathological changes in LPS-induced ALI mice. In conclusion, our results suggest that CL is a potential therapeutic candidate for the treatment of inflammatory diseases, including pneumonia.

Introduction

Inflammation is a complex protective response caused by endo- and exogenous stimuli such as bacterial lipopolysaccharide (LPS) (1). LPS, the major component in the outer membrane of gram-negative bacteria cell walls, induces the production of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, as well as inflammatory mediators, such as nitric oxide (NO) and prostaglandin E2 (PGE2), which are synthesized by inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), respectively (2). These pro-inflammatory cytokines and inflammatory mediators assist with the innate immune response. However, their overproduction results in acute phase endotoxemia that causes tissue injury, organ failure, shock and even death (3).

Nuclear factor-κB (NF-κB), which is a key player in the regulation of immune and inflammatory responses, has an important role in modulating the transcription of several inflammatory factors and cytokines such as TNF-α, IL-1β and IL-6 (4). In unstimulated cells, Rel protein dimers, which are composed mainly of p50 and p65 subunits, are normally sequestered in the cytosol as an inactive complex by binding to inhibitor κB-α (IκB-α) (5). The activation of NF-κB mostly occurs through the phosphorylation and subsequent degradation of IκB via the activation of inhibitor κB kinase (IKK). When IκB is phosphorylated, it is targeted for ubiquitination and subsequent degradation by the 26S proteosome (6). The resulting free NF-κB then translocates to the nucleus, where it binds to κB-binding sites in the promoter regions of target genes and induces the transcription of pro-inflammatory mediators (7).

The mitogen-activated protein kinases (MAPKs) are serine/threonine-specific protein kinases that play a critical role in the regulation and differentiation of cell survival/apoptosis, as well as in controlling the cell response to cytokines, growth factors and environmental stresses (8). These classical MAPKs, extracellular signal-regulated kinase 1/2 (ERK 1/2), p38 MAPK and c-jun NH2-terminal kinase (JNK) have been involved in the transcriptional regulation of inflammatory genes (9). Specifically, p38 MAPK signaling pathways constitute an additional level of gene regulation by NF-κB, especially of the p65 subunit (10,11). Synergistically, MAPKs and NF-κB can collaborate to induce pro-inflammatory cytokine gene products and excretion (12). Accordingly, therapy aimed at the inhibition of NF-κB and MAPKs have potential therapeutic advantages in curing inflammatory diseases (13).

Canarium lyi C.D. Dai & Yakovlev (CL) is a member of the Anacardiaceae family. To the best of our knowledge, no studies on its anti-inflammatory effects have yet been reported.

In the present study, we elucidated the assumptive anti-inflammatory mechanism of CL on LPS-stimulated RAW 264.7 macrophages by measuring the levels of cytokines, NF-κB and MAPK activation. In addition, we assessed the anti-inflammatory effects of CL in a murine model of LPS-induced acute lung injury by measuring the brochoalveolar lavage fluid (BALF) analysis, pro-inflammatory protein expression and histological alteration of lung tissue.

Materials and methods

In vitro experiment
Preparation of plant material

Canarium lyi C.D. Dai & Yakovlev of the Burseraceae family was collected from the area of Gia Lai, K Bang, So Pai, Vietnam in 2011. Plant samples were identified by Dr Tran The Bach of the Institute of Ecology and Biological Resources. A voucher specimen (KRIB 0036679) was deposited in the herbarium (KRIB) of the Korea Research Institute of Bioscience and Biotechnology. Canarium lyi (147 g) was treated with MeOH and sonicated several times at room temperature for 3 days to produce an extract (20.05 g).

Cell culture

RAW 264.7 cells were maintained at 1×105 cells/ml in Dulbecco’s modified Eagle’s medium (DMEM; Sigma, St. Louis, MO, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Invitrogen, Burlington, ON, Canada), and 1% (w/v) of an antibiotic-antimycotic solution (Invitrogen, Grand Island, NY, USA) in 95% air and 5% CO2 humidified atmosphere at 37°C.

Cytotoxicity assay

Cell viability was determined by assessing the mitochondrion-dependent reduction of MTT (Amresco LLC, Solon, OH, USA) to formazan. Briefly, 5 μl of a 5 mg/ml MTT solution was added to the cell supernatant, and then incubation for 4 h at 37°C. DMSO was added following removal of the medium. The optical density of formazan was measured using a microplate reader (VersaMax; Molecular Devices, San Diego, CA, USA) at 570 nm. The level of formazan generated by untreated cells was chosen as the 100% value.

Measurement of nitric oxide

Nitrite levels in the cultured media and serum, which reflect intracellular nitric oxide (NO) synthase activity, were determined by Griess reaction. The cells were incubated with samples in the presence of LPS (0.5 μg/ml, Sigma-Aldrich, La Jolla, CA, USA) at 37°C for 24 h. The cell supernatant was dispensed into new 96-well plates, and 100 μl of each supernatant was mixed with the same volume of the Griess reagent [1% sulfanilamide, 0.1% N-(1-naphathyl)-ethylenediamine dihydrochloride and 5% phosphoric acid] and incubated at room temperature for 10 min. Sodium nitrite was used to generate a standard curve, and the concentration of nitrite was measured for absorbance at 540 nm.

Enzyme-linked immunosorbent assay (ELISA) of IL-6

The levels of IL-6 in supernatant were determined using a commercially available ELISA kit (R&D Systems Inc., Minneapolis, MN, USA) according to the manufacturer’s instructions. IL-6 levels were determined from a standard curve. The concentrations were expressed as pg/ml.

Enzyme immune assay of prostaglandin E2

PGE2 levels in supernatants were determined using a PGE2 EIA kit (Cayman Chemical Co., Inc., Ann Arbor, MI, USA) according to the manufacturer’s instructions. Briefly, 50 μl diluted standards/samples were pipetted into the wells of a 96-well plate precoated with goat polyclonal anti-mouse IgG. Aliquots of a PGE2 monoclonal antibody solution and a PGE2 acetylcholine esterase conjugate solution were added to each well, and incubated at room temperature for 18 h. The wells were washed six times with a wash buffer containing 0.05% (v/v) Tween-20, followed by the addition of 200 μl Ellman’s reagent containing acetylthiocholine and 5,5′-dithio-bis-(2-nitrobenzoic acid). PGE2 concentrations were measured by absorbance at 405 nm.

Reverse transcriptase-polymerase chain reaction analysis (RT-PCR)

Total RNA was isolated using TRIzol™ reagent (Life Technologies Corp., Carlsbad, CA, USA). For RT-PCR, a single-strand cDNA was synthesized from 2 μg total RNA. The primer sequences used were: iNOS: sense, 5′-CAA GAG TTT GAC CAG AGG ACC-3′ and antisense, 5′-TGG AAC CAC TCG TAC TTG GGA-3′; COX-2: sense, 5′-GAA GTC TTT GGT CTG GTC TCC TG-3′ and antisense, 5′-GTC TGC TGG TTT GGA ATA GTT GC-3′; TNF-α: sense and 5′-CAT CTT GCA AAA TTC GAG TGA CAA-3′ and antisense, 5′-TGG GAG TAG ACA AGG TAC AAC CC-3′; IL-6, sense, 5′-GAG GAT ACC ACT CCC AAC AGA CC-3′ and antisense, 5′-AAG TGC ATC ATC GTT GTT CAT ACA-3′, β-actin: sense, 5′-CGC TCA TTG CCG ATA GTG AT-3′ and antisense 5′-TGT TTG AGA CCT TCA ACA CC-3′. PCR products were fractionated on 1.5% agarose gel electrophoresis and stained with 5 μg/ml ethidium bromide. Images were captured by an Olympus C-4000 Zoom camera system (Olympus America Inc., Center Valley, PA, USA).

Immunoblot analysis

Western blot analyses were performed as previously described (14). Immunoblotting was performed with the primary antibodies at 4°C overnight. A horseradish peroxidase-labeled secondary antibody (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) was then used for 1 h. The membranes were washed three times with TBST, and then developed using an enhanced chemiluminescence (ECL) kit (Thermo Fisher Scientific, San Jose, CA, USA). For quantitative analysis, densitometric band values were determined using a bio-imaging analyzer (LAS 4000 mini; Fujifilm, Tokyo, Japan).

In vivo experiment
Animals

Male C57BL/6 mice (6–8 weeks) were obtained from the Koatech Co. (Pyeongtaek, Korea). The mice were fed with food and water ad libitum in an animal facility with temperature ranging from 22 to 24°C and a 12-h light/dark cycle under a specific pathogen-free conditions. Prior to the initiation of the experiment, the mice were housed for a minimum of one week in order that they adapt to the environment. The animal experimental procedures were approved by the Korea Research Institute of Bioscience and Biotechnology and performed in compliance with the National Institute of Health Guidelines for the care and use of laboratory animals and the Korean national animal welfare law.

Experimental protocols

Mice were randomly allocated into 4 groups: Control, LPS, LPS + dexamethasone (LPS + DEX) and LPS + CL. Dexamethasone served as a positive control drug. The mice of the LPS + DEX and LPS + CL groups received dexamethasone (3 mg/kg) and CL (30 mg/kg) by oral gavage for 3 days, respectively. The mice from the control and LPS groups received oral gavage at an equal volume of PBS. The mice including the LPS, LPS + DEX and LPS + CL groups were instilled with 10 μg of LPS dissolved in 50 μl PBS intranasally to induce acute lung injury 1 h after final drug treatment. Mice in the control group were intranasally given 50 μl PBS without LPS for 18 h. To obtain BALF, ice-cold PBS (0.7 ml) was infused into the lung and withdrawn via tracheal cannulation twice (total volume 1.4 ml).

Inflammatory cell counts in bronchoalveolar lavage fluid

Total inflammatory cell numbers were assessed by counting cells in at least five squares of a hemocytometer after excluding dead cells by Trypan blue staining. To determine the differential cell counts, 100 μl of BALF was centrifuged onto slides using a Cytospin (Hanil Science Industrial Co., Ltd., Seoul, Korea) (200 × g for 5 min). The slides were dried, and the cells were fixed and stained using a Diff-Quik® staining reagent (B4132-1A; IMEB Inc., Deerfield, IL, USA) following the manufacturer’s instructions. The supernatant obtained from BALF was stored at −70°C for the biochemical analysis.

Western blot analysis of the lung tissue

Lung tissue was homogenized (1/10 w/v) using a homogenizer with a tissue lysis/extraction reagent (Sigma-Aldrich) containing a protease inhibitor cocktail (Roche Diagnostics, Indianapolis, IN, USA). Each protein concentration was determined using a Bradford reagent (Bio-Rad Laboratories, Hercules, CA, USA). Western blotting was performed as described above, and the levels of COX-2, IκB-α and NF-κB were determined.

Measurement of the levels of pro-inflammatory cytokines in the BALF

The levels of IL-6 (R&D Systems), TNF-α and IL-1β (BD Biosciences, San Jose, CA, USA) in BALF were measured using ELISA kits according to the manufacturer’s instructions.

Histological analysis

After BALF samples were obtained, lung tissues were fixed in 4% (v/v) paraformaldehyde. Tissues were embedded in paraffin, sectioned at 4 μm thickness, and stained with H&E solution (Sigma-Aldrich) to estimate inflammation.

Statistical analysis

Data are expressed as the means ± the standard error of the mean (SEM). Statistical significance was determined using analyses of variance (ANOVAs) followed by multiple comparison tests with Dunnet’s adjustment. P<0.05 was considered significant.

Results

In vitro study
Effect of CL on cell viability

We determined the effect of CL on cell viability by MTT assay after incubating cells for 24 h. The cytotoxicity of CL was preliminarily evaluated to establish the appropriate concentration ranges of CL for the analysis of ongoing experiments. Results showed that the cells’ viabilities were not affected by CL at the concentrations (5, 10, 20 and 40 μg/ml) used (Fig. 1A). Therefore, we used non-toxic concentrations (5–40 μg/ml) for the entire experiment.

Effect of CL on the production of nitrite, PGE2 and pro-inflammatory cytokines on LPS-stimulated RAW 264.7 cells

LPS-stimulated cells markedly increased in NO, whereas CL treatment significantly decreased NO production in a concentration-dependent manner (Fig. 1B). We also examined the effects of CL on PGE2 production following LPS stimulation in RAW 264.7 cells. The amount of PGE2 was increased by the LPS stimulation in the culture supernatant, and this increase was effectively reduced by treatment with CL (Fig. 1C).

Similarly, treatment of the RAW 264.7 cells with LPS alone resulted in a significant increase in cytokine production compared with the control group (Fig. 1D). However, CL treatment considerably inhibited the LPS induction of IL-6 in a dose-dependent manner (Fig. 1D).

Effect of CL on mRNA and protein expression levels of inflammatory mediators

The production of mRNA and protein of iNOS, COX-2 and pro-inflammatory cytokines, including IL-6 and TNF-α, increased in LPS-stimulated RAW 264.7 cells (Fig. 2). However, treatment of the cells with CL significantly decreased iNOS, COX-2 and pro-inflammatory cytokine production compared to the LPS-stimulated cells in a concentration-dependent manner (Fig. 2).

Effect of CL on LPS-induced MAPK activation

LPS-stimulated cells showed an increase in the phosphorylation levels of p38, ERK1/2 and JNK. By contrast, treatment of the cells with CL significantly reduced the phosphorylation of p38, ERK and JNK expression compared with the LPS-stimulated cells in a concentration-dependent manner (Fig. 3).

Effect of CL on LPS-induced NF-κB activity

We examined the effect of CL on LPS-stimulated IκB-α degradation. One hour of pre-treatment with CL followed by treatment with LPS for 30 min markedly suppressed the LPS-stimulated phosphorylation and degradation of IκB-α in a dose-dependent manner (Fig. 4A). We also investigated the translocation of the NF-κB subunit p65 from the cytosol to the nucleus using western blot analysis. LPS stimulation caused p65 translocation from the cytosol to the nucleus, while CL inhibited this translocation (Fig. 4B).

In vivo experiment
Effect of CL on inflammatory cell count in the BALF of LPS-induced ALI mice

The LPS group showed a significant increase in the number of total inflammatory cells and neutrophils in BALF compared with the negative control group. However, pre-treatment with CL significantly decreased the number of total inflammatory cells and neutrophils compared with the LPS group (Fig. 5).

Effect of CL on pro-inflammatory cytokines in the BALF of LPS-induced ALI mice

The levels of IL-6, IL-1β and TNF-α in BALF were significantly higher in LPS-induced mice when compared with the negative control group. By contrast, the CL-treated mice had considerably lower levels of all these pro-inflammatory cytokines compared to the LPS-treated mice (Fig. 6).

Effect of CL on COX-2 and NF-κB in the lung tissue of LPS-induced ALI mice

The LPS-induced mice exhibited an increased expression of COX-2, p-IκB and p-p65 in the lung tissue compared to the negative control group. However, treatment of the mice with CL significantly inhibited the expression of COX-2, IκB degradation and the phosphorylation of p65 in the lung tissue compared to the LPS-treated mice (Fig. 7).

Effect of CL on histopathological changes in the lung tissue of LPS-induced ALI mice

We confirmed an intact structure and clear pulmonary alveoli in the lung sections of mice in the negative control group. By contrast, lung sections obtained from mice in the LPS group showed evidence of histological changes, including areas of inflammatory cell infiltration, thickening of the alveolar wall, edema and pulmonary congestion. By contrast, treatment of the mice with CL attenuated the pathological changes that were observed in the LPS-treated mice (Fig. 8).

Discussion

Macrophages are major inflammatory cells and immune effector cells (15). The activity of macrophages plays an important role in the inflammatory responses when infected with pathogens such as LPS.

Macrophages can kill pathogens directly by phagocytosis and indirectly through the secretion of various pro-inflammatory mediators such as reactive oxygen and pro-inflammatory cytokines such as TNF-α, IL-1β and IL-6 (16). The overproduction of the inflammatory mediators by activated macrophages is involved in the pathophysiology of many inflammatory diseases, including arthritis, acute lung injury, chronic obstructive pulmonary disease (COPD), asthma and inflammatory bowel disease (IBD) (17). LPS can directly activate macrophages and trigger the production of inflammatory mediators, such as pro-inflammatory cytokines, NO and PGE2, which are the main cytotoxic and pro-apoptotic mechanisms participating in the innate response of many mammals. Thus, LPS-stimulated RAW 264.7 macrophages can be effectively used as a model to study inflammation and potential anti-inflammatory mediators with their action mechanisms (18). The LPS stimulation of murine macrophages has been known to induce the phosphorylation and activation of MAPKs such as ERK 1/2, JNK, and p38 (19,20). Previous studies have demonstrated that LPS stimulation accelerates the phosphorylation of MAPKs in an inflammatory response. Moreover, it has been reported that the activation of NF-κB is triggered by MAPK (21). In the present study, we found that CL inhibited the expression of iNOS, COX-2, IL-6 and TNF-α mRNA simultaneously and concentration-dependently, suggesting that the inhibition of NO and PGE2 release may be caused by the suppression of iNOS and COX-2 expression at the mRNA level. In addition, we investigated the effects of CL on the LPS-stimulated phosphorylation of MAPKs in RAW 264.7 cells. The results showed that CL significantly reduced the phosphorylation of MAPKs in the LPS-stimulated RAW 264.7 cells compared with LPS-stimulated RAW 264.7 cells not treated with CL. Therefore, these results indicate that CL may attenuate the inflammatory responses induced by LPS stimulation.

NF-κB plays a central role in immunity since it activates the pro-inflammatory genes that encode iNOS, COX-2 and TNF-α (22). NF-κB is a transcription factor and binds to the κB motifs in the promoters of target genes, and thus, induces the transcriptions of iNOS, COX-2 and TNF-α. In unstimulated cells, Rel protein dimers, which are composed mainly of p50 and p65 subunits, are sequestered in the cytoplasm as complexes with a family of inhibitors known as IκB (23). When the IκBs become phosphorylated, NF-κB is released from its inhibition by IκB and translocated to the nucleus where inflammation-associated genes are then activated. NF-κB activation mediates the transactivation of pro-inflammatory genes, including TNF-α and IL-6 (24,25). To elucidate the effect of CL on NF-κB activation, we evaluated the expression of phosphorylated IκB-α in vitro and in vivo. The results of the present study demonstrate that CL blocked LPS-induced IκB-α phosphorylation and, as a result, inhibited NF-κB activation. Since LPS induces changes in the NF-κB and MAPK signaling pathways, MAPKs may be another factor affected by CL exposure. These collective results provide convincing evidence that CL had an anti-inflammatory ability by inhibiting LPS-stimulated NF-κB and MAPK activation and subsequent cytokine production in RAW 264.7 cells.

To investigate the potential anti-inflammatory effects of CL in vivo, we evaluated the protective effects of CL on a murine model of LPS-induced ALI. ALI is characterized by interstitial edema, neutrophil accumulation, epithelial integrity disruption, and protein leakage into the alveolar space, severely altering gas exchange (26). Many sequela associated with ALI result from the excessive production of cytokine mediators such as TNF-α, IL-1β and IL-6 (27). Previous studies have shown that increased levels of TNF-α, IL-1β and IL-6 in BALF may be noted in the persistent elevation of pro-inflammatory cytokines in patients with ALI (28). TNF-α, IL-1β and IL-6 increase the inflammatory cascade, cause inflammatory injury and recruit neutrophils into the lung (29). In our results, CL markedly decreased the production of TNF-α, IL-1β and IL-6, as well as the infiltration of inflammation cells, including macrophages and neutrophils compared with LPS-induced ALI. These findings were consistent with the histopathology of lung tissue. These results indicate that the protective effects of CL on ALI induced by LPS may result from the inhibition of inflammatory mediators and the limitation of an inflammatory response in the lung.

In summary, the results of the present study have demonstrated the anti-inflammatory effects of CL in in vitro and in vivo experiments. CL inhibited the expression of pro-inflammatory mediators in LPS-stimulated RAW 264.7 cells and LPS-induced ALI mice, and it blocked the activation of MAPKs and NF-κB. These results suggest that CL may have therapeutic potential for effectively treating inflammatory diseases such as pneumonia.

Acknowledgments

The present study was supported by a grant from the Ministry of Science, ICT and Future planning (FGC 1011332) and the KRIBB Research Initiative Program (KGM1221413) of the Republic of Korea.

References

1 

Kim JJ, Jiang J, Shim DW, et al: Anti-inflammatory and anti-allergic effects of Agrimonia pilosa Ledeb extract on murine cell lines and OVA-induced airway inflammation. J Ethnopharmacol. 140:213–221. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Huang MH, Wang BS, Chiu CS, et al: Antioxidant, antinociceptive, and anti-inflammatory activities of Xanthii Fructus extract. J Ethnopharmacol. 135:545–552. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Hong YH, Chao WW, Chen ML, et al: Ethyl acetate extracts of alfalfa (Medicago sativa L.) sprouts inhibit lipopolysaccharide-induced inflammation in vitro and in vivo. J Biomed Sci 14. 16:642009. View Article : Google Scholar

4 

Su YW, Chiou WF, Chao SH, et al: Ligustilide prevents LPS-induced iNOS expression in RAW 264.7 macrophages by preventing ROS production and down-regulating the MAPK, NF-κB and AP-1 signaling pathways. Int Immunopharmacol. 11:1166–1172. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Oh JH, Kang LL, Ban JO, et al: Anti-inflammatory effect of 4-O-methylhonokiol, compound isolated from Magnolia officinalis through inhibition of NF-kappaB. Chem Biol Interact. 180:506–514. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Chen ZJ: Ubiquitin signalling in the NF-kappaB pathway. Nat Cell Biol. 7:758–765. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Park HH, Kim MJ, Li Y, et al: Britanin suppresses LPS-induced nitric oxide, PGE2 and cytokine production via NF-κB and MAPK inactivation in RAW 264.7 cells. Int Immunopharmacol. 15:296–302. 2013. View Article : Google Scholar

8 

Gao Y, Jiang W, Dong C, et al: Anti-inflammatory effects of sophocarpine in LPS-induced RAW 264.7 cells via NF-κB and MAPKs signaling pathways. Toxicol In Vitro. 26:1–6. 2012. View Article : Google Scholar

9 

Dong C, Davis RJ and Flavell RA: MAP kinases in the immune response. Annu Rev Immunol. 20:55–72. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Kim JY, Shin JS, Ryu JH, et al: Anti-inflammatory effect of anemarsaponin B isolated from the rhizomes of Anemarrhena asphodeloides in LPS-induced RAW 264.7 macrophages is mediated by negative regulation of the nuclear factor-κB and p38 pathways. Food Chem Toxicol. 47:1610–1617. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Saccani S, Pantano S and Natoli G: p38-dependent marking of inflammatory genes for increased NF-kappaB recruitment. Nat Immunol. 3:69–75. 2002. View Article : Google Scholar

12 

Craig R, Larkin A, Mingo AM, et al: p38 MAPK and NF-kappa B collaborate to induce interleukin-6 gene expression and release. Evidence for a cytoprotective autocrine signaling pathway in a cardiac myocyte model system. J Biol Chem. 275:23814–23824. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Huo M, Cui X, Xue J, et al: Anti-inflammatory effects of linalool in RAW 264.7 macrophages and lipopolysaccharide-induced lung injury model. J Surg Res. 180:e47–e54. 2013. View Article : Google Scholar

14 

Su YW, Chao SH, Lee MH, et al: Inhibitory effects of citronellol and geraniol on nitric oxide and prostaglandin E2 production in macrophages. Planta Med. 76:1666–1671. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Kim AR, Lee MS, Shin TS, et al: Phlorofucofuroeckol A inhibits the LPS-stimulated iNOS and COX-2 expressions in macrophages via inhibition of NF-κB, Akt, and p38 MAPK. Toxicol In Vitro. 25:1789–1795. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Himaya SW, Ryu B, Qian ZJ, et al: Sea cucumber, Stichopus japonicus ethyl acetate fraction modulates the lipopolysaccharide induced iNOS and COX-2 via MAPK signaling pathway in murine macrophages. Environ Toxicol Pharmacol. 30:68–75. 2010. View Article : Google Scholar

17 

Vodovotz Y, Chow CC, Bartels J, et al: In silico models of acute inflammation in animals. Shock. 26:235–244. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Huang SS, Chiu CS, Lin TH, et al: Antioxidant and anti-inflammatory activities of aqueous extract of Centipeda minima. J Ethnopharmacol. 147:395–405. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Fengyang L, Yunhe F, Bo L, et al: Stevioside suppressed inflammatory cytokine secretion by downregulation of NF-κB and MAPK signaling pathways in LPS-stimulated RAW264.7 cells. Inflammation. 35:1669–1675. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Tuntipopipat S, Muangnoi C, Chingsuwanrote P, et al: Anti-inflammatory activities of red curry paste extract on lipopolysaccharide-activated murine macrophage cell line. Nutrition. 27:479–487. 2011. View Article : Google Scholar

21 

Cho EJ, An HJ, Shin JS, et al: Roxatidine suppresses inflammatory responses via inhibition of NF-κB and p38 MAPK activation in LPS-induced RAW 264.7 macrophages. J Cell Biochem. 112:3648–3659. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Yun KJ, Shin JS, Choi JH, et al: Quaternary alkaloid, pseudocoptisine isolated from tubers of Corydalis turtschaninovi inhibits LPS-induced nitric oxide, PGE2, and pro-inflammatory cytokines production via the down-regulation of NF-kappaB in RAW 264.7 murine macrophage cells. Int Immunopharmacol. 9:1323–1331. 2009. View Article : Google Scholar : PubMed/NCBI

23 

An HJ, Kim IT, Park HJ, et al: Tormentic acid, a triterpenoid saponin, isolated from Rosa rugosa, inhibited LPS-induced iNOS, COX-2, and TNF-α expression through inactivation of the nuclear factor-κb pathway in RAW 264.7 macrophages. Int Immunopharmacol. 11:504–510. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Liu H, Sidiropoulos P, Song G, et al: TNF-alpha gene expression in macrophages: regulation by NF-kappa B is independent of c-Jun or C/EBP beta. J Immunol. 164:4277–4285. 2000. View Article : Google Scholar

25 

Yoshimura A: Signal transduction of inflammatory cytokines and tumor development. Cancer Sci. 97:439–447. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Y, Du Z, Zhou Q, et al: Remifentanil attenuates lipopolysaccharide-induced acute lung injury by downregulating the NF-κB signaling pathway. Inflammation. Apr 20–2014.Epub ahead of print. View Article : Google Scholar

27 

Ward PA: Role of complement, chemokines, and regulatory cytokines in acute lung injury. Ann NY Acad Sci. 796:104–112. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Minamino T and Komuro I: Regeneration of the endothelium as a novel therapeutic strategy for acute lung injury. J Clin Invest. 116:2316–2319. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Matthay MA and Zimmerman GA: Acute lung injury and the acute respiratory distress syndrome: four decades of inquiry into pathogenesis and rational management. Am J Respir Cell Mol Biol. 33:319–327. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 35 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hong J, Kwon O, Shin I, Jeon C, Shin N, Lee J, Park S, Bach TT, Hai DV, Oh S, Oh S, et al: Anti-inflammatory effects of methanol extract of Canarium lyi C.D. Dai & Yakovlev in RAW 264.7 macrophages and a murine model of lipopolysaccharide-induced lung injury. Int J Mol Med 35: 1403-1410, 2015
APA
Hong, J., Kwon, O., Shin, I., Jeon, C., Shin, N., Lee, J. ... Ahn, K. (2015). Anti-inflammatory effects of methanol extract of Canarium lyi C.D. Dai & Yakovlev in RAW 264.7 macrophages and a murine model of lipopolysaccharide-induced lung injury. International Journal of Molecular Medicine, 35, 1403-1410. https://doi.org/10.3892/ijmm.2015.2117
MLA
Hong, J., Kwon, O., Shin, I., Jeon, C., Shin, N., Lee, J., Park, S., Bach, T. T., Hai, D. V., Oh, S., Han, S., Ahn, K."Anti-inflammatory effects of methanol extract of Canarium lyi C.D. Dai & Yakovlev in RAW 264.7 macrophages and a murine model of lipopolysaccharide-induced lung injury". International Journal of Molecular Medicine 35.5 (2015): 1403-1410.
Chicago
Hong, J., Kwon, O., Shin, I., Jeon, C., Shin, N., Lee, J., Park, S., Bach, T. T., Hai, D. V., Oh, S., Han, S., Ahn, K."Anti-inflammatory effects of methanol extract of Canarium lyi C.D. Dai & Yakovlev in RAW 264.7 macrophages and a murine model of lipopolysaccharide-induced lung injury". International Journal of Molecular Medicine 35, no. 5 (2015): 1403-1410. https://doi.org/10.3892/ijmm.2015.2117