Subscribe to RSS
DOI: 10.1055/s-2006-947201
© Georg Thieme Verlag KG Stuttgart · New York
Gigantol Isolated from the Whole Plants of Cymbidium goeringii Inhibits the LPS-Induced iNOS and COX-2 Expression via NF-κB Inactivation in RAW 264.7 Macrophages Cells
Kyung-Tae Lee, Ph. D.
Department of Pharmaceutical Biochemistry
College of Pharmacy
Kyung-Hee University
Dongdaemun-Ku
Hoegi-Dong 130-701
Seoul
Korea
Phone: +82-2-961-0860
Fax: +82-2-966-3885
Email: ktlee@khu.ac.kr
Publication History
Received: April 19, 2006
Accepted: June 20, 2006
Publication Date:
21 August 2006 (online)
Abstract
During our efforts to find bioactive natural products with anti-inflammatory activity, we isolated gigantol from the whole plants of Cymbidium goeringii (Orchidaceae) by activity-guided chromatographic fractionation. Gigantol was found to have potent inhibitory effects on LPS-induced nitric oxide (NO) and prostaglandin E2 (PGE2) production in RAW 264.7 cells. Consistent with these findings, gigantol suppressed the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) at the protein and mRNA levels in RAW 264.7 cells in a concentration-dependent manner. Our data also indicate that gigantol is a potent inhibitor of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) release and influenced the mRNA expression levels of these cytokines in a dose-dependent manner. Furthermore, a reporter gene assay for nuclear factor kappa B (NF-κB) and an electromobility shift assay (EMSA) demonstrated that gigantol effectively inhibited the activation of NF-κB, which is necessary for the expression of iNOS, COX-2, TNF-α, IL-1β and IL-6. Thus, our studies suggest that gigantol inhibits LPS-induced iNOS and COX-2 expression by blocking NF-κB activation.
Key words
Cymbidium goeringii - Orchidaceae - gigantol - inducible nitric oxide synthase - cyclooxygenase-2 - NF-κB
Introduction
During inflammatory processes, large amounts of the pro-inflammatory mediators nitric oxide (NO) and prostaglandin E2 (PGE2) are generated by the inducible isoforms of NO synthase (iNOS) and by cyclooxygenase-2 (COX-2) [1]. In mammalian cells, NO is synthesized by three different isoforms of nitric oxide synthase (NOS), namely, neuronal NOS (nNOS), endothelial NOS (eNOS), and inducible NOS (iNOS). Although nNOS and eNOS are constitutively expressed, iNOS is expressed in response to interferon-γ, lipopolysaccharide (LPS), and a variety of pro-inflammatory cytokines [1]. Cyclooxygenase (COX) converts arachidonic acid to PGs, and has two isoforms, i. e., COX-1 and COX-2 [2]. COX-1 is expressed constitutively in most tissues and appears to be responsible for maintaining normal physiological functions, whereas COX-2 is only detectable in certain types of tissues and is induced transiently by growth factors, pro-inflammatory cytokines, tumor promoters, and bacterial toxins [3].
LPS is a major inflammatory molecule, and triggers the productions of pro-inflammatory toxins and cytokines, like TNF-α, in various cell types [4]. Moreover, TNF-α plays key roles in the induction and perpetuation of inflammation caused by autoimmune reactions by activating T cells and macrophages and by up-regulating other pro-inflammatory cytokines and endothelial adhesion molecules, such as, intercellular adhesion molecule 1 and vascular cell adhesion molecule 1, which enhance the recruitment of leukocytes to sites of inflammation [5]. Likewise, IL-1β is one of the most important inflammatory cytokines secreted by macrophages and is induced by LPS in macrophages. During inflammation, increased IL-1β release leads to cell or tissue damage [6], and thus, reduction of IL-1β release from macrophages may retard the inflammatory response to LPS stimulation. Moreover, in terms of response to noxious stimuli, the signal transduction pathway of IκB/NF-κB activation contributes to the regulation of IL-1β [7]. Additionally, the production of IL-6 is induced by several other factors, such as TNF-α and IL-1β as well as the bacterial endotoxin, LPS. IL-6 is a pro-inflammatory cytokine that acts as an endogenous pyrogen and has multiple effects on the immune system, particularly on hematopoiesis [7]. It is also regulated by the signal transduction pathway of IκB/NF-κB activation [8].
Nuclear transcription factor kappa-B (NF-κB) is one of the most ubiquitous transcription factors and regulates genes involved in cellular proliferation, inflammatory responses, and cell adhesion. The activation of NF-κB has been reported to induce the transcriptions of many pro-inflammatory mediators, e. g., iNOS, COX-2, TNF-α and IL-1β, -6 and -8 [9]. Functionally active NF-κB exists mainly as a heterodimer and is composed of subunits of the Rel family, i. e., p50 and p65, and is normally sequestered in the cytosol as an inactive complex due to its binding with inhibitory κB (IκBs) in unstimulated cells [10]. The activation of NF-κB involves the phosphorylation of IκBs at two critical serine residues (Ser32, Ser36) via the IκB kinase (IKK) signalosome complex [10].
Although Cymbidium goeringii (Orchidaceae) has been used as a traditional treatment for burns, frostbite, and bleeding [11], no phytochemical or pharmacological studies have been undertaken to date, with the exception of a single study on the isolation of sterols from this plant. [12]. However, no report has been issued on its anti-inflammatory activity or on its mode of action. Thus, as a part of our on-going screening program to evaluate the anti-inflammatory potential of natural compounds, we investigated the in vitro inhibition of NO production by the methanol extract of whole C. goeringii using activity-guided fractionation, which led to the isolation of gigantol (Fig. [1]). We then evaluated the effects of gigantol on NO induction by LPS and on the LPS-induced productions of PGE2, TNF-α, IL-1β, IL-6, and of other pro-inflammatory proteins and on their mRNA expressions in vitro.

Fig. 1 Chemical structure of gigantol.
Materials and Methods
#Isolation of gigantol from Cymbidium goeringii
Cymbidium goeringii was collected on Mt. Baekyang, Jeongeup, Chonbuk province, Korea during April 2003. The plant was identified by Dr. Sung Hoon Bang at the Department of Horticulture, Kyung Hee University and a voucher specimen (03 057) has been deposited at the Laboratory of Natural Product Chemistry, Kyung Hee University. Fresh whole plants (10 kg) were cut and extracted three times with 80 % MeOH (3 × 18 L). Extract solutions were filtered and dried using a rotatory evaporator under reduced pressure to give the MeOH extract (344 g), which (300 g) was suspended in H2O (2 L) and partitioned with EtOAc (2 L × 2). The EtOAc soluble portion obtained was concentrated under vacuum to yield the EtOAc extract (21 g). The aqueous layer was then successively partitioned with n-BuOH (2 L × 2), and these n-BuOH extracts were also evaporated under vacuum to give the n-BuOH extract (23 g) and the aqueous extract (256 g), respectively. The EtOAc extract (20 g) was found to be active by activity-guided fractionation and was chromatographed on a silica gel column (600 g, 7.0 × 60.0 cm; Merck; Darmstadt, Germany) using n-hexane-EtOAc (7 : 1 → 5 : 1 → 3 : 1 → 1 : 1) and CHCl3-MeOH (17 : 1 → 10 : 1), to produce twenty-eight fractions (SOE-1 - SOE-28). SOE-8 (1.6 g) was subjected to a silica gel column chromatography (100 g, 5 × 17 cm) using CHCl3-MeOH (5 : 1 → 3 : 1) as eluent to afford eleven fractions (SOE-8-1 - SOE-8-11). SOE-8-6 (152 mg) was purified by ODS column chromatography (70 g, 3 × 20 cm; Merck) and eluted with MeOH-H2O (3 : 1) to give gigantol (72 mg). The structure of this compound was confirmed by comparing its spectral data (IR, 1H-NMR, 13C-NMR, EI-MS) with those reported in the literature [13], [14]. Gigantol used for this study was checked by HPLC and was >98 % pure.
Gigantol: Colorless needles, m. p. 95 - 96 °C, IR (KBr window): νmax = 3510, 3010, 2960, 1610, 1520, 1470 cm-1; 1H-NMR (CDCl3, 400 MHz): δ = 6.82 (1H, d, J = 8.2 Hz, H-5′′), 6.77 (1H, dd, J = 8.2, 1.9 Hz, H-6′′), 6.65 (1H, d, J = 1.9 Hz, H-2′′), 6.28 - 6.29 (3H, br. s, H-2′,4′,6′), 3.85 (3H, s, H-OMe), 3.77 (3H, s, H-OMe), 2.83 - 2.85 (4H, m, H-1,2); 13C-NMR (CDCl3, 100 MHz): δ = 160.8 (C-5′), 156.5 (C-3′), 146.0 (C-3′′), 144.4 (C-1′), 143.6 (C-4′′), 133.6 (C-1′′), 121.0 (C-6′′), 114.1 (C-2′′), 111.2 (C-5′′), 108.0 (C-2′), 106.7 (C-6′), 99.1 (C-4′), 55.8 (C-OMe), 55.1 (C-OMe), 38.0 (C-2), 37.0 (C-1); EI/MS: m/z = 274 [M]+, 137.
#Chemicals and antibodies
Dulbecco’s modified Eagle’s minimum essential medium (DMEM), fetal bovine serum (FBS), penicillin, and streptomycin were obtained from Life Technologies Inc. (Grand Island, NY, USA). iNOS, COX-1, COX-2, and β-actin monoclonal antibodies and peroxidase-conjugated secondary antibody were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Enzyme immunoassay (EIA) kits for PGE2, TNF-α interleukin-1β (IL-1β) and IL-6 were obtained from R&D Systems (Minneapolis, MN, USA), and luciferase assay kits from Promega (Madison, WI, USA). pNF-κB-Luc reporter plasmid was purchased from BD Biosciences (San Jose, CA, USA). Superfect transfection reagent from Qiagen (Qiagen GmbH; Hilden, Germany), and RNA extraction kits from Intron Biotechnology (Seoul, Korea). iNOS, COX-2, TNF-α, IL-1β, IL-6 and β-actin oligonucleotide primers were from Bioneer (Seoul, Korea). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltertazolium bromide (MTT), aprotinin, leupeptin, phenylmethylsulfonyl fluoride (PMSF), dithiothreitol, Escherichia coli lipopolysaccharide (LPS), acetylsalicylic acid (aspirin), carrageenan, serotonin, indomethacin and all other chemicals were from Sigma (St. Louis, MO, USA).
#Cell culture and sample treatment
The RAW 264.7 murine macrophage cell line was obtained from the Korean Cell Line Bank (Seoul, Korea). Cells were grown at 37 °C in DMEM supplemented with 10 % FBS, penicillin (100 units/mL), and streptomycin sulfate (100 μg/mL) in a humidified 5 % CO2 atmosphere. Cells were incubated with test compounds at different concentrations and stimulated with LPS 1 μg/mL for the indicated times. Gigantol dissolved in DMSO was added to the medium in serial dilution (the final DMSO concentration in all assays did not exceed 0.1 %).
#Nitrite, PGE2, TNF-α, IL-1β and IL-6 assays
Nitrite accumulation, an indicator of NO synthesis, was measured in culture media using the Griess reaction [15]. PGE2, TNF-α, IL-1β and IL-6 levels in macrophage culture media were quantified using EIA kits, according to the manufacturer’s instructions (R&D Systems).
#Western blot analysis
Cellular proteins were extracted from control and gigantol treated RAW 264.7 cells, and washed cell pellets were resuspended in extraction lysis buffer (50 mM HEPES (pH 7.0), 250 mM NaCl, 5 mM EDTA, 0.1 % Nonidet P-40, 1 mM phenylmethylsulfonyl fluoride, 0.5 mM dithiothreitol, 5 mM sodium fluoride (NaF), 0.5 mM sodium orthovanadate) containing 5 μg/mL of each of leupeptin and aprotinin, and then incubated for 30 min at 4 °C. Cell debris was removed by microcentrifugation, and supernatants were quick frozen. Protein concentrations were determined using Bio-Rad protein assay reagent according to the manufacturer’s instruction. Forty micrograms of cellular proteins from treated and untreated cell extracts were electroblotted onto nitrocellulose membranes after 10 % SDS-polyacrylamide gel electrophoresis. Immunoblots were incubated overnight with blocking solution (5 % skim milk) at 4 °C, and then incubated for 4 h with a 1 : 1000 dilution of monoclonal anti-iNOS, a 1 : 1000 dilution of anti-COX-2 antibody, and a β-actin antibody (Santa Cruz Biotechnology Inc.). Blots were washed twice with Tween 20/Tris-buffered saline (TTBS) and then incubated with a 1 : 1000 dilution of horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology Inc.) for 1 h at room temperature. Blots were again washed three times with TTBS and then developed using enhanced chemiluminescence kits (Amersham Life Science; Arlington Heights, IL, USA).
#RNA preparation and RT-PCR
Total cellular RNA was isolated using Easy Blue® kits (Intron Biotechnology). RNA (1 μg) was reverse-transcribed (RT) from each sample using MuLV reverse transcriptase, 1 mM dNTP, and oligo (dT12 - 18) 0.5 μg/μL. PCR analyses were performed on aliquots of the cDNA preparations to detect iNOS, COX-2, TNF-α, IL-1β and IL-6 (using β-actin as the internal standard) gene expression using a thermal cycler (Perkin Elmer Cetus; Foster City, CA, USA). Reactions were carried out in 25μL containing (final concentrations) 1 unit of Taq DNA polymerase, 0.2 mM dNTP, × 10 reaction buffer, and 100 pmol of 5′ and 3′ primers. After an initial denaturation for 2 min at 95 °C, 30 amplification cycles were performed for iNOS (1 min at 95 °C, annealing 1 min at 60 °C, and extension 1.5 min at 72 °C), COX-2 (denaturation 1 min at 94 °C, annealing 1 min at 60 °C, and extension 1 min at 72 °C), TNF-α (denaturation1 min at 95 °C, annealing 1 min at 55 °C, extension and 1 min at 72 °C), IL-1β (denaturation 1 min at 94 °C, annealing 1 min at 60 °C, and extension 1 min at 72 °C) and IL-6 (denaturation 1 min at 94 °C, annealing 1 min at 57 °C, and extension 1 min at 72 °C). The PCR primers used in this study are listed below and were purchased from Bioneer (Seoul, Korea): sense strand iNOS, 5′-AATGGCAACATCAGGTCGGCCATCACT-3′, anti-sense strand iNOS, 5′-GCTGTGTGTCACAGAAGTCTCGAACTC-3′; sense strand COX-2, 5′-GGAGAGACTATCAAGATAGT-3′, anti-sense strand COX-2, 5′-ATGGTCAGTAGACTTTTACA-3′; sense strand TNF-α, 5′-ATGAGCACAGAAAGCATGATC-3′, anti-sense strand TNF-α, 5′-TACAGGCTTGTCACTCGAATT-3′; sense strand IL-1β, 5′-TGCAGAGTTCCCCAACTGGTACATC-3′; anti-sense strand IL-1β, 5′-GTGCTGCCTAATGTCCCCTTGAATC-3′; sense strand IL-6, 5′- GAGGATACCACTCCCAACAGACC-3′; anti-sense strand IL-6, 5′- AAGTGCATCATCGTTGTTCATACA-3′; sense strand β-actin, 5′-TCATGAAGTGTGACGTTGACATCCGT-3′, anti-sense strand β-actin, 5′-CCTAGAAGCATTTGCGGTGCACGATG-3′. After amplification, PCR reaction products were electrophoresed on 2 % agarose gels and visualized using ethidium bromide and UV [15].
#Transient transfection and luciferase assay (reporter gene assay)
RAW 264.7 cells were transfected using Superfect reagent (Qiagen GmbH) and pNF-κB-Luc reporter plasmid (BD Biosciences; San Jose, CA, USA), as instructed by the manufacturer. Cells were incubated for 2 h before adding 5 mL of DMEM/10 % FBS. Forty-eight hours after the start of transfection, cells were pretreated with gigantol for 1 h and stimulated with LPS (1 μg/mL). Following 3 h of stimulation, cells were lysed and luciferase activities were determined using the Promega luciferase assay system (Promega) and a luminometer (Perkin Elmer Cetus). Luciferase activities were normalized versus sample protein concentrations.
#Nuclear extraction and electrophoretic mobility shift assay (EMSA)
RAW 264.7 macrophages in 100-mm dishes (1 × 106 cells/mL) were preincubated with various concentrations of gigantol (25, 50, 100 μg/mL) and then stimulated with LPS (1 μg/mL) for 1 h. The cells were washed once with PBS, scraped into 1 mL of cold PBS, and pelleted by centrifugation. Nuclear extracts were prepared as described previously with slight modification [15]. Briefly, cell pellets were resuspended in hypotonic buffer (10 mM HEPES, pH 7.9, 1.5 mM MgCl2, 10 mM KCl, 0.2 mM PMSF, 0.5 mM DTT, 10 μg/mL aprotinin) and incubated on ice for 15 min. They were then lysed by adding 0.1 % Nonidet P-40 and vortexing vigorously for 10 s. Nuclei were pelleted by centrifugation at 12,000 × g for 10 min at 4 °C and resuspended in high salt buffer (20 mM HEPES, pH 7.9, 25 % glycerol, 400 mM KCl, 1.5 mM MgCl2, 0.2 mM EDTA, 0.5 mM DTT, 1 mM NaF, 1 mM sodium orthovanadate). Nuclear extract (10 μg) was mixed with double-stranded NF-κB oligonucleotide. 5′-AGTTGAGGGGACTTTCCCAGGC-3′ end-labeled by [γ-32P] dATP (the underline indicates a κB consensus sequence or a binding site for the NF-κB/cRel homodimeric or heterodimeric complex). Binding reactions were performed at 37 °C for 30 min in 30 μL of reaction buffer containing 10 mM Tris-HCl, pH 7.5, 100 mM NaCl, 1 mM EDTA, 4 % glycerol, 1 μg of poly(dI-dC), and 1 mM DTT. DNA-protein complexes were separated from the unbound DNA probe on native 5 % polyacrylamide gels at 100 V in 0.5 × TBE buffer. Gels were vacuum dried for 1 h at 80 °C and exposed to X-ray film at -70 °C for 24 h.
#Statistical analysis
Results are expressed as mean ± S.D. of at least three experiments performed using different cell preparations in vitro. Statistically significant values were compared using a non-parametric multiple comparisons test (Kruskal-Wallis test) followed by Dunn’s test. Statistical significance was set at P < 0.05.
#Results
Since it was recently suggested that inflammatory diseases are primarily associated with excessive NO formation [16], we undertook to identify the active principle of the MeOH extract of C. goeringii by focusing on its inhibitory effect on NO formation. The MeOH extract of C. goeringii was fractionated and aliquots were subjected to nitrite assays using RAW 264.7 macrophage cells. The MeOH extract has an inhibitory effect on NO formation in LPS-induced macrophage cells (data not shown), and this extract was further fractionated into EtOAc and BuOH extracts. The EtOAc fraction showed higher activity than the BuOH extract, and silica gel column chromatography and successive ODS column chromatography of this extract afforded gigantol (Fig. [1]). Our physical and spectroscopic data were in accord with those published for gigantol [13], [14].
To assess the effect of gigantol on LPS-induced NO production in RAW 264.7 cells, cells pretreated with/without gigantol for 1 h were treated with LPS (1 μg/mL) for 24 h; controls contained neither LPS nor sample. Cell culture media were harvested and NO concentrations were measured using the Griess reaction. Gigantol was found to inhibit LPS-induced NO production in a concentration-dependent (25, 50, 100 μg/mL) manner with an average IC50 of 42.1 μg/mL for three separate experiments (Fig. [2] A). l-N 6-(1-Iminoethyl)lysine (NIL) was used as a positive control. To examine whether gigantol inhibits PGE2 production, cells were pre-incubated with gigantol for 1 h, and then activated with 1 μg/mL LPS for 24 h. As shown in Fig. [2] B, PGE2 production was significantly inhibited by gigantol in a concentration-dependent manner. The cytotoxicity of gigantol was evaluated in the presence or absence of LPS by MTT assays, and it was found that gigantol had no affect on RAW 264.7 cell viability even at 150 μg/mL for 24 h. However, gigantol in the presence of LPS reduced cell viability by 27.5 % at the highest concentration used (150 μg/mL) (see Fig. 1S in Supporting Information).
Since gigantol was found to inhibit both NO and PGE2 production, we investigated whether these inhibitory effects are related to iNOS and COX-2 modulation using Western blot and RT-PCR. In unstimulated RAW 264.7 cells, iNOS and COX-2 at the protein and mRNA levels were undetectable. However, both iNOS and COX-2 were markedly induced by LPS treatment, and gigantol significantly inhibited these expressions in a concentration-dependent manner (Fig. [3] A). RT-PCR also showed that iNOS and COX-2 mRNA expressions were correlated with their protein levels. However, gigantol did not affect the expression of β-actin, the housekeeping gene. These results are consistent with the inhibitory effects of gigantol on NO and PGE2 production (Fig. [2]).
We also tested the effect of gigantol on LPS-induced TNF-α, IL-1β and IL-6 release using enzyme immunoassays (EIAs) and RT-PCR. We found that gigantol pretreatment reduced TNF-α, IL-1β and IL-6 expression at the protein (Fig. [4] A, B and C) and mRNA (Fig. [4] D) levels in RAW 264.7 macrophage cells in a concentration-dependent manner.
Since the activation of NF-κB is critically required for the activation of iNOS, COX-2, TNF-α, IL-1β and IL-6 by LPS [9], [17], we examined the effect of gigantol on LPS-stimulated NF-κB-dependent reporter gene expression. In this study, we used a pNF-κB-luc plasmid, which was generated by inserting four spaced NF-κB binding sites into pLuc-promoter vector. RAW 264.7 cells were transiently transfected with pNF-κB-luc plasmid and then stimulated with 1 μg/mL LPS in the presence or absence of gigantol. It was found that gigantol treatment significantly reduced the LPS-induced increase in NF-κB-dependent luciferase enzyme expression (Fig. [5] A). In addition, LPS-induced NF-κB-DNA binding and transcriptional activity in RAW 264.7 macrophage cells was significantly inhibited by gigantol in a concentration-dependent manner (Fig. [5] B). Taken together, the above findings demonstrate that gigantol suppresses iNOS, COX-2, TNF-α, IL-1β and IL-6 expressions at least in part via an NF-κB-dependent mechanism.

Fig. 2 Effects of gigantol on nitrite (A) and PGE2 (B) production by LPS in RAW 264.7 cells. A Cells were pretreated for 1 h with different concentrations (25, 50, 100 μg/mL) of gigantol and then LPS (1 μg/mL) was added and incubated for 24 h. Control (Con) values were obtained in the absence of LPS and gigantol. 10 μM of l-N 6-(1-iminoethyl)lysine (L-NIL) was used as a positive control. B The treatment conditions used were identical to those described in the legend of Fig. 2A. 10 μM of NS-398 were used as a positive control. Values represent mean ± S.D. of three independent experiments. * P < 0.05 compared with LPS alone.

Fig. 3 The effects of gigantol on LPS-induced iNOS and COX-2 expressions in RAW 264.7 cells. A Cells were treated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h. LPS (1 μg/mL) was then added, and cells were incubated for 24 h. Total cellular proteins (40 μg) were resolved by SDS-PAGE, transferred to nitrocellulose membranes, and detected with specific antibodies, as described in Materials and Methods. A representative immunoblot of three separate experiments is shown. B Total RNA was prepared for the RT-PCR analyses of iNOS and COX-2 expressions in RAW 264.7 macrophages pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h followed by LPS (1 μg/mL) treatment for 4 h. iNOS-specific sequences (807 bp) and COX-2-specific sequences (721 bp) were detected by agarose gel electrophoresis, as described in methods. PCR of β-actin was performed to verify that the initial cDNA contents of samples were similar. Experiments were repeated three times and similar results were obtained.

Fig. 4 The effects of gigantol on LPS-induced TNF-α, IL-1β and IL-6 in RAW 264.7 cells. A Cells were pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h. LPS (1 μg/mL) was then added and cells were incubated for 24 h. Control (Con) values were obtained in the absence of LPS and gigantol. The values shown represent the mean ± S.D. of three independent experiments. * P < 0.05 compared with LPS alone. B Total RNA was prepared for the RT-PCR analysis of TNF-α, IL-1β and IL-6 gene expressions in RAW 264.7 macrophages pretreated with different concentrations (25, 50, 100 μg/ml) of gigantol for 1 h and then with LPS (1 μg/mL) for 4 h. TNF-α specific sequences (351 bp), IL-1β-specific sequences (387 bp) and IL-6-specific sequences (142 bp) were detected by agarose gel electrophoresis. PCR of β-actin was performed to verify that the initial cDNA contents of the samples were similar. Experiments were repeated three times and similar results were obtained.

Fig. 5 The inhibition of p65-DNA binding and NF-κB activation by gigantol. A RAW 264.7 cells were transiently co-transfected with pNF-κB-Luc reporter and then left untreated (Con) or were pretreated with different concentrations (25, 50, 100 μg/mL of gigantol. LPS (1 μg/mL) was then added and the cells were further incubated for 3 h. The cells were then harvested and luciferase activities were determined using a Promega luciferase assay system and a luminometer. The values shown represent mean ± S.D. of three independent experiments. ** P < 0.01, *** P < 0.001 vs. the LPS only treated group; the significances of differences between the groups were evaluated using the Student’s t-test. B Nuclear extracts were prepared from untreated RAW 264.7 cells or cells pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h followed by LPS (1 μg/mL) for 1 h, and then analyzed for NF-κB binding by EMSA. The arrow indicates the NF-κB band. Data shown are representative of three independent experiments.
Discussion
During our continued searches for novel anti-inflammatory agents from natural products, we found that the ethyl acetate fraction of C. goeringii inhibited LPS-induced NO production in RAW 264.7 cells. Activity-guided fractionation led to the isolation of the bibenzyl derivative, gigantol. Although it has been previously reported that gigantol has various pharmacological effects, i. e., anti-mutagenic, spasmolytic, antifungal and antibacterial effects [18], [19], [20], [21], its anti-inflammatory effect and the mechanism involved have not been addressed.
In murine macrophage RAW 264.7 cells, LPS induces iNOS transcription and transduction, and this is followed by NO production. Furthermore, LPS stimulation is known to induce IκB proteolysis and NF-κB nuclear translocation [10]. Therefore, RAW 264.7 cells provide us with an excellent model for drug screening and for subsequently evaluating potential inhibitors of the pathway leading to the induction of iNOS and the production of NO. The reactive free radical NO is a major macrophage-derived inflammatory mediator and has also been reported to be involved in the pathogenesis of inflammatory diseases [22]. In addition, a large amount of evidence suggests that prostaglandins (PGs) are involved in various pathophysiological processes, including those of inflammation and carcinogenesis, and that the inducible isoform of cyclooxygenase (COX-2) is mainly responsible for the production of large amounts of these mediators [23]. Based on these findings, the anti-inflammatory activities of gigantol on LPS-induced NO, PGE2 and TNF-α production in murine macrophage RAW 264.7 cells were studied. To further investigate the inhibitory effect of gigantol on these pro-inflammatory molecules, we investigated the expression of iNOS and COX-2 proteins and of iNOS, COX-2 and TNF-α mRNA. Inhibition of the expression of iNOS and COX-2 at the gene level was evidenced by reductions in their mRNA levels in a concentration-dependent manner. Thus, the inhibition of NO and PGE2 release may be attributed to the suppression of iNOS and COX-2 mRNA transcriptions. Importantly, the inhibition of the LPS-induced expression of these molecules in RAW 264.7 cells by gigantol was not due to gigantol’s cytotoxicity, as assessed by MTT assay and expression of the housekeeping gene β-actin.
It has been reported that cytokines such as TNF-α, IL-1β and IL-6 are pro-inflammatory in vitro and in vivo [6]. Moreover, the production of TNF-α is crucial for the synergistic induction of NO synthesis in IFN-γ and/or LPS-stimulated macrophages [24]. TNF-α elicits a number of physiological effects, such as septic shock, inflammation, cachexia, and cytotoxicity [25]. In particular, IL-1 is a major pro-inflammatory cytokine, and mainly released by macrophages and is believed to play a considerable role in the pathophysiology of endometriosis [26]. IL-6 is also a pivotal pro-inflammatory cytokine, and is regarded as an endogenous mediator of LPS-induced fever. In the present study, we found that gigantol also significantly inhibits the releases of TNF-α and IL-1β, -6 and their mRNA expression.
NF-κB is known to play a critical role in the regulation of cell survival genes and to coordinate the expressions of pro-inflammatory enzymes and cytokines, such as, iNOS, COX-2, TNF-α, and IL-1β, IL-6 [9]. Since the expressions of these pro-inflammatory mediators are known to be modulated by NF-κB, we examined the possibility that gigantol inhibits NF-κB activity. Thus, we examined NF-κB activation and the DNA binding activity of p65 to confirm the inhibitions of the expressions of iNOS, COX-2, TNF-α, IL-1β and IL-6 because we believed that the inhibitory effects of gigantol on NO, PGE2, TNF-α, IL-1β and IL-6 production were regulated by the NF-κB signaling pathway. Our results suggest that NF-κB activation and the DNA binding activity of p65 are in fact inhibited in a concentration-dependent manner by gigantol.
In conclusion, we found that gigantol is a potent inhibitor of LPS-induced NO, PGE2, TNF-α, IL-1β and IL-6 production, and that it acts at the transcription level. These inhibitions were found to be caused by the prevention of NF-κB activation in RAW 264.7 macrophages. Therefore, we conclude that gigantol appears to have potential to prevent inflammatory disease.
#Acknowledgements
This research was supported by a grant from the ‘Investigation of biological activity components and evaluation of pharmacological efficacy in Ganghwa indigeous Crops’.
- Supporting Information for this article is available online at
- Supporting Information .
References
- 1 Vane J R, Mitchell J A, Appleton I, Tomlinson A, Bishop-Bailey D, Croxtall J. et al . Inducible isoforms of cyclooxygenase and nitric-oxide synthase in inflammation. Pharmacology. 1994; 91 2046-50
- 2 Funk C D, Funk L B, Kennedy M E, Pong A S, Fittzgerald G A. Human platelet/erythroleukemia cell prostaglandin G/H synthase: cDNA cloning, expression and gene chromosomal assignment. FASEB J. 1991; 5 2304-12
- 3 Hinz B, Brune K. Cyclooxygenase-2 - 10 years later. J Pharmacol Exp Ther. 2002; 300 367-75
- 4 Guha M, Mackman N. LPS induction of gene expression in human monocytes. Cell Signal. 2001; 13 85-94
- 5 Beutler B, Cerami A. The biology of cachectin/TNF-α primary mediator of the host response. Annu Rev Immunol. 1989; 7 625-55
- 6 Molloy R G, Mannick J A, Rodrick M L. Cytokines, sepsis and immunomodulation. Br J Surg. 1993; 80 289-97
- 7 Van Snick J. IL-6: an overview. Annu Rev Immunol. 1990; 8 253-78
- 8 Dendorfer U. Molecular biology of cytokines. Artif Organs. 1996; 20 437-44
- 9 Surh Y J, Chun K S, Cha H H, Han S S, Keum Y S, Park K K. et al . Molecular mechanism underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001; 480 - 1 243-68
- 10 Baeuerle P A. IkappaB-NF-kappaB structures: at the interface of inflammation control. Cell. 1998; 95 729-31
- 11 Jung B S, Shin M K. In: Jung BS, Shin MK, editors Hyang Yak Dae Sa Jun, 3rd edition. Seoul; Young Lim Book Center 1990: p 134-5
- 12 Lee J H, Kim D H, Bang M H, Yang H J, Bang S H, Chung I S. et al . Isolation of sterols from the methanol extracts of Cymbidium goeringii Reichb. Fil. J Korean Soc Appl Biol Chem. 2005; 48 263-6
- 13 Juneja R K, Sharma S C, Tandon J S. Two substituted bibenzyls and a dihydrophenanthrene from Cymbidium aloifolium . Phytochemistry. 1987; 26 1123-5
- 14 Juneja R K, Sharma S C, Tandon J S. A substituted 1,2-diarylethane from Cymbidium giganteum . Phytochemistry. 1985; 24 321-4
- 15 Shin K M, Kim I T, Park Y M, Ha J, Choi J W, Park H J. et al . Anti-inflammatory effect of caffeic acid methyl ester and its mode of action through the inhibition of prostaglandin E2, nitric oxide and tumor necrosis factor-alpha production. Biochem Pharmacol. 2004; 68 2327-36
- 16 Bredt D S, Snyder S H. Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci USA. 1990; 87 682-5
- 17 Lappas M, Permezel M, Georgiou H M, Rice G E. Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro . Biol Reprod. 2002; 67 668-73
- 18 Miyazawa M, Shimamura H, Nakamura S I, Kameoka H. Antimutagenic activity of gigantol from Dendrobium nobile . J Agric Food Chem. 1997; 45 2849-53
- 19 Hernandez-Romero Y, Rojas J I, Castillo R, Rojas A, Mata R. Spasmolytic effects, mode of action and structure-activity relationships of stilbenoids from Nidema boothii . J Nat Prod. 2004; 67 160-7
- 20 Estrada S, Rojas A, Mathison Y, Israel A, Mata R. Nitric oxide/cGMP mediates the spasmolytic action of 3,4"-dihydroxy-5,5"-dimethoxybibenzyl from Scaphyglottis livida . Planta Med. 1999; 65 109-14
- 21 Matsuura H, Saxena G, Farmer S W, Hancock R EW, Towers G HN. Antibacterial and antifungal compounds from Empetrum nigrum . Planta Med. 1995; 61 580
- 22 Xie Q W, Kashiwabara Y, Nathan C. Carboxyl terminus of inducible nitric oxide synthase: Contribution to NADPH binding and enzymatic activity. J Biol Chem. 1994; 269 4705-8
- 23 Simon L S. Role of regulation of cyclooxygenase-2 during inflammation. Am J Med. 1999; 106 S37-42
- 24 Jun C D, Choi B M, Kim H M, Chung H T. Involvement of protein kinase C during taxol-induced activation of murine peritoneal macrophages. J Immunol. 1995; 154 6541-7
- 25 Mannel D N, Echtenacher B. TNF in the inflammatory response. Chem Immunol. 2000; 74 141-61
- 26 Bergqvist A, Bruse C, Carlberg M, Carlstrom K. Interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in endometriotic tissue and in endometrium. Fertil Steril. 2001; 75 489-95
Kyung-Tae Lee, Ph. D.
Department of Pharmaceutical Biochemistry
College of Pharmacy
Kyung-Hee University
Dongdaemun-Ku
Hoegi-Dong 130-701
Seoul
Korea
Phone: +82-2-961-0860
Fax: +82-2-966-3885
Email: ktlee@khu.ac.kr
References
- 1 Vane J R, Mitchell J A, Appleton I, Tomlinson A, Bishop-Bailey D, Croxtall J. et al . Inducible isoforms of cyclooxygenase and nitric-oxide synthase in inflammation. Pharmacology. 1994; 91 2046-50
- 2 Funk C D, Funk L B, Kennedy M E, Pong A S, Fittzgerald G A. Human platelet/erythroleukemia cell prostaglandin G/H synthase: cDNA cloning, expression and gene chromosomal assignment. FASEB J. 1991; 5 2304-12
- 3 Hinz B, Brune K. Cyclooxygenase-2 - 10 years later. J Pharmacol Exp Ther. 2002; 300 367-75
- 4 Guha M, Mackman N. LPS induction of gene expression in human monocytes. Cell Signal. 2001; 13 85-94
- 5 Beutler B, Cerami A. The biology of cachectin/TNF-α primary mediator of the host response. Annu Rev Immunol. 1989; 7 625-55
- 6 Molloy R G, Mannick J A, Rodrick M L. Cytokines, sepsis and immunomodulation. Br J Surg. 1993; 80 289-97
- 7 Van Snick J. IL-6: an overview. Annu Rev Immunol. 1990; 8 253-78
- 8 Dendorfer U. Molecular biology of cytokines. Artif Organs. 1996; 20 437-44
- 9 Surh Y J, Chun K S, Cha H H, Han S S, Keum Y S, Park K K. et al . Molecular mechanism underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001; 480 - 1 243-68
- 10 Baeuerle P A. IkappaB-NF-kappaB structures: at the interface of inflammation control. Cell. 1998; 95 729-31
- 11 Jung B S, Shin M K. In: Jung BS, Shin MK, editors Hyang Yak Dae Sa Jun, 3rd edition. Seoul; Young Lim Book Center 1990: p 134-5
- 12 Lee J H, Kim D H, Bang M H, Yang H J, Bang S H, Chung I S. et al . Isolation of sterols from the methanol extracts of Cymbidium goeringii Reichb. Fil. J Korean Soc Appl Biol Chem. 2005; 48 263-6
- 13 Juneja R K, Sharma S C, Tandon J S. Two substituted bibenzyls and a dihydrophenanthrene from Cymbidium aloifolium . Phytochemistry. 1987; 26 1123-5
- 14 Juneja R K, Sharma S C, Tandon J S. A substituted 1,2-diarylethane from Cymbidium giganteum . Phytochemistry. 1985; 24 321-4
- 15 Shin K M, Kim I T, Park Y M, Ha J, Choi J W, Park H J. et al . Anti-inflammatory effect of caffeic acid methyl ester and its mode of action through the inhibition of prostaglandin E2, nitric oxide and tumor necrosis factor-alpha production. Biochem Pharmacol. 2004; 68 2327-36
- 16 Bredt D S, Snyder S H. Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci USA. 1990; 87 682-5
- 17 Lappas M, Permezel M, Georgiou H M, Rice G E. Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro . Biol Reprod. 2002; 67 668-73
- 18 Miyazawa M, Shimamura H, Nakamura S I, Kameoka H. Antimutagenic activity of gigantol from Dendrobium nobile . J Agric Food Chem. 1997; 45 2849-53
- 19 Hernandez-Romero Y, Rojas J I, Castillo R, Rojas A, Mata R. Spasmolytic effects, mode of action and structure-activity relationships of stilbenoids from Nidema boothii . J Nat Prod. 2004; 67 160-7
- 20 Estrada S, Rojas A, Mathison Y, Israel A, Mata R. Nitric oxide/cGMP mediates the spasmolytic action of 3,4"-dihydroxy-5,5"-dimethoxybibenzyl from Scaphyglottis livida . Planta Med. 1999; 65 109-14
- 21 Matsuura H, Saxena G, Farmer S W, Hancock R EW, Towers G HN. Antibacterial and antifungal compounds from Empetrum nigrum . Planta Med. 1995; 61 580
- 22 Xie Q W, Kashiwabara Y, Nathan C. Carboxyl terminus of inducible nitric oxide synthase: Contribution to NADPH binding and enzymatic activity. J Biol Chem. 1994; 269 4705-8
- 23 Simon L S. Role of regulation of cyclooxygenase-2 during inflammation. Am J Med. 1999; 106 S37-42
- 24 Jun C D, Choi B M, Kim H M, Chung H T. Involvement of protein kinase C during taxol-induced activation of murine peritoneal macrophages. J Immunol. 1995; 154 6541-7
- 25 Mannel D N, Echtenacher B. TNF in the inflammatory response. Chem Immunol. 2000; 74 141-61
- 26 Bergqvist A, Bruse C, Carlberg M, Carlstrom K. Interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in endometriotic tissue and in endometrium. Fertil Steril. 2001; 75 489-95
Kyung-Tae Lee, Ph. D.
Department of Pharmaceutical Biochemistry
College of Pharmacy
Kyung-Hee University
Dongdaemun-Ku
Hoegi-Dong 130-701
Seoul
Korea
Phone: +82-2-961-0860
Fax: +82-2-966-3885
Email: ktlee@khu.ac.kr

Fig. 1 Chemical structure of gigantol.

Fig. 2 Effects of gigantol on nitrite (A) and PGE2 (B) production by LPS in RAW 264.7 cells. A Cells were pretreated for 1 h with different concentrations (25, 50, 100 μg/mL) of gigantol and then LPS (1 μg/mL) was added and incubated for 24 h. Control (Con) values were obtained in the absence of LPS and gigantol. 10 μM of l-N 6-(1-iminoethyl)lysine (L-NIL) was used as a positive control. B The treatment conditions used were identical to those described in the legend of Fig. 2A. 10 μM of NS-398 were used as a positive control. Values represent mean ± S.D. of three independent experiments. * P < 0.05 compared with LPS alone.

Fig. 3 The effects of gigantol on LPS-induced iNOS and COX-2 expressions in RAW 264.7 cells. A Cells were treated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h. LPS (1 μg/mL) was then added, and cells were incubated for 24 h. Total cellular proteins (40 μg) were resolved by SDS-PAGE, transferred to nitrocellulose membranes, and detected with specific antibodies, as described in Materials and Methods. A representative immunoblot of three separate experiments is shown. B Total RNA was prepared for the RT-PCR analyses of iNOS and COX-2 expressions in RAW 264.7 macrophages pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h followed by LPS (1 μg/mL) treatment for 4 h. iNOS-specific sequences (807 bp) and COX-2-specific sequences (721 bp) were detected by agarose gel electrophoresis, as described in methods. PCR of β-actin was performed to verify that the initial cDNA contents of samples were similar. Experiments were repeated three times and similar results were obtained.

Fig. 4 The effects of gigantol on LPS-induced TNF-α, IL-1β and IL-6 in RAW 264.7 cells. A Cells were pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h. LPS (1 μg/mL) was then added and cells were incubated for 24 h. Control (Con) values were obtained in the absence of LPS and gigantol. The values shown represent the mean ± S.D. of three independent experiments. * P < 0.05 compared with LPS alone. B Total RNA was prepared for the RT-PCR analysis of TNF-α, IL-1β and IL-6 gene expressions in RAW 264.7 macrophages pretreated with different concentrations (25, 50, 100 μg/ml) of gigantol for 1 h and then with LPS (1 μg/mL) for 4 h. TNF-α specific sequences (351 bp), IL-1β-specific sequences (387 bp) and IL-6-specific sequences (142 bp) were detected by agarose gel electrophoresis. PCR of β-actin was performed to verify that the initial cDNA contents of the samples were similar. Experiments were repeated three times and similar results were obtained.

Fig. 5 The inhibition of p65-DNA binding and NF-κB activation by gigantol. A RAW 264.7 cells were transiently co-transfected with pNF-κB-Luc reporter and then left untreated (Con) or were pretreated with different concentrations (25, 50, 100 μg/mL of gigantol. LPS (1 μg/mL) was then added and the cells were further incubated for 3 h. The cells were then harvested and luciferase activities were determined using a Promega luciferase assay system and a luminometer. The values shown represent mean ± S.D. of three independent experiments. ** P < 0.01, *** P < 0.001 vs. the LPS only treated group; the significances of differences between the groups were evaluated using the Student’s t-test. B Nuclear extracts were prepared from untreated RAW 264.7 cells or cells pretreated with different concentrations (25, 50, 100 μg/mL) of gigantol for 1 h followed by LPS (1 μg/mL) for 1 h, and then analyzed for NF-κB binding by EMSA. The arrow indicates the NF-κB band. Data shown are representative of three independent experiments.
- www.thieme-connect.de/ejournals/toc/plantamedica