Research Article

Korean Journal of Plant Resources. 31 October 2018. 478-488
https://doi.org/10.7732/kjpr.2018.31.5.478

ABSTRACT


MAIN

  • Introduction

  • Materials and Methods

  •   ME preparation and preparation of extracts

  •   Cell lines

  •   Cell viability assay

  •   NO assay

  •   Total RNA extraction, cDNA synthesis, and reverse transcription polymerase chain reaction (RT-PCR) analysis

  •   Immunoblotting analysis

  •   Statistical analysis

  • Results

  •   Identification of mushroom extracts (MEs)

  •   Effects of MEs on LPS-induced cell proliferation in RAW264.7 cells

  •   Effects of MEs on LPS-stimulated NO production in RAW264.7 cells

  •   Effects of MEs on the LPS-induced increase in TNF-α, IL-6, and IL-1β mRNAs in RAW264.7 cells

  •   Effects of MEs on LPS-induced iNOS, COX-2, and p65 expression in RAW264.7 cells

  • Discussion

Introduction

Mushrooms have a long history of use as a part of the human diet in many regions worldwide due to their organoleptic characteristics and nutritional value (Reid et al., 2017). They have a high protein content with almost all essential amino acids and are rich in diverse minerals and vitamin B, representing a good dietary source of these important nutrients (Robaszkiewicz et al., 2010). In addition to attracting a great deal of interest in many areas of foods, mushrooms are also useful for preventing diseases, such as hypertension, hypercholesterolemia, and cancer (Lull et al., 2005). Medicinal mushrooms have an established history of use in traditional Eastern therapies. Based on historical customs of North Eastern Asia, medicinal mushrooms such as Ganoderma lucidum (Reishi), Lentinus edodes (Shiitake), and Inonotus obliquus (Chaga) have been collected and hot-water-soluble fractions have been extracted (Wasser, 2002). However, appropriate preparation methods for mushrooms in food processing and pharmaceutical production to regulate cellular immune responses are still elusive. Furthermore, the types of mushrooms that have greater biological and pharmaceutical activities for enhancing the immune response have not yet been identified.

Inflammation is a biological defense response of the body to harmful stimuli, including tissue damage or pathogen infection (Park et al., 2018); this response is required for removal of damaging agents. Chronic inflammation can be differentially classified from acute inflammation and is generally associated with the development of diseases, such as atherosclerosis, asthma, rheumatoid arthritis, and cancer (Libby et al., 2002; Du et al., 2015).

During the inflammatory response, inflammatory cells such as macrophages secrete inflammatory mediators including nitric oxide (NO), tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β (Mann et al., 2005). Lipopolysaccharide (LPS), a well-known endotoxin, activates the transcription factor nuclear factor-κB (NF-κB) and induces the expression of inflammatory cytokines, such as inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), resulting in the production of inflammatory mediators (Guzik et al., 2003; Yi et al., 2017). Currently, many anti-inflammatory drugs cause side effects such as gastritis, nephritis, and cardiac disorders. Accordingly, many studies have attempted to identify safer anti-inflammatory substances among commonly consumed dietary foods.

In this study, we aimed to investigate whether mushroom extracts possessed anti-inflammatory effects. Primarily, we considered that mushroom extracts should be easily prepared and ready to eat. For this purpose, the solvents were applied either water or ethanol. To this end, we examined the production of LPS-induced NO using RAW264.7 cells and evaluated the mRNA levels of cytokines and protein accumulation of iNOS and COX-2 following modification of NF-κB protein levels. Our data provide insights into the utility of mushroom extracts (MEs) in the regulation of immune responses and anti-inflammatory activities.

Materials and Methods

ME preparation and preparation of extracts

Mushrooms were purchased from Yeoncheon, South Korea in June, 2014. The dried fruiting body at 50℃ for 3 days of 22 species of mushroom was placed into a flask and extracted three times with 10 volume of 70% ethanol or water at room temperature for 1 day, and then filtered through a PTEE syringe. The filtrate was condensed by evaporation under reduced pressure at 50℃ and freeze-dried, and the weight was measured.

Cell lines

RAW264.7 macrophages were obtained from the Korean Cell Line Bank (KCLB NO. 40071, Seoul, Korea). The cells were grown in Dulbecco’s modified Eagle’s medium (Gibco Life Technologies, Carlsbad, CA, USA) containing 10% fetal bovine serum, 50 ㎍/㎖ penicillin, and 50 ㎍/㎖ streptomycin in a 5% CO chamber at 37℃.

Cell viability assay

RAW264.7 cells were seeded in 96-well plates at a density of 1 × 104 cells/well. The cultured cells were treated with 1 ㎍/㎖ LPS for 4 h and MEs were then added in dimethylsulfoxide (DMSO) at various concentrations. Solvent control cells were treated with LPS alone. After 24 h of culture with or without samples, MTS working solution (Promega, CA, USA) was added into the culture plates for 4 h at 37℃. The absorbance of individual wells was measured at 540 nm using a microplate reader (Molecular Devices, CA, USA), and cell viability was calculated as the percent absorbance relative to LPS-treated control cells.

NO assay

The level of NO production in RAW264.7 cell supernatants was determined using Griess reagent (Promega), according to the manufacturer’s instructions. Briefly, the extracts were treated as described for the cell viability assays, and the cell culture supernatants were then collected in 96-well plates for nitrite assays. The absorbance was measured at 540 nm using a microplate reader (Molecular Devices, CA, USA). The concentration of nitrite was calculated from a standard curve.

Total RNA extraction, cDNA synthesis, and reverse transcription polymerase chain reaction (RT-PCR) analysis

Total RNA was isolated from RAW264.7 cells using TRIzol Reagent (Invitrogen, NY, USA), and 1 ㎍ of RNA was used for cDNA synthesis. RT-PCR was performed where each cDNA was amplified using gene-specific primers (Table 1) and a Maxime PCR RedMix kit (iNtRON Biotechnology, Seongnam, Korea). GADPH expression was used to normalize the expression value in each sample and relative expression values were determined against the average value of LPS-treated samples without MEs using ImageJ software.

Table 1. The pro-inflammatory gene primer used for RT-PCR

GeneSense and antisense sequencesExpected product size (bp)
IL-65'-GTACTCCAGAAGACCAGAGG-3'308
5'-TGCTGGTGACAACCACGGCC-3'
TNF-α5'-TTGACCTCAGCGCTGAGTTG-3'364
5'-CCTGTAGCCCACGTCGTAGC-3'
IL-1β5'-CAGGATGAGGACATGAGCACC-3'447
5'-CTCTGCAGACTCAAACTCCAC-3'
GAPDH5'-CGGAGTCAACGGATTTGGTCGTAT-3'306
5'-AGCCTTCTCCATGGTGGTGAAGAC-3'

Immunoblotting analysis

RAW264.7 cells were collected and lysed in ice-cold RIPA buffer (20 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM Na2-ethylenediamintetraacetic acid, 1 mM ethylene glycol-bis[β-aminoethyl ether]-N,N,N',N'-tetraacetic acid, 1% NP-40, 1% sodium deoxycholate, 2.5 mM sodium pyrophosphate, 1 mM β-glycerophosphate, 1 mM Na3VO4, and 1 ㎍/㎖ leupeptin) for 30 min, and protein levels were measured using the BCA assay (Thermo Scientific, Rockford, IL, USA). The extracted protein was separated using 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then transferred onto nitrocellulose membranes (GE Healthcare, Chalfont St. Giles, UK). After blocking with Tris-buffered saline containing 5% nonfat dry milk and 0.1% (w/v) Tween 20, the membranes were immunoblotted with specific antibodies and then subjected to enhanced chemiluminescence (ECL kit; GE Healthcare). The relative band intensities were determined using chemiluminescence (Davinch-chemi Chemiluminescence Imaging System; CAS-400SM; Seoul, Korea). The relative protein intensity of each sample (200 ㎍/㎖) was calculated by normalizing against the intensity of β-actin as a loading control.

Statistical analysis

Unless otherwise indicated, values represent the mean ± standard deviation (SD) of data obtained from triplicate experiments. Statistical analysis was performed using Student’s t-tests.

Results

Identification of mushroom extracts (MEs)

To obtain MEs, either water or 70% ethanol, herein referred to simply as ethanol, was used as the extraction solvent based on the consideration that MEs should be directly edible. We chose the dried fruiting body of the mushrooms, and extraction was performed at room temperature for 1 day. The MEs are listed in Table 2.

Table 2. Relative effects of mushroom extracts on LPS-treated RAW264.7 cells

Extracted
Solvent
MushroomThe rate of
cell growth
change
NO
contents
(μM)
The rate of
NO change
WaterPleurotus eryngii (DC.) Quél. 0.928 26.45±1.430.926
Tricholoma giganteum Massee1.018 26.25±0.520.988
Tricholoma matsutake0.902 26.26±1.700.879
Sparassis crispa (Wulfen) Fr. 0.951 28.95±2.361.052
Agaricus bisporus (J.E. Lange) Imbach1.051 26.21±1.200.993
Pleurotus cornucopiae (paulet) Rolland var,
citrinopileatus (Sing.) Ohira
0.942 3.75±0.690.134
Pleurotus ostreatus (Jacq.: Fr.) P. Kumm1.096 27.86±0.150.985
Pleurotus salmoneostramineus Lj.N. Vassiljeva1.206 25.48±1.960.975
Hypsizygus marmoreus (Peck) H.E. Bigelow1.064 29.12±3.441.023
Isaria japonica Yasuda0.8494.25±0.740.258
Inonotus obliquus (Fr.) Pilat1.023 26.68±0.760.930
Lentinula edodes (Berk.) Pegler0.989 9.92±0.700.338
Ganoderma lucidum (Curtis) P. Karst. 0.953 0.17±0.360.006
Antler-Ganoderma lucidum (Curtis) P. Karst. 1.104 26.47±1.170.940
Flat Ganoderma lucidum (Curtis) P. Karst. 1.208 28.20±1.191.001
Grifola frondosa (Dicks.) Gray 1.017 16.74±0.650.596
Flammulina velutipes (Curtis) Singer1.285 26.35±2.450.880
Hypsizygus marmoreus (Peck) H.E. Bigelow0.941 21.02±1.890.818
Gold-Hypsizygus marmoreus (Peck) H.E. Bigelow0.956 26.17±2.220.976
EthanolPleurotus eryngii (DC.) Quél. 1.082 17.68±2.500.635
Tricholoma giganteum Massee0.928 18.40±2.630.652
Tricholoma matsutake1.370 17.28±1.180.658
Sparassis crispa (Wulfen) Fr. 1.384 15.21±0.990.597
Agaricus bisporus (J.E. Lange) Imbach0.897 17.22±1.170.642
Pleurotus ostreatus (Jacq.: Fr.) P. Kumm1.307 17.31±1.030.657
Pleurotus salmoneostramineus L. Vass0.900 14.18±1.040.540
Hypsizygus marmoreus (Peck) H.E. Bigelow1.379 13.77±1.550.554
Isaria japonica Yasuda0.979 0.035±0.120.008
Inonotus obliquus (Fr.) Pilat0.998 2.17±0.350.219
Lentinula edodes (Berk.) Pegler1.452 15.68±1.790.593
Ganoderma lucidum (Curtis) P. Karst. 0.997 4.96±1.220.201
Antler-Ganoderma lucidum (Curtis) P. Karst. 1.114 17.30±0.880.648
Grifola frondosa (Dicks.) Gray 0.737 15.40±1.190.582
Phellinus linteus (Berk. Et Curt.) Teng 1.211 0.05±0.140.013
Umbilicaria esculenta (Miyoshi) Minks0.9522.00±1.100.119
Cordyceps militaris1.246 18.77±1.000.716
DMSO1.000 30.68±2.171.000

Effects of MEs on LPS-induced cell proliferation in RAW264.7 cells

To examine the effects of cell viability in the presence of MEs, we performed MTS assays using RAW264.7 cells, which were derived from mouse leukemic monocyte macrophages. The MTS assay is commonly used for analyze cell cytotoxicity and its result is correlated with cell proliferation. Cell proliferation was not changed by LPS (Table 3) or ME treatment in the absence of LPS (data not shown). LPS-induced cell proliferation was not altered by around 36 different spices of MEs (Table 2, Table 3). However, some MEs caused reduced cell proliferation under the LPS treatment. For example, Isaria japonica Yasuda (I. japonica) and Inonotus obliquus (Fr.) Pilát (I. obliquus) ethanol extracts showed only 53.64% ± 9.10% and 58.66% ± 2.76% cell proliferation, respectively, relative to the control, indicating that these MEs may exhibit cytotoxicity in RAW264.7 cells (Table 3). Indeed, even when using a relatively low concentration of 100 ㎍/㎖ I japonica and I. obliquus MEs, cell growth reached only 54.81% ± 6.77% and 58.77% ± 5.35%, respectively (Table 3). Thus, although some MEs affected cell proliferation, most MEs yielded 100% or higher viability, indicating that these MEs did not exert cytotoxic effects in RAW264.7 cells.

Table 3. Table 3. The mushroom extracts effect of cell proliferation and NO release content

Exracted SolventMushroom Cell proliferation
(% of LPS-treated control)
NO contents
(μM)
100 μg/ml200 μg/ml100 μg/ml200 μg/ml
WaterPleurotus eryngii (DC.) Quél. 103.3 ±10.2995.90±12.7628.56±0.2826.45±1.43
Tricholoma giganteum Massee106.88±13.17108.84±3.6826.56±1.3726.25±0.52
Tricholoma matsutake105.69±13.6195.37±8.2229.88±1.2926.26±1.70
Sparassis crispa (Wulfen) Fr. 91.65±3.4787.12±10.3227.51±1.0528.95±2.36
Agaricus bisporus (J.E. Lange) Imbach89.74±8.9294.33±17.9026.39±1.0326.21±1.20
Pleurotus cornucopiae (paulet)
Rolland var, citrinopileatus (Sing.)
Ohira
80.68±3.2076.04±4.0028.08±1.353.75±0.69
Pleurotus ostreatus (Jacq.: Fr.)
P. Kumm
88.94±12.9097.45±7.4328.28±3.3727.86±0.15
Pleurotus salmoneostramineus Lj.N.
Vassiljeva
93.77±11.56113.09±7.5126.12±0.3525.48±1.96
Hypsizygus marmoreus (Peck) H.E.
Bigelow
89.57±3.9395.27±8.6028.46±0.2729.12±3.44
Isaria japonica Yasuda82.01±7.8181.13±9.3416.45±3.154.25±0.74
Inontus obliquus79.61±10.3581.46±5.0928.69±1.5526.68±0.76
Lentinula edodes (Berk.) Pegler87.54±10.5286.59±5.5729.32±0.939.92±0.70
Ganoderma lucidum (Curtis) P. Karst. 91.74±12.9987.39±3.9527.12±1.480.17±0.36
Antler-Ganoderma lucidum (Curtis)
P. Karst.
107.82±17.47118.98±7.9028.16±0.7726.47±1.17
Flat Ganoderma lucidum (Curtis)
P. Karst.
94.81±8.64114.57±10.3928.18±1.1428.20±1.19
Grifola frondosa (Dicks.) Gray 105.27±9.67107.03±5.8128.10±1.2016.74±0.65
Flammulina velutipes (Curtis) Singer90.78±15.24116.67±10.1029.93±0.7426.35±2.45
Hypsizygus marmoreus (Peck)
H.E. Bigelow
88.41±6.4483.16±13.4225.70±1.0021.02±1.89
Gold-Hypsizygus marmoreus (Peck)
H.E. Bigelow
93.31±5.5189.21±11.7026.82±0.8126.17±2.22
EthanolPleurotus eryngii (DC.) Quél. 91.74±3.4399.29±10.7227.86±0.1517.68±2.50
Tricholoma giganteum Massee124.06±18.08115.18±12.3328.20±1.1918.40±2.63
Tricholoma matsutake97.02±8.68132.92±8.8326.28±0.8617.28±1.18
Sparassis crispa (Wulfen) Fr. 95.45±27.14132.11±8.2125.48±1.9615.21±0.99
Agaricus bisporus (J.E. Lange) Imbach106.27±6.6895.37±8.2226.84±0.8117.22±1.17
Pleurotus ostreatus (Jacq.: Fr.)
P. Kumm
101.21±2.53132.33±9.1126.35±2.4517.31±1.03
Pleurotus salmoneostramineus L. Vass130.85±11.59117.76±20.2626.26±1.7014.18±1.04
Hypsizygus marmoreus (Peck)
H.E. Bigelow
97.59±5.22134.57±10.2724.86±1.3213.77±1.55
Isaria japonica Yasuda54.81±6.7753.64±10.504.25±0.740.035±0.12
Inontus obliquus58.77±5.3558.66±3.199.92±0.702.17±0.35
Lentinula edodes (Berk.) Pegler94.09±10.98136.6±11.1726.45±1.4315.68±1.79
Ganoderma lucidum (Curtis) P. Karst. 85.08±11.0984.80±10.3824.65±0.214.96±1.22
Antler-Ganoderma lucidum (Curtis)
P. Karst.
92.71±6.47103.26±13.3726.68±0.7617.30±0.88
Grifola frondosa (Dicks.) Gray 113.86±10.3983.90±3.0326.47±1.1715.40±1.19
Phellinus linteus (Berk. Et Curt.) Teng 54.30±4.8165.78±9.883.75±0.690.05±0.14
Umbilicaria esculenta (Miyoshi) Minks102.38±10.2897.48±5.1916.74±0.652.00±1.10
Cordyceps militaris101.65±8.48126.62±16.7526.21±1.2018.77±1.00
-LPS103.01±2.492.17±0.02
1μg/mlLPS100±0030.68±2.17

Effects of MEs on LPS-stimulated NO production in RAW264.7 cells

To observe LPS-induced NO production in RAW264.7 cells in the presence of MEs, cells were treated with LPS (1 ㎍/㎖) for 4 h and then treated with DMSO or various MEs for 24 h. After 24 h of incubation, the NO levels in the cellular supernatants were assessed by Griess assays. Treatment with LPS increased NO production (30.68 ± 2.17 μM) compared with that in the absence of LPS (2.17 ± 0.02 μM; Table 2, Table 3). In contrast, post-treatment with 200 ㎍/㎖ MEs resulted in reduced LPS-induced NO generation (Table 3). Specifically, MEs prepared using Pleurotus cornucopiae (paulet) Rolland var. citrinopileatus (Sing.) Ohira, and I japonica extracts with water as well as those prepared using I. obliquus t and U. esculenta extracts with ethanol reduced NO contents to 3.75 ± 0.69 μM, 4.25 ± 0.74 μM, and 2.17 ± 0.35 μM, 2.00 ± 1.10 μM, respectively, similar to that without LPS (2.17 ± 0.02 μM; Table 3). We further tested NO production in the presence of 100 ㎍/㎖ MEs to investigate the effects of a lower concentration of MEs. Notably, 100 ㎍/㎖ MEs yielded results similar to those above, except that the NO concentration was higher than that induced by 200 ㎍/㎖ MEs (Table 3), indicating that LPS-induced NO contents were reduced in an ME concentration-dependent manner. Statistically significant differences were calculated by Student’s t-test, p < 0.05 (*), p < 0.01(**), and p < 0.001 (***). As NO is a signaling molecule involved in acute and chronic inflammatory diseases, NO levels are used to be representing inflammation. These results suggest that extracts from several mushrooms including I japonica, I. obliquus, U. esculenta, and P. linteus exerted anti-inflammatory effects via reduction of NO in LPS-stimulated RAW264.7 cells.

Before further investigating the anti-inflammatory effects of MEs, we summarized the above results for cell proliferation and NO reduction. The values were obtained by comparing the changes in cell proliferation and NO contents between 200 ㎍/㎖ MEs and 100 ㎍/㎖ MEs (Table 2). Thus, we chose two ME candidates: I. japonica water extract and U. esculenta ethanol extract due to that the change rate of cell growth was close to 1 for no change (i.e less cytotoxic), and the change rate of NO content was around 0.2 for 80% reduction on the treatment of 200 ㎍/㎖ MEs (i.e effective for NO reduction) (Table 2).

Effects of MEs on the LPS-induced increase in TNF-α, IL-6, and IL-1β mRNAs in RAW264.7 cells

To evaluate the anti-inflammatory effects of these two MEs, we analyzed the expression of inflammatory marker genes by reverse transcription PCR. Because murine macrophages produce pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, during the inflammatory response, LPS treatment increases the these transcripts (Mann et al., 2005). Given the screening results for the chosen two MEs, RAW264.7 cells were pre-incubated for 4 h with 1 ㎍/㎖ LPS and cultured 24 h more with the indicated concentrations (50–200 ㎍/㎖) of MEs. LPS treatment induced TNF-α, IL-6, and IL-1β gene expression. However, LPS-induced accumulation of TNF-α, IL-6, and IL-1β mRNA was significantly reduced after treatment with both I. japonica extracts (Fig. 1) and U. esculenta extracts (Fig. 2). In addition, reduction of the these transcripts was much more apparent following treatment with the high concentration (200 ㎍/㎖) of MEs compared with that after treatment with a low concentration (50 ㎍/㎖) of MEs (Fig. 3B–D and Fig. 4B–D). Statistically, the values revealed that the difference were significant with p < 0.05 (*), p < 0.01(**), and p < 0.001(***) by Student’s t-test. This result indicated that both I. japonica and U. esculenta extracts suppressed LPS-mediated transcription of pro-inflammatory cytokines.

http://static.apub.kr/journalsite/sites/kjpr/2018-031-05/N0820310505/images/kjpr_31_05_05_F1.jpg
Fig. 1.

Effects of water extracts of I. japonica on pro-inflammatory gene expression in RAW264.7 cells. (A) The expression levels of TNF-α, IL-6, and IL-1β mRNAs were determined by RT-PCR analysis. Cells were treated with 1 μg/mL LPS and then I. japonica extracts at various concentrations for 24 h. GADPH was used as an internal control. The expression levels of TNF-α (B), IL-6 (C), and IL-1β (D) were determined by densitometry analysis. Values are expressed as the mean ± SD of four determinations. Significance was determined using Student’s t-tests: *p < 0.05, **p < 0.01, and ***p < 0.001 versus cells treated with LPS alone.

http://static.apub.kr/journalsite/sites/kjpr/2018-031-05/N0820310505/images/kjpr_31_05_05_F2.jpg
Fig. 2.

Effects of ethanol extract of U. esculenta on pro-inflammatory gene expression in RAW264.7 cells. (A) The expression levels of TNF-α, IL-6, and IL-1β mRNA was determined by RT-PCR analysis. Cells were treated with 1 ㎍/㎖ LPS and then U. esculenta extracts at various concentrations for 24 h. GADPH was used as an internal control. The expression levels of TNF-α (B), IL-6 (C), and IL-1β (D) were determined by densitometry analysis. Values are represented as the mean ± SD of four determinations. Significance was determined using Student’s t-tests: *p < 0.05, **p < 0.01, and ***p < 0.001 versus cells treated with LPS alone.

Effects of MEs on LPS-induced iNOS, COX-2, and p65 expression in RAW264.7 cells

Next, we examined whether I. japonica and U. esculenta extracts suppressed the expression of transcription factors related to the inflammatory process, e.g., iNOS and COX-2. iNOS and COX-2 are key pro-inflammatory proteins, and the expression of these proteins is controlled by signaling downstream of NF-κB (T and A, 2006). To test this hypothesis, we investigated iNOS and COX-2 protein expression and p65 protein modification by western blotting following treatment with MEs. p65, a subunit of NF-κB, undergoes phosphorylation (p-P65) to activate gene expression, including COX-2 and iNOS genes (T and A, 2006).

LPS treatment increased iNOS and COX-2 expression by inducing the inflammatory response in RAW264.7 cells. As expected, immunoblotting analyses indicated that iNOS and COX-2 expression and post-translation modification of p65 by LPS pretreatment were inhibited after incubation with I. japonica (Fig. 3) and U. esculenta (Fig. 4) MEs. In addition, p65 protein abundance was slightly increased by LPS, whereas p65 protein phosphorylation was dramatically decreased (Fig. 3D and Fig. 4D). These results demonstrated that both I. japonica and U. esculenta extracts suppress LPS-stimulated p65 phosphorylation and affected COX-2 and iNOS protein abundance. Because the pro-inflammatory enzyme iNOS is responsible for NO production, inhibition of iNOS protein accumulation resulted in reduced NO content following treatment with I. japonica and U. esculenta extracts.

http://static.apub.kr/journalsite/sites/kjpr/2018-031-05/N0820310505/images/kjpr_31_05_05_F3.jpg
Fig. 3.

Effects of water extracts of I. japonica on the NF-κB pathway in RAW264.7 cells. (A) COX-2, iNOS, and p65 protein abundance was determined by a western blotting. Relative protein abundance of IL-6 (B) and IL-1β (C) and the level of Phosphorylated p65 (D), and were determined by densitometry analysis. Values are expressed the mean ± SD of four determinations. Significance was determined using Student’s t-tests: *p < 0.05, **p < 0.01, and ***p < 0.001 versus cells treated with LPS alone.

http://static.apub.kr/journalsite/sites/kjpr/2018-031-05/N0820310505/images/kjpr_31_05_05_F4.jpg
Fig. 4.

Effects of ethanol extracts of U. esculenta on the NF-κB pathway in RAW264.7 cells. (A) COX-2, iNOS, and p65 protein levels were determined by western blot analysis. IL-6 (B) and IL-1β (C) protein levels were determined by densitometry analysis. Phosphorylated p65 levels were determined (D). Values are expressed as the mean ± SD of four determinations. Significance was determined using one-factor t-tests: *p < 0.05, **p < 0.01, and ***p < 0.001 versus cells treated with LPS alone.

Discussion

Macrophages play important roles in inflammation through the production of several pro-inflammatory molecules, including NO. Uncontrolled, excessive NO has been shown to be associated with a range of inflammatory diseases, including arteriosclerosis, ischemic reperfusion, hypertension, and septic shock (Alvarez and Evelson, 2007; Pacher et al., 2007). Recent studies have demonstrated that plant foods, including fruits, vegetables, and medicinal herbs, are excellent sources of antioxidant molecules that effectively inhibit the inflammatory process by influencing different molecular targets (Tsai et al., 2007).

Edible mushrooms are used as functional foods due to their high content of bioactive compounds. However, their anti-inflammatory properties and the effects of processing steps on their bioactivity have not been systemically investigated. The anti-inflammatory effect of mushrooms of water extracts were reported using Sparassis cispa (Wulfen) Fr. (Kim et al., 2012), Ganoderma lucidum (Curtis) P. Karst (Kuo et al., 2006) and Grifola frondosa (Dicks.) Gray (Radic et al., 2010). The anti-inflammatory effect of mushrooms of ethanol extracts were reported Pleurotus eryngii (DC.) Quél (Lin et al., 2014), Agaricus bisporus (J.E. Lange) Imbach (Gunawardena et al., 2014; Taofiq et al., 2015), Pleurotus ostreatus (Jacq.: Fr.) P. Kumm (Taofiq et al., 2015), Hypsizygus marmoreus (Peck) H.E. Bigelow (Chien et al., 2016), Inonotus obliquus (Fr.) Pilat (Park et al., 2005; Van et al., 2009; Ma et al., 2013), Lentinula edodes (Berk.) Pegler (Gunawardena et al., 2014), Ganoderma lucidum (Curtis) P. Karst (Yoon et al., 2013). Accordingly, in this study, we found that MEs effectively suppressed LPS-induced inflammation. In addition, we showed that water extracts of I. japonica and ethanol extracts of U. esculenta inhibited the LPS-induced increase in NO production and pro-inflammatory cytokine expression in RAW264.7 cells.

LPS, a component of the outer membrane of gram-negative bacteria, activates macrophages and promotes the synthesis and release of molecules involved in the inflammatory response, such as cytokines, NO, and pro-inflammatory enzymes (O'Connellet al., 1998). We observed that MEs prepared using water or ethanol effectively reduced LPS- induced NO accumulation in RAW264.7 macrophages, suggesting that the MEs mitigated the pro-inflammatory reaction. Accumulating evidence has shown that NO is directly involved in the development of inflammation (Aktan, 2004; Pacher et al., 2007). In particular, high concentrations of NO, synthesized by iNOS, can mediate inflammation and cause cell death by triggering apoptosis (Wink and Mitchell, 1998).

Interestingly, both water extracts of I. japonica and ethanol extracts of U.esculenta elicited changes in TNF-α, IL-1β, and IL-6 mRNA expression and in accumulation of iNOS and COX-2 enzymes, and they modulated p65 phosphorylation, suggesting that these MEs could inhibit the expression of the pro-inflammatory cytokines and modulate NF-κB signaling to control NO. Therefore, the findings of our study reveal that water extracts of I. japonica and ethanol extracts of U. esculenta possess a protective effect in LPS-induced RAW264.7 cells. In addition, these results support that mushrooms are healthy foods and sources of medicinal components. One benefit of these MEs is that they can be prepared easily, and this benefit might contribute to developing healthy beverages or drugs to prevent or treat inflammatory diseases.

Acknowledgements

This study was performed with the support of the Cooperative Research Program for Agriculture Science and Technology Development (project no. PJ01134401), the Rural Development Administration, Republic of Korea.

References

1
Aktan, F. 2004. iNOS-mediated nitric oxide production and its regulation. Life Sci. 75:639-653.
10.1016/j.lfs.2003.10.04215172174
2
Alvarez, S. and P.A. Evelson. 2007. Nitric oxide and oxygen metabolism in inflammatory conditions: sepsis and exposition to polluted ambients. Front Biosci. 12:964-974.
10.2741/211717127352
3
Chien, R.C., Y.C. Yang, E.I. Lai and J.L. Mau. 2016. Anti- inflammatory effects of extracts from the medicinal mushrooms Hypsizygus marmoreus and Pleurotus eryngii (Agaricomycetes). International Journal of Medicinal Mushrooms 18:477-487.
10.1615/IntJMedMushrooms.v18.i6.20
4
Du, C., M. Bhatia, S.C. Tang, M. Zhang and T. Steiner. 2015. Mediators of inflammation: Inflammation in cancer, chronic diseases, and wound healing. Mediators Inflamm. 2015:570653.
10.1155/2015/57065326549940PMC4624916
5
Gunawardena, D., L. Bennett, K. Shanmugam, K. King, R. Williams, D. Zabaras, R. Head, L. Ooi, E. Gyengesi and G. Münch. 2014. Anti-inflammatory effects of five commercially available mushroom species determined in lipopolysaccharide and interferon-γ activated murine macrophages. Food Chemistry 148:92-96.
10.1016/j.foodchem.2013.10.01524262531
6
Guzik, T.J., R. Korbut and T. Adamek-Guzik. 2003. Nitric oxide and superoxide in inflammation and immune regulation. J Physiol Pharmacol. 54:469-487.PMid:14726604
7
Kim, H.H., S.Y. Lee, T.S. Singh, J.K. Choi, T.Y. Shin and S.H. Kim. 2012. Sparassis crispa suppresses mast cell-mediated allergic inflammation: Role of calcium, mitogen-activated protein kinase and nuclear factor-κB. International Journal of Molecular Medicine 30:344-350.
10.3892/ijmm.2012.100022614038
8
Kuo, M.C., C.Y. Weng, C.L. Ha and M.J. Wu. 2006. Ganoderma lucidum mycelia enhance innate immunity by activating NF-κB. Journal of Ethnopharmacology 103:217-222.
10.1016/j.jep.2005.08.01016169168
9
Libby, P., P.M. Ridker and A. Maseri. 2002. Inflammation and atherosclerosis. Circulation 105:1135-1143.
10.1161/hc0902.10435311877368
10
Lin, J.T., C.W. Liu, Y.C. Chen, C.C. Hu, L.D. Juang, C.C. Shiesh and D.J. Yang. 2014. Chemical composition, antioxidant and anti-inflammatory properties for ethanolic extracts from Pleurotus eryngii fruiting bodies harvested at different time. LWT-Food Science and Technology 55:374-382.
10.1016/j.lwt.2013.08.023
11
Lull, C., H.J. Wichers and H.F. Savelkoul. 2005. Antiinflammatory and immunomodulating properties of fungal metabolites. Mediators Inflamm. 2005:63-80.
10.1155/MI.2005.6316030389PMC1160565
12
Ma, L., H. Chen, P. Dong and X. Lu. 2013. Anti-inflammatory and anticancer activities of extracts and compounds from the mushroom Inonotus obliquus. Food Chemistry 139:503-508.
10.1016/j.foodchem.2013.01.03023561137
13
Mann, J.R., M.G. Backlund and R.N. DuBois. 2005. Mechanisms of disease: Inflammatory mediators and cancer prevention. Nat Clin Pract Oncol. 2:202-210.
10.1038/ncponc014016264935
14
O'Connell, M.A., B.L. Bennett, F. Mercurio, A.M. Manning and N. Mackman. 1998. Role of IKK1 and IKK2 in lipopolysaccharide signaling in human monocytic cells. J Biol Chem. 273:30410- 30414.
10.1074/jbc.273.46.304109804806
15
Pacher, P., J.S. Beckman and L. Liaudet. 2007. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 87:315-424.
10.1152/physrev.00029.200617237348PMC2248324
16
Park, S.B., H.M. Song, H.N. Kim, G.H. Park, H.J. Son, Y. Um, J.A. Park and J.B. Jeong. 2018. Anti-inflammatory effect of Biji (Soybean curd residue) on LPS-stimulated RAW264.7 Cells. Korean J. Plant Res. 31:117-123.
17
Park, Y.M., J.H. Won, Y.H. Kim, J.W. Choi, H.J. Park and K.T. Lee. 2005. In vivo and in vitro anti-inflammatory and anti- nociceptive effects of the methanol extract of Inonotus obliquus. Journal of Ethnopharmacology 101:120-128.
10.1016/j.jep.2005.04.00315905055
18
Radic, N., Z. Jevnikar, N. Obermajer, J. Kristl, J. Kos, F. Pohleven and B. Strukelj. 2010. Infuence of culinary- medicinal Maitake mushroom, Grifola frondosa (Dicks.: Fr.) SF Gray (Aphyllophoromycetideae) polysaccharides on gene expression in Jurkat T lymphocytes. International Journal of Medicinal Mushrooms 12:245-255.
10.1615/IntJMedMushr.v12.i3.30
19
Reid, T., M. Munyanyi and T. Mduluza. 2017. Effect of cooking and preservation on nutritional and phytochemical composition of the mushroom Amanita zambiana. Food Sci Nutr. 5:538- 544.
10.1002/fsn3.42828572939PMC5448345
20
Robaszkiewicz, A., G. Bartosz, M. Lawrynowicz and M. Soszynski. 2010. The role of polyphenols, beta-carotene, and lycopene in the antioxidative action of the extracts of dried, edible mushrooms. J Nutr Metab. 2010:173274.
10.1155/2010/17327421234347PMC3017951
21
Trianthi, P. and A. Aggarwal. 2006. NF-kB transcription factor: a key player in the generation of immune response. Current Science 90:519-531.
22
Taofiq, O., R.C. Calhelha, S. Heleno, L. Barros, A. Martins, C. Santos-Buelga, M.J.R. Queiroz and I.C. Ferreira. 2015. The contribution of phenolic acids to the anti-inflammatory activity of mushrooms: Screening in phenolic extracts, individual parent molecules and synthesized glucuronated and methylated derivatives. Food Research International. 76:821-827.
10.1016/j.foodres.2015.07.04428455068
23
Tsai, P.J., T.H. Tsai, C.H. Yu and S.C. Ho. 2007. Evaluation of NO-suppressing activity of several Mediterranean culinary spices. Food Chem Toxicol. 45:440-447.
10.1016/j.fct.2006.09.00617074427
24
Van, Q., B. Nayak, M. Reimer, P. Jones, R. Fulcher and C. Rempel. 2009. Anti-inflammatory effect of Inonotus obliquus, Polygala senega L., and Viburnum trilobum in a cell screening assay. Journal of Ethnopharmacology 125:487-493.
10.1016/j.jep.2009.06.02619577624
25
Wasser, S.P. 2002. Medicinal mushrooms as a source of antitumor and immunomodulating polysaccharides. Appl Microbiol Biotechnol. 60:258-274.
10.1007/s00253-002-1076-712436306
26
Wink, D.A. and J.B. Mitchell. 1998. Chemical biology of nitric oxide: Insights into regulatory, cytotoxic, and cytoprotective mechanisms of nitric oxide. Free Radic Biol Med. 25:434-456.
10.1016/S0891-5849(98)00092-6
27
Yi, M., C. Kang and H. Bu. 2017. Anti-inflammatory and tyrosinase inhibition effects of amaranth (Amaranthus spp. L.) seed extract. Korean J. Plant Res. 30:144-151.
10.7732/kjpr.2017.30.2.144
28
Yoon, H.M., K.J. Jang, M.S. Han, J.W. Jeong, G.Y. Kim, J.H. Lee and Y.H. Choi. 2013. Ganoderma lucidum ethanol extract inhibits the inflammatory response by suppressing the NF-κB and toll-like receptor pathways in lipopolysaccharide- stimulated BV2 microglial cells. Experimental and Therapeutic Medicine 5:957-963.
10.3892/etm.2013.89523408713PMC3570243
페이지 상단으로 이동하기