c‑Jun N‑terminal kinase‑mediated anti‑inflammatory effects of Garcinia subelliptica in macrophages

  • Authors:
    • Young‑Chang Cho
    • Sayeon Cho
  • View Affiliations

  • Published online on: January 18, 2016     https://doi.org/10.3892/mmr.2016.4791
  • Pages: 2293-2300
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Garcinia plants have been traditionally used to treat inflammatory diseases, such as skin infections and pain, in many regions including South‑East Asia. Garcinia subelliptica, a plant of the Garcinia species widely distributed from Japan to Thailand, has been reported to contain components similar to other Garcinia plants that exhibit anti‑inflammatory effects. The present study aimed to explore the anti‑inflammatory effects of ethanol extracts of Garcinia subelliptica (EGS) in macrophages, as there are no previous systemic studies that have investigated the effects of Garcinia subelliptica on inflammation. Non‑cytotoxic concentrations of EGS (≤200 µg/ml) were observed to reduce nitric oxide production by modulating iNOS expression in lipopolysaccharide (LPS)‑stimulated RAW 264.7 macrophages. The expression of cyclooxygenase‑2, the enzyme responsible for the production of prostaglandin E2, was notably reduced by EGS. EGS treatment inhibited the production of pro‑inflammatory cytokines, including IL‑6 and IL‑1β, however, not TNF‑α. Reduced production of inflammatory mediators by EGS was followed by reduced phosphorylation of c‑Jun N‑terminal kinase (JNK) however, not of other mitogen‑activated protein kinases and nuclear factor‑κB. These results indicate that EGS selectively inhibits the excessive production of inflammatory mediators in LPS‑stimulated murine macrophages by reducing the activation of JNK, suggesting that EGS is a candidate for modulating severe inflammation.

Introduction

Macrophages are important cells in the immune system that act as the first defense against invading agents (bacteria, viruses, and fungi) by releasing cellular signaling molecules and antimicrobial agents (14). Inflammatory responses in macrophages are induced upon the stimulation of macrophages with external stimuli, such as lipopolysaccharides (LPS). Inflammatory responses include the expression of various pro-inflammatory mediators, such as nitric oxide (NO), which is produced by inducible nitric oxide synthase (iNOS), and prostaglandin E2 (PGE2), which is produced by cyclooxygenase (COX)-2, in addition to the expression of various pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α (510). Excessive amounts of these mediators cause severe inflammatory diseases, including septic shock, rheumatoid arthritis, systemic lupus erythematosus and inflammatory bowel disease, although the enhanced production of inflammatory mediators is important for the host defense against external stimuli (1016). Therefore, inhibiting the excessive production of inflammatory mediators in macrophages by regulating mRNA and protein expression levels may be a viable strategy to develop effective anti-inflammatory agents.

A study from the World Health Organization has indicated that greater than 80% of the population in developing countries still use natural plants as their main medicinal source (17). The Garcinia species have been used traditionally for the treatment of inflammatory diseases worldwide. The ethanol and dichloromethane extracts of the fruit hull of Garcinia mangostana, a Thai traditional medicine for the treatment of abscesses and skin infections (18), have been reported to exhibit anti-inflammatory activity (19) and potent antinociceptive effects in mice (20). The ethyl acetate extract of Garcinia hanburyi, a Thai folk medicine used to treat infected wounds (21,22), has been assessed for anti-inflammatory, analgesic and antipyretic activities (23). The hydroalcoholic extracts of the leaves from Garcinia gardneriana, a plant traditionally used in southern Brazil to treat skin disorders (24), have shown anti-inflammatory activity in several experimental models (25).

Polyphenols and their metabolites in Garcinia plants are known to exhibit anti-inflammatory properties. Of these poly-phenols, kolaviron, a biflavonoid complex from Garcinia kola, and garcinol, a polyisoprenylated benzophenone derivative isolated from Garcinia indica, have been applied to animal models to evaluate their anti-inflammatory effects (2629). Garcinia subelliptica (G. subelliptica) is a Garcinia species that is distributed widely from Okinawa Island in Japan to Thailand, and contains secondary polyphenolic metabolites which are known to exert anti-inflammatory effects (3032). Furthermore, G. subelliptica has been reported to contain phloroglucinols and terpenoids that exhibit anti-inflammatory properties similar to those of other Garcinia plants (33). Garcinielliptones exert anti-inflammatory effects by inhibiting chemical mediators and xanthine oxidase in mast cells, neutrophils and macrophages (3335). However, systemic studies for the anti-inflammatory effects of the ethanol extract of G. subelliptica (EGS) have not been reported.

In the present study, the anti-inflammatory effects of EGS exerted through the modulation of the production of inflammatory mediators in LPS-stimulated RAW 264.7 macrophages (ATSS, Manassas, VA, USA) were investigated to evaluate the potential of EGS for the treatment of excessive inflammation. Furthermore, the associated signaling pathways, such as the phosphorylation of the inhibitor of κB (IκB) and the mitogen-activated protein kinase (MAPK) pathway, were investigated to assess the molecular targets of EGS.

Materials and methods

Cell culture and reagents

A 95% ethanol extract (Code no.: FBM124-035) of the Garcinia subelliptica Merr. (Clusiaceae) was purchased from the International Biological Material Research Center (Daejeon, Korea). The RAW 264.7 macrophages, a mouse monocytic cell line, were cultured in Dulbecco's modified Eagle's medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.), 50 unit/ml penicillin, and 50 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in humidified air containing 5% CO2. The cytokines that were produced, including TNF-α and IL-6, were measured using enzyme-linked immunosorbent assay (ELISA) Ready-SET-Go!® kits (eBioscience, Inc., San Diego, CA, USA). Rabbit anti-IκBα (sc-371) and rabbit anti-glyceraldehyde 3-phosphate dehydrogenase (GAPDH; sc-25778) polyclonal antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Mouse anti-phosphoylated (p)-IκBα (Ser32/36; 9246) and anti-p-extracellular signal-regulated kinase 1/2 (ERK 1/2; 9106) monoclonal antibodies; and rabbit anti-iNOS (2982), anti-COX-2 (4842), anti-p-p38 (9211), anti-p38 (9212), anti-ERK 1/2 (9102), anti-p-c-Jun N-terminal kinase (JNK; 9251) and anti-JNK (9252) polyclonal antibodies; and goat anti-rabbit horseradish peroxidase (HRP)-conjugated immunoglobulin (Ig)G (7074) and horse anti-mouse HRP-conjugated IgG (7076) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Cell viability assay

RAW 264.7 macrophages were seeded in 96 well plates (4×104/well). Following adhesion overnight, cells were incubated with 20, 50, 100, 200, and 400 µg/ml EGS and 1 µg/ml LPS for 24 h. Following incubation, cell viability was measured using an EZ-Cytox Cell Viability Assay kit (Daeil Lab Services Co., Ltd., Seoul, Korea). Briefly, the EZ-Cytox solution that contained a water soluble tetrazolium salt was added to each well for 2 h at 37°C and 100 µl of supernatants were transferred to 96 well plates. The absorbance was measured at 450 nm with a Synergy H1 Microplate Reader (BioTek Instruments, Inc., Winooski, VT, USA).

Nitrite assay

RAW 264.7 macrophages were incubated with 20, 50, 100 and 200 µg/ml EGS and 1 µg/ml LPS for 24 h. Following incubation the levels of NO synthesis were determined by assaying the culture supernatants for nitrite using the Griess reagent (1% sulfanilamide, 0.1% N-1-naphthylenediamine dihydrochloride, and 2.5% phosphoric acid). Nitrate is the stable product of the reaction between NO and molecular oxygen. The absorbance was measured at 540 nm, using a Synergy H1 Microplate Reader, following incubation for 10 min.

ELISA

RAW 264.7 macrophages were stimulated with 1 µg/ml LPS and 20, 50, 100 and 200 µg/ml EGS for 24 h. Following stimulation, the supernatants were obtained via centrifugation at 2,339 × g for 3 min at 4°C, and a sandwich ELISA was performed according to the manufacturer's instructions, using monoclonal antibodies specific to each mediator to determine the quantity of TNF-α and IL-6 in the culture supernatants. Prior to the application of samples, the plate was pre-coated with the coating antibody in the supplied buffer. Following incubation overnight at 4°C, the plate was washed with 1X phosphate-buffered saline (PBS)-Tween 20 (0.5%) and treated with 1X assay diluents for 1 h. Samples were loaded into each well and incubated for 2 h at room temperature. Following washing, the plate was treated with a biotinylated secondary antibody solution and HRP-streptavidin solution for 1.5 h, and the substrate solution was added to the washed-plate. Following 10 min incubation in dark conditions, 1 N phosphoric acid (H3PO4) was added and the optical density of the individual wells was measured at 450 nm using a Synergy Microplate Reader.

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

RAW 264.7 macrophages were stimulated with 50 ng/ml LPS and 20, 50, 100 and 200 µg/ml EGS for 6 h. Total RNA was extracted from the cells via isoproponal precipitation using Accuzol reagent (Bioneer Corporation, Daejeon, Korea), reverse-transcribed into complementary DNA (cDNA) using a TOPscript™ cDNA synthesis kit (Enzynomics, Daejeon, Korea) and then PCR amplified. Quantification of mRNA was performed using a real-time PCR reagent, iTaq™ Universal SYBR Green Supermix (Bio-Rad Laboratories, Inc., Hercules, CA, USA), according to the manufacturer's instructions. The PCR was run for 40 cycles of denaturation at 94°C (5 sec) and annealing/extension at 60°C (30 sec) using a CFX Connect™ Real-Time Thermal Cycler (Bio-Rad Laboratories, Inc.). According to the 2−∆∆Cq method (36), the results were normalized to the reference genes, β-actin and GAPDH, and were expressed as the ratio of gene expressions to the LPS treated group (100%). PCR primers were designed using Beacon Designer 7.0 (Premier Biosoft, Palo Alto, CA, USA) and subsequently synthesized by Bioneer Corporation. The sequences of PCR primers used are as follows: Mouse iNOS, sense 5′-TGG CCA CCA AGC TGA ACT-3′ and antisense 5′-TCA TGA TAA CGT TTC TGG CTCTT-3′; COX-2, sense 5′-GAT GCT CTT CCG AGC TGTG-3′ and antisense 5′-GGA TTG GAA CAG CAA GGA TTT-3′; TNF-α, sense 5′-CTG TAG CCC ACG TCG TAGC-3′ and antisense 5′-TTG AGA TCC ATG CCG TTG-3′; IL-6, sense 5′-TCT AAT TCA TAT CTT CAA CCA AGAGG-3′ and antisense 5′-TGG TCC TTA GCC ACT CCTTC-3′; IL-1β, sense 5′-TTG ACG GAC CCC AAA AGAT-3′ and antisense 5′-GAT GTG CTG CTG CGA GATT-3′; β-actin, sense 5′-CGT CAT ACT CCT GCT TGCTG-3′ and antisense 5′-CCA GAT CAT TGC TCC TCC TGA-3′; and GAPDH, sense 5′-GCT CTC TGC TCC TCC TGTTC-3′ and antisense 5′-ACG ACC AAA TCC GTT GACTC-3′.

Semi-quantitative RT-PCR

PCR primers were designed using Primer3 software (version 4.0.0), as previously described (37), and were subsequently synthesized by Bioneer Corporation. The sequences of the PCR primers used study are as follow: Mouse iNOS, sense 5′-GCA TGG AAC AGT ATA AGG CAA ACA-3′ and antisense 5′-GTT TCT GGT CGA TGT CAT GAG CAA-3′; COX-2, sense 5′-GCA TGG AAC AGT ATA AGG CAA ACA-3′ and antisense 5′-GTT TCT GGT CGA TGT CAT GAG CAA-3′; TNF-α, sense 5′-GTG CCA GCC GAT GGG TTG TACC-3′ and antisense 5-′AGG CCC ACA GTC CAG GTC ACTG-3′; IL-6, sense 5′-TCT TGG GAC TGA TGC TGG TGAC-3′ and antisense 5′-CAT AAC GCA CTA GGT TTG CCGA-3′; IL-1β, sense 5′-AGC TGT GGC AGC TAC CTGTG-3′ and antisense 5′-GCT CTG CTT GTG AGG TGCTG-3′; and GAPDH, sense 5′-GTC TTC ACC ACC ATG GAG AAGG-3′ and antisense 5′-CCT GCT TCA CCA CCT TCT TGCC-3′. The PCR was run for 20–25 cycles of 94°C (30 sec), 60°C (30 sec), and 72°C (30 sec) using a Genetouch thermal cycler (Bioer Technology Co., Ltd., Hangzhou, China). Following amplification, 10 µl of the RT-PCR products were separated in 1.5% (w/v) agarose gels and stained with ethidium bromide.

Preparation of total cell lysates

LPS-stimulated RAW 264.7 cells were treated with EGS and 1 µg/ml LPS for 15 min and washed with ice-cold PBS. The cells were lysed in lysis buffer containing 0.5% NP-40, 0.5% Triton X-100, 150 mM NaCl, 20 mM Tris-HCl (pH 8.0), 1 mM ethylenediaminetetraacetic acid, 1% glycerol, 1 mM phenylmethylsulfonyl fluoride and 1 µg/ml aprotinin, and collected in microtubes prior to centrifugation at 15,814 x g for 30 min at 4°C. The supernatants were prepared in fresh microtubes.

Immunoblot analysis

Protein concentration was measured using the Bradford method (Bio-Rad Laboratories, Inc.). Aliquots (20 µg) of the cell lysates were separated by 10% sodium dodecyl sulfate-polyacrylamide gel in a Mini-Protein II gel apparatus (both Bio-Rad Laboratories, Inc.) and transferred onto nitrocellulose membranes (GE Healthcare Life Sciences, Pittsburgh, WI, USA) with transfer buffer [192 mM glycine, 25 mM Tris-HCl (pH 8.8), and 20% MeOH (v/v)]. Following blocking of non-specific sites with 5% bovine serum albumin solution, the membrane was incubated overnight at 4°C with the primary antibodies (all 1:1,000). Subsequently, each membrane was incubated for 1 h at room temperature with secondary HRP-conjugated goat anti-rabbit or horse anti-mouse IgG (1:5,000). The target proteins were detected using an enhanced chemiluminescence solution (Daeil Lab Services Co., Ltd.). Protein levels were quantified by scanning the immunoblots and analyzing them with LabWorks software (version 4.6; UVP, Inc., Upland, CA, USA).

Statistical analysis and experimental replicates

Data are presented as the mean ± standard error of the mean. Statistical differences between each result were assessed by comparing with the LPS-treated group using the Mann-Whitney U test. Mann-Whitney U test was performed using Prism 3.0 (GraphPad Software, Inc., San Diego, CA, USA) and P<0.01 was considered to indicate a statistically significant difference. The data from nine replicates were analyzed, including three independent experiments with three replicates in each.

Results

Determination of the non-cytotoxic concentration of EGS in macrophages

Selective regulation of inflammatory mediators and the identification of specific target signaling molecules are valuable strategies for the development of novel anti-inflammatory reagents. Based on this concept, the current study investigated a number of natural plant extracts, which are known traditionally to have pharmacological effects, and evaluated their inhibitory effects on inflammation by measuring the production of NO, a major inflammatory mediator, in LPS-stimulated RAW 264.7 macrophages. Of the extracts that exerted an inhibitory effect on NO production, EGS was selected as Garcinia species have been used traditionally to treat numerous inflammatory diseases. Considering that the anti-inflammatory effects of EGS should be studied at concentrations that do not influence cell viability, the non-cytotoxic and maximally effective concentrations of EGS were identified by the dose-dependent treatment of LPS-stimulated RAW 264.7 macrophages. Cell viability was determined by measuring the ability of cells to metabolically reduce a water-soluble tetrazolium salt to a formazan dye. As shown in Fig. 1, the cell viability of the RAW 264.7 macrophages was not reduced by EGS treatment at doses of ≤200 µg/ml in the presence of 1 µg/ml LPS. However, a number of dead cells (31.79±3.14%) were detected when the cells were treated with 400 µg/ml EGS. These results suggest that a low dose of EGS (200 µg/ml) does not affect the viability of RAW 264.7 macrophages. Therefore, concentrations below 200 µg/ml were used in subsequent experiments.

Inhibitory effect of EGS on the production of NO in LPS-stimulated macrophages

To evaluate the anti-inflammatory function of EGS in macrophages, the profile of NO production was measured. As expected, RAW 264.7 macrophages stimulated with LPS (1 µg/ml) produced a large quantity of NO compared with the non-treated control group. When the cells were treated with EGS, the enhanced production of NO by LPS was inhibited in a dose-dependent manner, and the LPS-induced NO production was nearly abolished at a dose of 200 µg/ml (Fig. 2A). Considering that NO production is tightly regulated by iNOS expression, the effects of EGS on the expression of iNOS mRNA and protein were investigated using PCR and immunoblot analysis. The RT-qPCR results revealed that LPS notably induces iNOS expression, which was inhibited by EGS in a dose-dependent manner (Fig. 2B). Semi-quantitative PCR data indicated a similar profile of expression as the RT-qPCR data, which supported the inhibitory effect of EGS on the expression of iNOS mRNA (Fig. 2C). In addition, LPS-induced expression of iNOS protein was notably reduced by EGS in a dose-dependent manner, and the maximum dose of EGS resulted in a similar iNOS expression profile as the non-treated control group (Fig. 2D). These results imply that EGS inhibits NO production in activated macrophages by modulating iNOS expression.

Inhibitory effect of EGS on the expression of COX-2 in LPS-stimulated macrophages

As COX-2 is the enzyme responsible for the production of PGE2 in activated macrophages, the present study explored the effect of EGS on the expression levels of COX-2 mRNA and protein to measure its anti-inflammatory function. As shown in Fig. 3A, induction of COX-2 mRNA expression by LPS was detected by RT-qPCR analysis. EGS treatment notably inhibited the enhanced expression of COX-2 mRNA at concentrations of 100 and 200 µg/ml. Furthermore, the inhibitory effect of EGS on the COX-2 mRNA expression detected by RT-qPCR was supported by semi-quantitative PCR (Fig. 3B). To examine the effect of EGS on the expression of COX-2 protein in activated macrophages, RAW 264.7 macrophages were treated with EGS in the presence of LPS, and total cell lysates were prepared. The increased levels of COX-2 protein expression mediated by LPS were reduced markedly by EGS in a dose-dependent manner (Fig. 3C). From these data, COX-2 expression was observed to be downregulated by EGS, and the subsequent production of PGE2 may therefore be regulated by EGS.

Differential regulation of inflammatory cytokine production by EGS in LPS-stimulated macrophages

To assess the anti-inflammatory properties of EGS in activated macrophages, the effects of EGS on the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α, were measured in LPS-stimulated RAW 264.7 macrophages. Production of IL-6 and TNF-α was increased notably following LPS stimulation (Fig. 4A and B). However, EGS treatment inhibited the production of IL-6 whilst not altering the expression of TNF-α (Fig. 4A and B). The mRNA expression of pro-inflammatory cytokines was assessed to determine whether the production of pro-inflammatory cytokines is regulated at the transcriptional level. As presented in Fig. 4C, RT-qPCR analysis indicated that EGS treatment inhibited the LPS-stimulated expression of IL-6 and IL-1β but not TNF-α. Similarly, semi-quantitative PCR analysis showed that EGS selectively reduced the mRNA expression levels of IL-6 and IL-1β (Fig. 4D). Taken together, these results suggest that EGS negatively regulates the LPS-induced production of pro-inflammatory cytokines by transcriptional repression of IL-6 and IL-1β however, not of TNF-α.

Inhibitory effect of EGS on the activation of JNK in LPS-stimulated macrophages

The phosphorylation levels of JNK and IκB were measured to investigate which inflammatory signaling pathways are associated with the inhibitory effects of EGS in LPS-stimulated macrophages. As shown in Fig. 5A, LPS treatment altered the expression of p-IκBα and IκBα. However, EGS did not affect the expression profiles of p-IκBα and IκBα. In addition, the modulation of JNK phosphorylation by EGS was measured. Immunoblot analyses revealed that EGS inhibited the phosphorylation of JNK without affecting the protein levels of JNK. EGS did not influence the phosphorylation or protein expression of additional MAPK pathway mediators, including ERK and p38. Collectively, these results suggest that EGS exerts anti-inflammatory effects by inhibiting the activation of JNK.

Discussion

In the present study, EGS was observed to markedly inhibit the production of IL-6 and IL-1β, however, not TNF-α, in LPS-stimulated RAW 264.7 macrophages. Numerous reports have indicated that interleukins and TNF-α are not simultaneously inhibited by natural compounds (38,39). One possibility for the differential regulation of pro-inflammatory cytokines by EGS is that they possess different promoter binding regions for transcription factors. Previous studies have demonstrated that the signal transducer and activator of transcription (STAT) protein-binding region is contained in the IL-6 and IL-1β promoter regions, however, not in the TNF-α promoter region (40). This implies that the specific regulation of IL-6 and IL-1β production by EGS in macrophages may be regulated through the inactivation of STAT signaling.

In the present study, LPS treatment of murine macrophages significantly enhanced the production of inflammatory mediators via the stimulation of the MAPK pathway; thus, the inhibition of p38, ERK and JNK phosphorylation may represent potential target pathways for the alleviation of severe inflammatory states (2,8,41). However, numerous reports have suggested that MAPK signaling cascades are differentially involved in the production of inflammatory mediators by macrophages (4244). Watters et al (45) suggested that the MAPK/ERK pathway is not essential for the production of iNOS and IL-1β in macrophages. Caivano (46) and Paul et al (47) reported that the p38 and MAPK/ERK pathways are not essential for the production of inflammatory mediators. In the present study, EGS inhibited the LPS-induced phosphorylation of JNK, however, the total MAPK levels, including p38, ERK and JNK, were unaltered. Collectively, these results suggest that the regulation of JNK activity, rather than total JNK expression, is the key regulatory mechanism of EGS-mediated inhibition of inflammatory mediators.

Considering that macrophages serve pivotal roles in the pathogenesis of many inflammatory diseases, the EGS-mediated selective regulation of inflammatory mediators suggests that EGS may have therapeutic potential against inflammatory diseases. However, further studies are required to analyze the major components of EGS that are responsible for the reduction of inflammatory mediators and to elucidate the exact mechanism underlying the differences in the production of the pro-inflammatory cytokines, IL-6 and TNF-α.

Acknowledgments

The current study was supported by a Chung-Ang University Research Grant in 2014 and a National Research Foundation of Korea (NRF) grant funded by the Ministry of Education, Science and Technology (grant no. NRF-2013R1A1A2062389).

References

1 

Boscá L, Zeini M, Través PG and Hortelano S: Nitric oxide and cell viability in inflammatory cells: A role for NO in macrophage function and fate. Toxicology. 208:249–258. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Fujihara M, Muroi M, Tanamoto K, Suzuki T, Azuma H and Ikeda H: Molecular mechanisms of macrophage activation and deactivation by lipopolysaccharide: Roles of the receptor complex. Pharmacol Ther. 100:171–194. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Fujiwara N and Kobayashi K: Macrophages in inflammation. Curr Drug Targets Inflamm Allergy. 4:281–286. 2005. View Article : Google Scholar : PubMed/NCBI

4 

MacMicking J, Xie QW and Nathan C: Nitric oxide and macrophage function. Annu Rev Immunol. 15:323–350. 1997. View Article : Google Scholar : PubMed/NCBI

5 

Jones BW, Heldwein KA, Means TK, Saukkonen JJ and Fenton MJ: Differential roles of toll-like receptors in the elicitation of proinflammatory responses by macrophages. Ann Rheum Dis. 60(Suppl 3): iii6–iii12. 2001.

6 

Jones BW, Means TK, Heldwein KA, Keen MA, Hill PJ, Belisle JT and Fenton MJ: Different toll-like receptor agonists induce distinct macrophage responses. J Leukoc Biol. 69:1036–1044. 2001.PubMed/NCBI

7 

Rhee SH and Hwang D: Murine TOLL-like receptor 4 confers lipopolysaccharide responsiveness as determined by activation of NF kappa B and expression of the inducible cyclooxygenase. J Biol Chem. 275:34035–34040. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Schroder K, Sweet MJ and Hume DA: Signal integration between IFNgamma and TLR signalling pathways in macrophages. Immunobiology. 211:511–524. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Stalińska K, Guzdek A, Rokicki M and Koj A: Transcription factors as targets of the anti-inflammatory treatment. A cell culture study with extracts from some mediterranean diet plants. J Physiol Pharmacol. 56(Suppl 1): S157–S169. 2005.

10 

Szabó C and Thiemermann C: Regulation of the expression of the inducible isoform of nitric oxide synthase. Adv Pharmacol. 34:113–153. 1995. View Article : Google Scholar : PubMed/NCBI

11 

Clancy RM, Amin AR and Abramson SB: The role of nitric oxide in inflammation and immunity. Arthritis Rheumatol. 41:1141–1151. 1998. View Article : Google Scholar

12 

Guadagni F, Ferroni P, Palmirotta R, Portarena I, Formica V and Roselli M: Review. TNF/VEGF cross-talk in chronic inflammation-related cancer initiation and progression: An early target in anticancer therapeutic strategy. In Vivo. 21:147–161. 2007.PubMed/NCBI

13 

Kröncke KD, Fehsel K and Kolb-Bachofen V: Inducible nitric oxide synthase in human diseases. Clin Exp Immunol. 113:147–156. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Nathan C and Xie QW: Nitric oxide synthases: Roles, tolls and controls. Cell. 78:915–918. 1994. View Article : Google Scholar : PubMed/NCBI

15 

Nathan C and Xie QW: Regulation of biosynthesis of nitric oxide. J Biol Chem. 269:13725–13728. 1994.PubMed/NCBI

16 

Nishimoto N and Kishimoto T: Interleukin 6: From bench to bedside. Nat Clin Pract Rheumatol. 2:619–626. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Calixto JB: Efficacy, safety, quality control, marketing and regulatory guidelines for herbal medicines (phytotherapeutic agents). Braz J Med Biol Res. 33:179–189. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Wutthithamavet W: Thai traditional medicine. revised edition. Odean Store Press; Bangkok: 1997

19 

Tewtrakul S, Wattanapiromsakul C and Mahabusarakam W: Effects of compounds from Garcinia mangostana on inflammatory mediators in RAW264.7 macrophage cells. J Ethnopharmacol. 121:379–382. 2009. View Article : Google Scholar

20 

Cui J, Hu W, Cai Z, Liu Y, Li S, Tao W and Xiang H: New medicinal properties of mangostins: Analgesic activity and pharmacological characterization of active ingredients from the fruit hull of Garcinia mangostana L. Pharmacol Biochem Behav. 95:166–172. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Duke JA: Handbook of medical herbs. Library of congress; New York: 1985

22 

Evan WC: Trease and Evans pharmacognosy. Saunders Company Limited; Nottingham: 1996

23 

Panthong A, Norkaew P, Kanjanapothi D, Taesotikul T, Anantachoke N and Reutrakul V: Anti-inflammatory, analgesic and antipyretic activities of the extract of gamboge from Garcinia hanburyi Hook f. J Ethnopharmacol. 111:335–340. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Guimarães CL, Otuki MF, Beirith A and Cabrini DA: A review on the therapeutic potential of Garcinia gardneriana. Dynamis. 12:6–12. 2004.In Portuguese.

25 

Castardo JC, Prudente AS, Ferreira J, Guimarães CL, Monache FD, Filho VC, Otuki MF and Cabrini DA: Anti-inflammatory effects of hydroalcoholic extract and two biflavonoids from Garcinia gardneriana leaves in mouse paw oedema. J Ethnopharmacol. 118:405–411. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Ayepola OR, Chegou NN, Brooks NL and Oguntibeju OO: Kolaviron, a Garcinia biflavonoid complex ameliorates hyperglycemia-mediated hepatic injury in rats via suppression of inflammatory responses. BMC Complement Altern Med. 13:3632013. View Article : Google Scholar : PubMed/NCBI

27 

Farombi EO, Adedara IA, Ajayi BO, Ayepola OR and Egbeme EE: Kolaviron, a natural antioxidant and anti-inflammatory phyto-chemical prevents dextran sulphate sodium-induced colitis in rats. Basic Clin Pharmacol Toxicol. 113:49–55. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Li F, Shanmugam MK, Chen L, Chatterjee S, Basha J, Kumar AP, Kundu TK and Sethi G: Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev Res (Phila). 6:843–854. 2013. View Article : Google Scholar

29 

Tsai ML, Chiou YS, Chiou LY, Ho CT and Pan MH: Garcinol suppresses inflammation-associated colon carcinogenesis in mice. Mol Nutr Food Res. 58:1820–1829. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Minami H, Takahashi E, Fukuyama Y, Kodama M, Yoshizawa T and Nakagawa K: Novel xanthones with superoxide scavenging activity from Garcinia subelliptica. Chem Pharm Bull (Tokyo). 43:347–349. 1995. View Article : Google Scholar

31 

Minami H, Hamaguchi K, Kubo M and Fukuyama Y: A benzophenone and a xanthone from Garcinia subelliptica. Phytochemistry. 49:1783–1785. 1998. View Article : Google Scholar

32 

Ito T, Yokota R, Watarai T, Mori K, Oyama M, Nagasawa H, Matsuda H and Iinuma M: Isolation of six isoprenylated biflavonoids from the leaves of Garcinia subelliptica. Chem Pharm Bull (Tokyo). 61:551–558. 2013. View Article : Google Scholar

33 

Weng JR, Tsao LT, Wang JP, Wu RR and Lin CN: Anti-inflammatory phloroglucinols and terpenoids from Garcinia subelliptica. J Nat Prod. 67:1796–1799. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Weng JR, Lin CN, Tsao LT and Wang JP: Terpenoids with a new skeleton and novel triterpenoids with anti-inflammatory effects from Garcinia subelliptica. Chemistry. 9:5520–5527. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Weng JR, Lin CN, Tsao LT and Wang JP: Novel and anti-inflammatory constituents of Garcinia subelliptica. Chemistry. 9:1958–1963. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCt method. Methods. 25:402–408. 2001. View Article : Google Scholar

37 

Koressaar T and Remm M: Enhancements and modifications of primer design program Primer3. Bioinformatics. 23:1289–91. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Samavati L, Rastogi R, Du W, Hüttemann M, Fite A and Franchi L: STAT3 tyrosine phosphorylation is critical for interleukin 1 beta and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol Immunol. 46:1867–1877. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Zhu ZG, Jin H, Yu PJ, Tian YX, Zhang JJ and Wu SG: Mollugin Inhibits the inflammatory response in lipopolysaccharide-stimulated RAW264.7 macrophages by blocking the Janus kinase-signal transducers and activators of transcription signaling pathway. Biol Pharm Bull. 36:399–406. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Lee C, Lim HK, Sakong J, Lee YS, Kim JR and Baek SH: Janus kinase-signal transducer and activator of transcription mediates phosphatidic acid-induced interleukin (IL)-1beta and IL-6 production. Mol Pharmacol. 69:1041–1047. 2006.

41 

Cario E, Rosenberg IM, Brandwein SL, Beck PL, Reinecker HC and Podolsky DK: Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing toll-like receptors. J Immunol. 164:966–972. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Burk DR, Senechal-Willis P, Lopez LC, Hogue BG and Daskalova SM: Suppression of lipopolysaccharide-induced inflammatory responses in RAW 264.7 murine macrophages by aqueous extract of Clinopodium vulgare L. (Lamiaceae). J Ethnopharmacol. 126:397–405. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Liu H, Bargouti M, Zughaier S, Zheng Z, Liu Y, Sangadala S, Boden SD and Titus L: Osteoinductive LIM mineralization protein-1 suppresses activation of NF-kappaB and selectively regulates MAPK pathways in pre-osteoclasts. Bone. 46:1328–1335. 2010. View Article : Google Scholar :

44 

Shan J, Fu J, Zhao Z, Kong X, Huang H, Luo L and Yin Z: Chlorogenic acid inhibits lipopolysaccharide-induced cyclo-oxygenase-2 expression in RAW264.7 cells through suppressing NF-kappaB and JNK/AP-1 activation. Int Immunopharmacol. 9:1042–1048. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Watters JJ, Sommer JA, Pfeiffer ZA, Prabhu U, Guerra AN and Bertics PJ: A differential role for the mitogen-activated protein kinases in lipopolysaccharide signaling: The MEK/ERK pathway is not essential for nitric oxide and interleukin 1beta production. J Biol Chem. 277:9077–9087. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Caivano M: Role of MAP kinase cascades in inducing arginine transporters and nitric oxide synthetase in RAW264 macrophages. FEBS Lett. 429:249–253. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Paul A, Cuenda A, Bryant CE, Murray J, Chilvers ER, Cohen P, Gould GW and Plevin R: Involvement of mitogen-activated protein kinase homologues in the regulation of lipopolysaccharide-mediated induction of cyclo-oxygenase-2 but not nitric oxide synthase in RAW 264.7 macrophages. Cell Signal. 11:491–497. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 13 Issue 3

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
Cho YC and Cho YC: c‑Jun N‑terminal kinase‑mediated anti‑inflammatory effects of Garcinia subelliptica in macrophages. Mol Med Rep 13: 2293-2300, 2016
APA
Cho, Y., & Cho, Y. (2016). c‑Jun N‑terminal kinase‑mediated anti‑inflammatory effects of Garcinia subelliptica in macrophages. Molecular Medicine Reports, 13, 2293-2300. https://doi.org/10.3892/mmr.2016.4791
MLA
Cho, Y., Cho, S."c‑Jun N‑terminal kinase‑mediated anti‑inflammatory effects of Garcinia subelliptica in macrophages". Molecular Medicine Reports 13.3 (2016): 2293-2300.
Chicago
Cho, Y., Cho, S."c‑Jun N‑terminal kinase‑mediated anti‑inflammatory effects of Garcinia subelliptica in macrophages". Molecular Medicine Reports 13, no. 3 (2016): 2293-2300. https://doi.org/10.3892/mmr.2016.4791