Open Access

Protective effect of Gloeostereum incarnatum on ulcerative colitis via modulation of Nrf2/NF‑κB signaling in C57BL/6 mice

  • Authors:
    • Xiao Li
    • Xin Liu
    • Yongfeng Zhang
    • Yaqin Zhang
    • Shuyan Liu
    • Nan Zhang
    • Yu Li
    • Di Wang
  • View Affiliations

  • Published online on: August 5, 2020     https://doi.org/10.3892/mmr.2020.11420
  • Pages: 3418-3428
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chronic non‑specific inflammatory cell infiltration of the colon is generally considered to be the cause of ulcerative colitis (UC). Gloeostereum incarnatum (GI), a fungus rich in amino acids and fatty acids, exhibits a variety of biological functions. In the present study, GI was identified to contain 15 fatty acids, 17 amino acids and 11 metallic elements. The protective effect of GI against UC was investigated in C57BL/6 mice with UC induced by free drinking 3.5% dextran sulfate sodium (DSS). After a 21‑day oral administration, GI prevented weight loss, enhancement of the disease activity index and colonic pathological alterations in mice with UC. GI reduced the levels of pro‑inflammatory factors including interleukin (IL)‑1β, IL‑2, IL‑6 and IL‑12, tumor necrosis factor α and ‑β, interferon α and ‑γ, and pro‑oxidative factors including reactive oxygen species and nitric oxide. In addition, it enhanced the levels of immunological factors including immunoglobulin (Ig)A, IgM and IgG, and antioxidative factors including superoxide dismutase and catalase in the serum and/or colon tissues. GI enhanced the expression levels of nuclear factor erythroid 2‑related factor 2 (Nrf2) and its downstream proteins and suppressed the phosphorylation of NF‑κB signaling in colon tissues. Together, GI was shown to alleviate the physiological and pathological state of DSS‑induced UC in mice via its antioxidant and anti‑inflammatory functions, which may be associated with its modulation of the activation of Nrf2/NF‑κB signaling.

Introduction

Inflammatory bowel disease is a chronic, repetitive and non-specific gastrointestinal inflammatory disease comprising two conditions: Ulcerative colitis (UC) and Crohn's disease (1). As a chronic intestinal disease, UC produces inflammatory reactions and immune response at the colonic mucosa (2), accompanied by weight loss, diarrhea and bloody stools (3). Long-term severe colitis is known to cause colorectal cancer (4).

The pathogenesis of UC involves tissue damage in the colon and deterioration of the digestive and absorptive functions (5). Inflammation of the colon is due to an imbalance between the expression of pro-inflammatory and anti-inflammatory factors, and the migration of neutrophils to the damaged site, leading to the accumulation of inflammatory factors (6). Interleukin (IL)-6, closely associated with the differentiation and maturation of B cells, promotes the function of pro-inflammatory cells and inhibits the regulation of immune-related cells including T cells (7), both of which increase the levels of pro-inflammatory factors including IL-1 and IL-17 (8). Therefore, blocking the expression of pro-inflammatory cytokines may be a method for the potential treatment of UC (9). NF-κB inhibitor α is involved in the regulation of the development of early inflammation (10). The inflammatory response leads to the aggregation of peroxides and the imbalance of oxidation and antioxidation in the colon, particularly the over-accumulation of reactive oxygen species (ROS), causes oxidative damage (11). Antioxidant enzymes, including superoxide dismutase (SOD), are the first line of defense against oxidative stress, helping to remove peroxides from the body, but generally tend to promote oxidation in UC (12). During this process, the nuclear factor erythroid-2-related factor 2 (Nrf2) recognizes the transcription-enhancing sequence associated with antioxidation and repairs bodily damage by enhancing the expression of antioxidant enzymes (13).

Dextran sulfate sodium (DSS) affects the normal replication of DNA in cells, causes the aggregation of inflammatory factors and destruction of the intestinal microenvironment via the overproduction of ROS and eventually leads to the onset of UC (14). DSS was used to induce the occurrence of UC in the present study. Sulfasalazine (SASP), which reduces the infiltration of inflammatory monocytes, has been used to treat UC in clinics (15) and was applied as a positive control agent in this study.

Gloeostereum incarnatum (GI) is an edible and medicinal fungus, commonly cultivated in north-central China and Hokkaido, Japan (16). The anti-inflammatory activities of GI extracts were found to reduce nitric oxide (NO) production in lipopolysaccharide (LPS)-induced RAW264.7 cells and delay melanin synthesis in B16/F10 cells (17). In our previous study, GI was confirmed to show immunomodulatory function in a cyclophosphamide monohydrate-induced mouse model by increasing immune-related factors including immunoglobulin (Ig) (7). Due to its anti-microbial activities, GI shows beneficial effects on gastrointestinal diseases including gastric ulcer and enteritis (18). However, thus far, there has been no systematic research on the protective effect of GI against UC.

In the present study, based on the detection of the main components of GI, its anti-inflammation and antioxidative properties were successfully confirmed in C57BL/6 mice with UC induced via oral administration of 3.5% DSS. The data provide experimental evidence for the applicability of GI for protection against UC.

Materials and methods

Detection of GI components

GI was purchased from TengHui Agriculture, identified by Professor Yu Li (Jilin Agricultural University, China) and pulverized by a pulverizer (XL-06A, Guangzhou Xulang Machinery Equipment Co., Ltd.).

Detection of main components

In GI powder, the contents of total sugar, reducing sugar, mannitol, total ash, total flavonoids, total triterpenoids, crude fat and crude fiber were detected using phenol-sulfuric acid assay (19), 3,5-dinitrosalicylic acid colorimetry (20), iodometry (21), regression analysis (22), the alumina colorimetric method (23), vanillin-glacial acetic acid-perchloric acid colorimetry (24), petroleum ether extraction (25) and the Ankom filter bag method (26), respectively.

Detection of minerals

Inductively coupled plasma optical emission spectrometry was used to detect the contents of mercury (Hg), lead (Pb), selenium (Se), arsenic (As), cadmium (Cd), zinc (Zn), iron (Fe), manganese (Mn), chromium (Cr), calcium (Ca), copper (Cu), sodium (Na) and potassium (K) in GI (27).

Detection of fatty acids

An oil sample of GI was extracted using a chloroform and methanol solution (2:1) and treated by alkaline hydrolysis to prepare the corresponding fatty acid methyl esters. Fatty acids were detected using gas chromatography-mass spectrometry (QP2010, Shimadzu Corporation) based on retention times (28).

Detection of amino acids

The protein in GI was hydrolyzed to single amino acid residues by hydrochloric acid hydrolysis and the contents of different amino acids were analyzed using an automatic amino acid analyzer (L-8900, Hitachi, Ltd.) (29).

Establishment of UC model mice and agent administration

A total of 48 male C57BL/6 mice [22–25 g; 8–10 weeks old; SPF grade, SCXK (LIAO) 2015-001], purchased from Liaoning Changsheng Biotechnology Co., Ltd., were housed in an exhaust ventilation cage system with a temperature of 22±2°C and humidity of 50±10%, with ad libitum access to food and water and a 12-h light/dark cycle. All animal experiments were approved by the Experimental Animal Ethics Committee of Jilin University [approval no. SYXK(JI)2014-0013]. The study was conducted using the Laboratory Guidelines for Animal Care (30,31) and the Guide for the Care and Use of Laboratory Animals (eighth edition) (32).

After 7 days of adaptive feeding, all mice with the exception of the control mice drank freely available clean water containing 3.5% DSS (S14048, Shanghai Yuanye Biological Technology Co., Ltd.) every day for 7 days. From the 8th to the 28th day, the mice drank 3.5% DSS on 1 day out of every 3 days. At the 7th day, the mice were randomly divided into 6 groups: The control group (n=8) that orally received double distilled (D.D.) water, the model group (n=8) that orally received D.D. water, the positive control group (n=8) that orally received 0.6 g/kg of SASP dissolved in D.D. water and the GI-treated groups that orally received 1.0 g/kg (n=8), 2.0 g/kg (n=8) and 4.0 g/kg (n=8) of GI suspended in D.D. water for the remaining 21 days. The doses of GI and SASP were selected based on previous studies (7,33) and our preliminary experiments (data not shown). On the 28th day, blood samples were collected from the orbital venous plexus of the mice and the mice were then euthanized by intraperitoneal injection of 100 mg/kg sodium pentobarbital (Xiya Reagent Co., Ltd.); mortality was characterized by cessation of heartbeat (34). Tissues including the colon, liver, spleen and kidney of each mouse were collected for further detection. The length of the colon and the organ index of the liver, spleen and kidney were calculated. The following formula was used to calculate the organ index (35): Organ index (%)=mean organ weight/mean body weight ×100.

During the entire experimental period, the activity, physiological status and body weight were monitored daily. The status of the UC mice was scored using the disease activity index (DAI) according to the degree of weight loss, fecal viscosity and bleeding (36).

Detection of biochemical factors in the serum and colon of UC mice

Colon tissue was homogenized in D.D. water contained 1.0% 50-mM phenylmethanesulfonyl fluoride (PMSF; Sigma-Aldrich; Merck KGaA) and 1.0% protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA) using a homogenizer [S-18KS, Leopard scientific instruments (Beijing) Co., Ltd.]. After centrifuging the homogenate at 1,000 × g for 10 min at 4°C, the supernatant was collected, and the protein content was determined using a BCA protein assay kit (EMD Millipore). The levels of IL-1β (E20180501A), IL-2 (E20180501A), IL-6 (E20180501A), IL-12 (E20180501A), tumor necrosis factor (TNF)-α (E20180501A), TNF-β (E20180501A), interferon (IFN)-α (E20180501A) and IFN-γ (E20180501A) in the colon and IgA (E20180401A), IgM (E20180401A), IgG (E20170601A), SOD (E20180401A), catalase (CAT) (E20180401A), ROS (E20180401A) and NO (E20180401A) in the colon and serum were measured using ELISA kits purchased from Shanghai Yuanye Biological Technology Co., Ltd.

Histopathological examination of organs in the UC mice

The tissues were fixed in 4% paraformaldehyde at 37°C for 24 h and dehydrated in an ascending series of ethanol, embedded in paraffin and then cut into 5–8-µm-thick sections by a microtome (Leica Microsystems GmbH). The samples were dewaxed with xylene at 37°C and rehydrated in a descending series of ethanol. The samples were stained with 0.45% hematoxylin for 10 min and 0.5% eosin for 3 min, both at 37°C, and then observed and images captured using a light microscope (magnification, ×400; Olympus Corporation).

Western blot analysis of colon tissue

The proteins were extracted from colon tissue as detailed in our previous study (37). Total protein was quantified using a bicinchoninic acid protein assay kit (EMD Millipore) and 40 µg protein/well was separated by 12% SDS-PAGE and transferred to polyvinylidene fluoride membranes (EMD Millipore) using the same method as that used in our previous study (37). The membranes were blocked with 5% BSA (Sigma-Aldrich; Merck KGaA) at 4°C for 4 h and then incubated overnight with primary antibodies against nuclear factor erythroid 2-related factor 2 (Nrf2; 1:2,000; cat. no. ab89443), CAT (1:2,000; cat. no. ab16731), heme oxygenase-1 (HO-1; 1:2,000; cat. no. ab137749), SOD-1 (1:2,000; cat. no. ab16831), SOD-2 (1:5,000; cat. no. ab13533), phosphorylated (p)-NF-κB (1:2,000; cat. no. ab86299), total (T)-NF-κB (1:1,000; cat. no. ab7970), p-inhibitor of NF-κB kinase α+β (IKKα+β; 1:1,000; cat. no. ab195907), T-IKKα+β (1:1,000; cat. no. ab178870), p-inhibitor of NF-κB (IκBα; 1:500; cat. no. ab12135), T-IκBα (1:1,000; cat. no. ab32518) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; 1:1,000; cat. no. ab8245; all from Abcam) at 4°C. The antibodies were diluted according to the ratio given in the manufacturer's protocols. Following washing with a Tris-buffered saline solution containing 0.05% Tween 20, the membranes were incubated with goat anti-rabbit (1:5,000; cat. no. IH-0011) or goat anti-mouse secondary antibody (1:5,000; cat. no. IH-0031; both Beijing Dingguo Changsheng Biotechnology Co Ltd.) at 4°C for 4 h. Electrochemiluminescence detection kits (EMD Millipore) and a Gel Imaging System (UVP, LLC) were used to detect changes in the protein content. The optical densities of bands were measured using ImageJ 1.48u software (National Institutes of Health).

Statistical analysis

Data are expressed as the mean ± standard error of the mean. One-way analysis of variance (ANOVA) and subsequent post-hoc multiple comparisons (Tukey's test) were performed using SPSS 16.0 software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Evaluation of GI components

GI contained 16.6% of total sugar, 16.8% of crude fiber, 2.1% of mannitol, 13.9% of total ash, 6.3% of reducing sugar, 0.394% of total flavonoids, 5.328×10−3% of total triterpenes and 2.8% of crude fat (Table I). The presence of 35 fatty acids was detected, with linoleic acid (1.626%), oleic acid (0.236%) and hexadecenoic acid (0.229%) being the most abundant; by contrast, the fatty acids caprylic acid, undecanoic acid, translinoleic acid and docosadienoic acid were not detected (Table I). GI contained 17 amino acids and 11 minerals, with no harmful elements (Table I).

Table I.

Composition of Gloeostereum incarnatum.

Table I.

Composition of Gloeostereum incarnatum.

A, Main components

CompoundsContents, %
Total sugar16.6
Total ash13.9
Total triterpenes (×10−3)   5.328
Crude fiber16.8
Reducing sugar6.3
Crude fat2.8
Mannitol2.1
Total flavonoids   0.394

B, Fatty acids

CompoundsContents, %

Caprylic acid (C8:0)ND
Capric acid (C10:0)ND
Undecanoic acid (C11:0)ND
Lauric acid (C12:0)0.015
Tridecanoic acid (C13:0)ND
Myristic acid (C14:0)0.003
Myristoleic acid (C14:1)ND
Pentadecanoic acid (C15:0)0.028
Pentadecenoic acid (C15:1)ND
Hexadecanoic acid (C16:0)0.229
Palmitoleic acid (C16:1)0.021
Heptadecanoic acid (C17:0)0.005
Margaroleic acid (C17:1)0.004
Stearic acid (C18:0)0.014
Elaidic acid (C18:1n9t)ND
Oleic acid (C18:1n9c)0.236
Translinoleic acid (C18:2n6t)ND
Linolic acid (C18:2n6c)1.626
α-Linolenic acid (C18:3n3)ND
γ-Linolenic acid (C18:3n6)ND
Arachidic acid (C20:0)ND
Cis-11-Eicosenoic acid C20:1n9)0.067
Docosanoic acid (C22:0)ND
Erucic acid (C22:1n9)ND
Docosadienoic acid (C22:2n6)ND
Docosahexaenoic acid (C22:6n3)ND
Tricosanoic acid (C23:0)0.005
Tetracosanoic acid (C24:0)0.008
Nervonic acid (C24:1n9)0.016
Dihomo-gamma-linolenic acid (C20:3n6)ND
Arachidonic acid (C20:4n6)ND
Eicosapentaenoic acid (C20:5n3)ND
Heneicosanoic acid (C21:0)ND
Eicosadienoic acid (C20:2)0.005
Eicosatrienoic acid (C20:3n3)ND

C, Amino acids

CompoundsContents, %

Aspartic acid0.728
L-Threonine0.405
Serine0.471
Glutamic acid1.012
Glycine0.400
Alanine0.463
Cystine0.266
Valine0.406
Methionine0.096
Isoleucine0.343
Leucine0.616
Tyrosine0.248
Phenylalanine0.335
Lysine0.221
Histidine0.188
Arginine0.510
Proline0.377

D, Minerals

CompoundsContents, mg/kg

MercuryND
Lead0.11493
Selenium0.03450
Arsenic0.12343
CadmiumND
Zinc44.64
Iron105.9
Manganese6.135
Chromium3.15357
Calcium601.5
Copper9.230
Sodium345.8
Potassium48,605

[i] ND, not detected.

Protective effect of GI against UC

In the model mice with UC, DSS free-drinking caused marked body weight loss (P<0.05; Table II) and the worsening of the DAI (P<0.001; Fig. 1A), both of which were reversed after 21-day GI administration (P<0.05; Table II and Fig. 1A). Compared with the model mice, GI treatment in the UC mice prevented the increase of the spleen and liver indexes (P<0.05; Table II) and increased the kidney indexes (P<0.05; Table II); however, SASP demonstrated no significant effects on the liver indexes (Table II). Inflammatory cell infiltration and ulceration of the colon can lead to the shortening of the colon, which was noted in the model mice (Fig. 1B). SASP and GI notably prevented this pathological reduction of the colon length (Fig. 1B). In the colons of the model mice with UC, a large number of exfoliated lymphocytes were identified; meanwhile, the goblet cells were reduced and the crypt cells were damaged, which were all significantly alleviated following SASP and GI treatment (Fig. 1C and D). Furthermore, inflammatory cell infiltration in the spleen (Fig. 1E), interstitial edema in the liver (Fig. 1F) and eosinophilic renal tubular epithelial cells in the kidney (Fig. 1G) were all observed in the DSS-only-treated mice (the model mice). These pathological alterations were all alleviated following SASP and GI administration (Fig. 1E-G).

Table II.

Effect of GI on body weight and organ indexes.

Table II.

Effect of GI on body weight and organ indexes.

3.5% dextran sulfate sodium

Gl, g/kg

VariableControlModelSulfasalazine, 0.6 g/kg1.02.04.0
Body weight, g
  Day 122.8±0.222.6±0.422.3±0.422.5±0.322.6±0.422.6±0.3
  Day 722.4±0.3 20.3±0.3a20.2±0.320.5±0.420.3±0.320.5±0.3
Day 1424.4±0.3 20.6±0.8b21.2±0.521.4±0.721.6±0.321.2±0.7
  Day 2125.4±0.4 20.8±0.8b22.1±0.422.3±0.5 22.9±0.3d22.0±0.5
  Day 2824.3±0.4 21.2±0.5b22.1±0.3 22.7±0.5d 23.4±0.3d21.5±0.3
Organ index, %
  Spleen0.25±0.05 0.56±0.10c 0.45±0.05d0.49±0.14 0.44±0.10d 0.38±0.11e
  Liver3.72±0.31 4.34±0.24a4.32±0.13 3.95±0.44d 3.79±0.40d 3.58±0.27e
  Kidney1.16±0.05 1.03±0.05a 1.13±0.06d1.10±0.10 1.11±0.05d1.01±0.07

{ label (or @symbol) needed for fn[@id='tfn2-mmr-22-04-3418'] } Data were analyzed using a one-way ANOVA and are expressed as mean ± SEM (n=8).

a P<0.05

b P<0.01

c P<0.001 vs. control mice

d P<0.05

e P<0.01 vs. DSS-induced UC mice. GI, Gloeostereum incarnatum.

Anti-inflammatory effects of GI

Inflammation of the mucosal surface of the colon leads to the development of UC (38). The free-drinking of DSS in the present study increased the levels of pro-inflammatory factors including IL-1β, IL-2, IL-6, IL-12, TNF-α, TNF-β, IFN-α and IFN-γ (P<0.05; Table III). GI and SASP demonstrated strong anti-inflammatory effects, as evidenced by the regulation of inflammatory cytokines (P<0.05; Table III). Compared with the model mice, 21-day GI administration in the UC mice resulted in >19.3% (P<0.05), >31.7% (P<0.05), >31.5% (P<0.05), >30.8% (P<0.05), >23.4% (P<0.05), >30.2% (P<0.05), >28.1% (P<0.05) and >33.5% (P<0.05) reduction in IL-1β, IL-2, IL-6, IL-12, TNF-α, TNF-β, IFN-α and IFN-γ levels, respectively, in the colon (Table III).

Table III.

Effect of GI on inflammatory factors of colon tissues of mice with ulcerative colitis.

Table III.

Effect of GI on inflammatory factors of colon tissues of mice with ulcerative colitis.

3.5% dextran sulfate sodium

GI, g/kg

Inflammatory factorsControlModelSASP, 0.6 g/kg1.02.04.0
IL-1β, pg/mgprot45.7±3.9 60.0±3.5a 44.1±3.1d 48.4±1.4c 42.2±3.8c 39.7±3.7d
IL-2, pg/mgprot158.1±9.4 219.1±14.6a 153.8±11.9c 130.2±11.0d 149.5±16.4c 128.4±10.1d
IL-6, pg/mgprot61.6±3.7 83.7±6.3a 54.5±8.2c 57.3±1.4c62.1±6.8 50.0±4.8d
IL-12, pg/mgprot37.3±2.6 57.2±6.3a 31.9±3.9d 30.5±2.0d 39.6±2.0c 27.8±2.1d
TNF-α, pg/mgprot407.5±21.9 489±22.2a429.0±28.9 374.5±27.2c 358.0±43.1c 337.5±14.6d
TNF-β, pg/mgprot78.6±7.8 119.8±11.5b 62.9±7.7d 70.1±4.1d 83.6±10.9c 63.3±4.4d
IFN-α, pg/mgprot19.7±1.6 28.8±2.6b 16.3±1.9d 20.7±1.2c 20.2±1.6c 17.2±1.8d
IFN-γ, pg/mgprot72.4±6.6 126.6±14.8b 72.9±9.5d 75.3±5.4d 84.2±5.5c 69.8±7.0c

{ label (or @symbol) needed for fn[@id='tfn8-mmr-22-04-3418'] } Data were analyzed using a one-way ANOVA and are expressed as mean ± SEM (n=8).

a P<0.05

b P<0.01 vs. control mice

c P<0.05

d P<0.01 vs. DSS-induced UC mice. GI, Gloeostereum incarnatum; IL, interleukin; TNF, tumor necrosis factor; IFN, interferon.

The immune response is involved in the pathogenesis of UC (39). Compared with the control mice, the UC mice (the model group) demonstrated lower levels of IgA, IgG and IgM in the colon and serum (P<0.05; Fig. 2). GI enhanced the levels of IgA (P<0.05) (Fig. 2A) and IgG (P<0.05; Fig. 2C) in the colon and serum and the levels of IgM (P<0.001) (Fig. 2B) in the serum of the UC mice, but did not enhance the levels of IgM in the colon (Fig. 2B).

Effect of GI on oxidative factors

Oxidative stress destroys the cellular macromolecules of the colon, which is the key to the pathogenesis of UC (40). Compared with the control mice, the UC mice demonstrated lower levels of SOD and CAT and higher levels of ROS and NO in the serum and colon tissues (P<0.05; Table IV). In the serum, GI resulted in >14.8% (P<0.01) and >17.4% (P<0.05) increase in SOD and CAT levels and >12.9% (P<0.05) reduction in NO levels, but no effect on ROS levels (Table IV). In the colon tissues, GI failed to affect CAT levels, but resulted in >36.3% (P<0.05) increase in SOD levels and >25.8% (P<0.05) and >45.2% (P<0.05) reductions in ROS and NO levels respectively (Table IV). SASP significantly prevented the pathological alterations in the levels of anti- and pro-oxidative factors in the colon tissues (P<0.05; Table IV).

Table IV.

Effect of GI on oxidative factors in serum and colon tissues of mice with ulcerative colitis.

Table IV.

Effect of GI on oxidative factors in serum and colon tissues of mice with ulcerative colitis.

A, Serum

DSS

GI, g/kg
Oxidative factorsCTRLModelSASP, 0.6 g/kg1.02.04.0
SOD, U/ml99.7±3.3 81.7±2.6c 94.8±3.1e83.9±3.1 93.8±2.2e 98.2±1.9f
CAT, U/ml26.8±0.9 21.9±1.1b 28.1±1.6e24.7±1.7 29.5±1.7e 25.7±0.5d
ROS, U/ml177.1±3.1174.2±3.9165.6±2.8167.3±5.7172.4±2.1179.9±0.7
NO, µmol/l14.0±0.6 17.8±0.8b 14.3±0.6d 15.3±0.8d 15.5±0.8d 15.1±0.4d

B, Colon

DSS

GI, g/kg

Oxidative factorsCTRLModelSASP, 0.6 g/kg1.02.04.0

SOD, U/mgprot152.0±9.6 104.5±9.1b 145.0±11.2d113.8±3.7119.7±8.6 142.4±4.3d
CAT, U/mgprot31.3±2.2 21.4±2.1a 27.8±0.9d22.2±0.625.3±1.922.2±1.4
ROS, U/mgprot310.7±23.9 418.2±33.7a 308.5±22.7d 292.2±19.3d 310.3±13.2d 284.1±13.0d
NO, µmol/gprot13.8±1.1 21.9±2.8a 11.5±1.9e 11.6±0.9d 12.0±1.7d 8.0±0.5e

{ label (or @symbol) needed for fn[@id='tfn13-mmr-22-04-3418'] } Data were analyzed using a one-way ANOVA and are expressed as the mean ± SEMs (n=8).

a P<0.05

b P<0.01

c P<0.001 vs. control mice

d P<0.05

e P<0.01

f P<0.001 vs. DSS-induced UC mice. GI, Gloeostereum incarnatum; CTRL, control; DSS, dextran sulfate sodium.

GI regulates the Nrf2/NF-κB signaling in the colon of UC mice

Compared with the model mice, the UC mice administered with GI demonstrated lower expression levels, in the colon, of Nrf2 and its downstream proteins, including CAT, HO-1, SOD-1 and SOD-2, and higher phosphorylated activities of NF-κB, IKKα/β and IκBα, all of which were prevented by GI administration (P<0.05; Fig. 3).

Discussion

The commonly used anti-colitis drugs mainly include non-steroidal drugs, which exhibit side effects including intestinal ulcers (41,42). Recently, edible mushrooms have attracted researchers' attention due to their diverse pharmacological effects and minimal adverse effects. For example, Ganoderma lucidum alleviates intestinal damage induced by indomethacin in mice (43) and Hericium erinaceus improves testicular damage via its antioxidative effects and regulation of the GPR41/43 receptor in UC rats and Caco-2 cells (44). Based on the immunomodulatory and anti-inflammatory effects of GI reported previously (7), the present study successfully confirmed its protective effect against UC in C57BL/6 mice.

GI is a medicinal and edible fungus with complex ingredients and multiple nutritional value components. Its natural and crude character not only supports its low toxicity with various pharmacological efficacy, but also helps to explain its non-dose dependent effects during the experiments in the present study, which may show anti-UC effects via multiple targets. Indeed, the non-dose dependent manner can be noted in the research associated with Traditional Chinese Medicine (37). GI contains 6.3% crude fiber, which is beneficial for intestinal peristalsis and eases constipation (45). Mixed fiber can markedly regulate blood sugar levels, in addition to inflammatory factors, and exhibits a beneficial regulatory effect on the intestinal microbiota (46). GI contains 0.394% flavonoids, which exert antioxidant effects by regulating the body's oxygen free radical levels. In an LPS-damaged mice model, a flavonoid-rich fraction of Ocimum gratissimum leaves regulated oxidative stress and inflammation in the liver and brain by reducing the levels of TNF-α and malondialdehyde (47). The flavonoid apigenin can reduce the elevation of pro-inflammatory cytokines in the colon, reduce the density of eosinophils and transform M1 pro-inflammatory macrophages to the M2 anti-inflammatory phenotype in diet-induced obese mice (48). GI contains 5.328%x10−3 triterpenes, which trigger the anti-inflammatory response and reduce the expression of pro-inflammatory cytokines in a streptozotocin-induced diabetes model (49). Thus, GI shows a good nutritional foundation for its antioxidation and anti-inflammation activities.

UC is a process driven by T helper type 2 (Th-2)-like T-cells combined with the infiltration of lymphocytes and macrophages (50). DSS-induced damage to the colon tissue in mice causes the activation of macrophages, destruction of the composition of the lamina propria cells and aggregation of inflammatory factors, which eventually leads to imbalance in Th-1/2 cells (51). Cluster of differentiation 1a acts as an inflammatory mediator of UC, inducing T cell activation to elicit an immune response in the body (52). As a central factor in immune mediation, the elevated levels of secretory IgA in the colon help to prevent intestinal damage and restore mucosal barrier function (53). IgG exerts important protective and regulatory effects in the placental barrier (54) and IgM serves an important role in its own immune regulation (55). In the present study, GI increased the levels of Ig in the UC mice to stimulate an immune response.

Furthermore, TGF-β serves a role in pro-inflammatory mediator production during the development of UC, which can regulate the levels of ILs, TNFs and IFNs (56). During the development of UC, signaling of the receptor-interacting protein kinase 3 can upregulate the expression of repair-associated cytokines, including cyclooxygenase 2 and IL-22 (57). As a pro-inflammatory factor, abnormal release of TNF-α recruits more macrophages and neutrophils, which in turn increases inflammatory damage and intestinal permeability (58). In the morphological observations of the current study, reduced goblet cells, crypt abscesses and inflammatory cell infiltration were noted in the colon tissue following DSS administration; these were alleviated following GI administration. The overexpression of IL-6 may aggravate the inflammatory cell infiltration at the injured site and recruit more pro-inflammatory factors (8), which further activate the inducible nitric oxide synthase and cyclooxygenase-2 pathways, in turn increasing neutrophil aggregation (59). In the present study, the regulatory activity of GI on inflammatory factors may have been involved in its protective effect against UC in C57BL/6 mice.

As an upstream regulatory protein, NF-κB can also be activated by increased levels of pro-inflammatory factors (60). The IKKα/β complex is an aggregate composed of a catalytic subunit (61) and is essential for the initiation of the NF-κB pathway (62). In general, IκBα forms a dimer with NF-κB in the cytoplasm. When the inflammatory factor stimulates the IKKα/β complex, IKKα/β phosphorylates IκBα, thereby separating it from NF-κB. NF-κB is thus activated and enters the nucleus to induce the expression of relevant genes to reduce the expression of pro-inflammatory cytokines (63). In the present study, GI markedly reduced the phosphorylated activation of NF-κB by suppressing the phosphorylated IKKα/β and IκBα.

Inflammatory cells in the colon produce ROS and the accumulation of ROS damages proteins and nucleic acids, leading to oxidative stress (64). SOD and CAT are enzymatic antioxidants that catalyze the decomposition of oxides or peroxides to avoid oxidative damage (65). A previous study indicates that SOD reduces inflammation by enhancing the body's antioxidant function (66). SOD decomposes superoxide into hydrogen peroxide (H2O2) and H2O2 can be further decomposed into H2O by CAT to exert antioxidant effects (67). The abnormal levels of anti- and pro-oxidation factors caused by DSS in the present study were all restored by GI treatment. Nrf2 is an important starting element in antioxidant systems (68) that can counteract the damaging effect of peroxide by activating the peroxiredoxin-1 gene (69). Under peroxidative conditions, Nrf2 dissociates from the dimer and transfers to the nucleus, binding to the antioxidant responsive element (70). Binding to this promoter activates the expression of a cytoprotective enzyme, including HO-1, CAT, or SOD (71). Nrf2 upregulates the GST-A4 gene expression via the mitogen-activated protein kinase pathway to protect against UC (69). Nrf2 and NF-κB are inseparable in the process of anti-inflammation and antioxidation. The activation of Nrf2 attenuates inflammatory signaling by inhibiting NF-κB entry into the nucleus, whereas NF-κB directly inhibits the Nrf2 pathway at the RNA level (72). The data from the present study suggested that Nrf2/NF-κB signaling is involved in GI-mediated protection against UC; however, more experiments are required to investigate the synergistic process between Nrf2 and NF-κB signaling.

There remains a limitation the present study. Although the constituents contained in GI, based on the present data, were detected, the active ingredients with the anti-UC effect were not isolated and purified; this will be further investigated.

In summary, GI was identified to alleviate the physiological and pathological state of DSS-induced UC in mice via its antioxidant and anti-inflammatory functions, which may be associated with its modulation of the activation of Nrf2/NF-κB signaling.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Key Research & Development Program of China (grant no. 2018YFE0107800), the Science and Technology Development Program of Jilin Province in China (grant no. 20170623092-TC), the Science and Technology Bureau of Changchun in China (grant no. 15SS11) and the Special Projects of the Cooperation between Jilin University and Jilin Province of China (grant no. SXGJSF2017-1).

Availability of data and materials

All data generated and analyzed during this study are included in this published article.

Authors' contributions

DW and YL contributed to the conceptual design of the research. XiaL, XinL, YoZ, YaZ, SL and NZ performed the experiments. DW, YL, XiaL and XinL analyzed the data and wrote the manuscript. DW and YL helped perform the analysis with constructive discussions. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The experimental animal protocol was approved by the Animal Ethics Committee of Jilin University [approval no. SYXK (JI) 2014-0013]. The present study was carried out under relevant guidelines and regulations.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yue B, Ren YJ, Zhang JJ, Luo XP, Yu ZL, Ren GY, Sun AN, Deng C, Wang ZT and Dou W: Anti-inflammatory effects of fargesin on chemically induced inflammatory bowel disease in mice. Molecules. 23:34182018. View Article : Google Scholar

2 

Cawthorpe D and Davidson M: Temporal comorbidity of mental disorder and ulcerative colitis. Perm J. 19:52–57. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Vargas Robles H, Citalán Madrid AF, García Ponce A, Silva Olivares A, Shibayama M, Betanzos A, Del Valle Mondragón L, Nava P and Schnoor M: Experimental colitis is attenuated by cardioprotective diet supplementation that reduces oxidative stress, inflammation, and mucosal damage. Oxid Med Cell Longev. 2016:84732422016. View Article : Google Scholar : PubMed/NCBI

4 

Bopanna S, Ananthakrishnan AN, Kedia S, Yajnik V and Ahuja V: Risk of colorectal cancer in Asian patients with ulcerative colitis: A systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2:269–276. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Tatiya-Aphiradee N, Chatuphonprasert W and Jarukamjorn K: Immune response and inflammatory pathway of ulcerative colitis. J Basic Clin Physiol Pharmacol. 30:1–10. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Chaussade S and Denizot Y: Mediators of inflammation and hemorrhagic rectocolitis. Ann Gastroenterol Hepatol (Paris). 27:117–121. 1991.(In French). PubMed/NCBI

7 

Wang D, Li Q, Qu Y, Wang M, Li L, Liu Y and Li Y: The investigation of immunomodulatory activities of Gloeostereum incaratum polysaccharides in cyclophosphamide-induced immunosuppression mice. Exp Ther Med. 15:3633–3638. 2018.PubMed/NCBI

8 

Zhang H and Chen W: Interleukin 6 inhibition by triptolide prevents inflammation in a mouse model of ulcerative colitis. Exp Ther Med. 14:2271–2276. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Jeon YD, Bang KS, Shin MK, Lee JH, Chang YN and Jin JS: Regulatory effects of Glycyrrhizae radix extract on DSS-induced ulcerative colitis. BMC Complement Altern Med. 16:4592016. View Article : Google Scholar : PubMed/NCBI

10 

Ai XY, Qin Y, Liu HJ, Cui ZH, Li M, Yang JH, Zhong WL, Liu YR, Chen S, Sun T, et al: Apigenin inhibits colonic inflammation and tumorigenesis by suppressing STAT3-NF-κB signaling. Oncotarget. 8:100216–100226. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Wang Z, Li S, Cao Y, Tian X, Zeng R, Liao DF and Cao D: Oxidative stress and carbonyl lesions in ulcerative colitis and associated colorectal cancer. Oxid Med Cell Longev. 2016:98752982016. View Article : Google Scholar : PubMed/NCBI

12 

Jin Y, Yang J, Lin L, Lin Y and Zheng C: The attenuation of Scutellariae radix extract on oxidative stress for colon injury in lipopolysaccharide-induced RAW264.7 cell and 2,4,6-trinitrobenzene sulfonic acid-induced ulcerative colitis rats. Pharmacogn Mag. 12:153–159. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Bellezza I, Giambanco I, Minelli A and Donato R: Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res. 1865:721–733. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Zhang ZL, Fan HY, Yang MY, Zhang ZK and Liu K: Therapeutic effect of a hydroxynaphthoquinone fraction on dextran sulfate sodium-induced ulcerative colitis. World J Gastroenterol. 20:15310–15318. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Shin MR, Kim KJ, Kim SH, Kim SJ, Seo BI, An HJ and Roh SS: Comparative evaluation between sulfasalazine alone and in combination with herbal medicine on DSS-induced ulcerative colitis mice. BioMed Res Int. 2017:67426522017. View Article : Google Scholar : PubMed/NCBI

16 

Asai R, Mitsuhashi S, Shigetomi K, Miyamoto T and Ubukata M: Absolute configurations of (−)-hirsutanol A and (−)-hirsutanol C produced by Gloeostereum incarnatum. J Antibiot (Tokyo). 64:693–696. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Soo LT: Free radical scavenging, anti-inflammatory and melannin synthesis inhibitory activities of Gloeostereum incarnatum. J Mushrooms. 12:107–116. 2014. View Article : Google Scholar

18 

Bunbamrung N, Intaraudom C, Dramae A, Boonyuen N, Veeranondha S, Rachtawee P and Pittayakhajonwut P: Antimicrobial activity of illudalane and alliacane sesquiterpenes from the mushroom Gloeostereum incarnatum BCC41461. Phytochem Lett. 20:274–281. 2017. View Article : Google Scholar

19 

Rover MR, Johnston PA, Lamsal BP and Brown RC: Total water-soluble sugars quantification in bio-oil using the phenol-sulfuric acid assay. J Anal Appl Pyrolysis. 104:194–201. 2013. View Article : Google Scholar

20 

Lindsay H: A colorimetric estimation of reducing sugars in potatoes with 3,5-dinitrosalicylic acid. Potato Res. 16:176–179. 1973. View Article : Google Scholar

21 

Devapriya F, Sajith P, Ranganathan R and Shanmugam J: Prevalence of biofilm and beta-lactamase producing Staphylococcus in nasal and throat isolates from healthy volunteers: A medical alert. Nepal J Med Sci. 3:79–83. 2014. View Article : Google Scholar

22 

Popek S: Application of regression analysis as a method to determine total ash content in some selected nectar honeys. Nahrung. 47:36–38. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Bahloul N, Bellili S, Aazza S, Chérif A, Faleiro ML, Antunes MD, Miguel MG and Mnif W: Aqueous extracts from tunisian diplotaxis: Phenol content, antioxidant and anti-acetylcholinesterase activities, and impact of exposure to simulated gastrointestinal fluids. Antioxidants. 5:122016. View Article : Google Scholar

24 

Li JJ, Hu XQ, Zhang XF, Liu JJ and Cao LS: Study on variation of main ingredients from spores and fruiting bodies of Ganoderma lucidum. Zhongguo Zhong Yao Za Zhi. 39:4246–4251. 2014.(In Chinese). PubMed/NCBI

25 

Dong QF, Wang JL, Zhang SF, Wang Z, Zhang CX, Gao H, Zhang HM and Zhang L: Antifungal activity of crude extracts and fat-soluble constituents of Holotrichia diomphalia larvae. Bioresour Technol. 99:8521–8523. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Marichal MJ, Trujillo AI, Cadenazzi M and Arias G: Fiber analysis: Evaluation of screen printing fabric filters bags by three statistical approaches. Anim Feed Sci Technol. 169:79–85. 2011. View Article : Google Scholar

27 

da Costa WKOC, da Silva CS, Figueiredo JFD, Nóbrega JA and Paim APS: Direct analysis of deodorants for determination of metals by inductively coupled plasma optical emission spectrometry. J Pharm Biomed Anal. 155:247–252. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Utami PI, Rahayu WS, Nugraha I and Rochana AN: Fatty acid analysis of lipid extracted from rats by gas chromatography-mass spectrometry method. 2ND Annual applied science and engineering conference (AASEC 2017). IOP Conf Series Mater Sci Eng. 2882018.

29 

Paquin CS and Paquin R: Integration, identification and concentration measurement of amino acids in plant samples by means of an automatic amino acid analyzer linked to a mini-computer. J Chromatogr A. 156:79–85. 1978. View Article : Google Scholar

30 

Couto M and Cates C: Laboratory guidelines for animal care. Methods Mol Biol. 1920:407–430. 2019. View Article : Google Scholar : PubMed/NCBI

31 

China sets lab animal guidelines. Science. 351:1372–1373. 2016.

32 

Committee for the Update of the Guide for the Care and Use of Laboratory Animals: Guide for the Care and Use of Laboratory Animals: Eighth Edition. The National Academies Press; Washington, DC: 2011

33 

Gan HT, Chen YQ and Ouyang Q: Sulfasalazine inhibits activation of nuclear factor-kappaB in patients with ulcerative colitis. J Gastroenterol Hepatol. 20:1016–1024. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Cao Z, Dai W, Zhang R, Chen L, Yang X, Hu L, Chiang LY and Liu W: Opening of the adenosine triphosphate-sensitive potassium channel attenuates morphine tolerance by inhibiting JNK and astrocyte activation in the spinal cord. Clin J Pain. 32:617–623. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Taba MY, Mohammadi S, Jalali M, Beheshti F and Attari SS: Effects of different doses of curcumin on testicular histopathology, apoptosis, and reproductive organs weight index in mice D-galactose-induced aging model. Comp Clin Pathol. 28:997–1002. 2019. View Article : Google Scholar

36 

Wang R, Wang L, Luo Y, Wang D, Du R, Du J and Wang Y: Maggot protein ameliorates dextran sulphate sodium-induced ulcerative colitis in mice. Biosci Rep. 38:BSR201817992018. View Article : Google Scholar : PubMed/NCBI

37 

Meng B, Zhang Y, Wang Z, Ding Q, Song J and Wang D: Hepatoprotective effects of Morchella esculenta against alcohol-induced acute liver injury in the C57BL/6 mouse related to Nrf-2 and NF-κB signaling. Oxid Med Cell Longev. 6029876:20192019.

38 

Sarlos P, Kovesdi E, Magyari L, Banfai Z, Szabo A, Javorhazy A and Melegh B: Genetic update on inflammatory factors in ulcerative colitis: Review of the current literature. World J Gastrointest Pathophysiol. 5:304–321. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Gong Y, Niu W, Tang Y, Zhang Q, Liu S, Liu X, Wang X and Xu Y: Aggravated mucosal and immune damage in a mouse model of ulcerative colitis with stress. Exp Ther Med. 17:2341–2348. 2019.PubMed/NCBI

40 

Yan H, Wang H, Zhang X, Li X and Yu J: Ascorbic acid ameliorates oxidative stress and inflammation in dextran sulfate sodium-induced ulcerative colitis in mice. Int J Clin Exp Med. 8:20245–20253. 2015.PubMed/NCBI

41 

Stadnicki A and Frysz-Naglak D: Non-steroidal anti-inflammatory drugs and intestinal side effects. Wiad Lek. 60:286–290. 2007.(In Polish). PubMed/NCBI

42 

Lychkova AÉ and Puzikov AM: Non-steroidal anti-inflammatory drugs in the correction of experimental ulcerative colitis. Eksp Klin Gastroenterol. 7:59–63. 2014.(In Russian).

43 

Nagai K, Ueno Y, Tanaka S, Hayashi R, Shinagawa K and Chayama K: Polysaccharides derived from Ganoderma lucidum fungus mycelia ameliorate indomethacin-induced small intestinal injury via induction of GM-CSF from macrophages. Cell Immunol. 320:20–28. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Wang D, Zhang Y, Yang S, Zhao D and Wang M: A polysaccharide from cultured mycelium of Hericium erinaceus relieves ulcerative colitis by counteracting oxidative stress and improving mitochondrial function. Int J Biol Macromol. 125:572–579. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Mehmood MH, Aziz N, Ghayur MN and Gilani AH: Pharmacological basis for the medicinal use of Psyllium husk (Ispaghula) in constipation and diarrhea. Dig Dis Sci. 56:1460–1471. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Zhai X, Lin D, Zhao Y, Li W and Yang X: Effects of dietary fiber supplementation on fatty acid metabolism and intestinal microbiota diversity in C57BL/6J mice fed with a high-fat diet. J Agric Food Chem. 66:12706–12718. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Ajayi AM, Ben-Azu B, Onasanwo SA, Adeoluwa O, Eduviere A and Ademowo OG: Flavonoid-rich fraction of Ocimum gratissimum attenuates lipopolysaccharide-induced sickness behavior, inflammatory and oxidative stress in mice. Drug Res (Stuttg). 69:151–158. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Gentile D, Fornai M, Colucci R, Pellegrini C, Tirotta E, Benvenuti L, Segnani C, Ippolito C, Duranti E, Virdis A, et al: The flavonoid compound apigenin prevents colonic inflammation and motor dysfunctions associated with high fat diet-induced obesity. PLoS One. 13:e01955022018. View Article : Google Scholar : PubMed/NCBI

49 

Wu YS and Chen SN: Extracted triterpenes from Antrodia cinnamomea reduce the inflammation to promote the wound healing via the STZ inducing hyperglycemia-diabetes mice model. Front Pharmacol. 7:1542016. View Article : Google Scholar : PubMed/NCBI

50 

Majewska-Szczepanik M, Góralska M, Marcińska K, Zemelka-Wiącek M, Strzępa A, Dorożyńska I and Szczepanik M: Epicutaneous immunization with protein antigen TNP-Ig alleviates TNBS-induced colitis in mice. Pharmacol Rep. 64:1497–1504. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Queen D, Hedayat AA, Magro C and Geskin LJ: An unusual cause of bilateral orbital swelling: Immunoglobulin G4-related orbital disease arising in a patient with ulcerative colitis. JAAD Case Rep. 5:634–638. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Al-Amodi O, Jodeleit H, Beigel F, Wolf E, Siebeck M and Gropp R: CD1a-expressing monocytes as mediators of inflammation in ulcerative colitis. Inflamm Bowel Dis. 24:1225–1236. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Cui Y, Zhu C, Ming Z, Cao J, Yan Y, Zhao P, Pang G, Deng Z, Yao Y and Chen Q: Molecular mechanisms by which casein glycomacropeptide maintains internal homeostasis in mice with experimental ulcerative colitis. PLoS One. 12:e01810752017. View Article : Google Scholar : PubMed/NCBI

54 

Ignat'eva NV, Ziganshina MM, Shilova NV, Khasbiullina NR, Bovin NV, Tyutyunnik VL and Sukhikh GT: Isolation of IgG associated with human placenta. Bull Exp Biol Med. 167:120–122. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Kubagawa H, Honjo K, Ohkura N, Sakaguchi S, Radbruch A, Melchers F and Jani PK: Functional roles of the IgM Fc receptor in the immune system. Front Immunol. 10:9452019. View Article : Google Scholar : PubMed/NCBI

56 

Binabaj MM, Asgharzadeh F, Avan A, Rahmani F, Soleimani A, Parizadeh MR, Ferns GA, Ryzhikov M, Khazaei M and Hassanian SM: EW-7197 prevents ulcerative colitis-associated fibrosis and inflammation. J Cell Physiol. 234:11654–11661. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Xu YL, Tang HL, Zhu SY, Peng HR, Qi ZT and Wang W: RIP3 deficiency exacerbates inflammation in dextran sodium sulfate-induced ulcerative colitis mice model. Cell Biochem Funct. 35:156–163. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Liang J, Liang J, Hao H, Lin H, Wang P, Wu Y, Jiang X, Fu C, Li Q, Ding P, et al: The extracts of Morinda officinalis and its hairy roots attenuate dextran sodium sulfate-induced chronic ulcerative colitis in mice by regulating inflammation and lymphocyte apoptosis. Front Immunol. 8:9052017. View Article : Google Scholar : PubMed/NCBI

59 

Yang Y, He J, Suo Y, Lv L, Wang J, Huo C, Zheng Z, Wang Z, Li J, Sun W and Zhang Y: Anti-inflammatory effect of taurocholate on TNBS-induced ulcerative colitis in mice. Biomed Pharmacother. 81:424–430. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Gupta RA, Motiwala MN, Dumore NG, Danao KR and Ganjare AB: Effect of piperine on inhibition of FFA induced TLR4 mediated inflammation and amelioration of acetic acid induced ulcerative colitis in mice. J Ethnopharmacol. 164:239–246. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Koppe C, Verheugd P, Gautheron J, Reisinger F, Kreggenwinkel K, Roderburg C, Quagliata L, Terracciano L, Gassler N, Tolba RH, et al: IκB kinaseα/β control biliary homeostasis and hepatocarcinogenesis in mice by phosphorylating the cell-death mediator receptor-interacting protein kinase 1. Hepatology. 64:1217–1231. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Li Y, Li J, Li B, Qin H, Peng X, Zhao Y and Chen Y: Anthocyanin suppresses CoCrMo particle-induced osteolysis by inhibiting IKKα/β mediated NF-κB signaling in a mouse calvarial model. Mol Immunol. 85:27–34. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Reale C, Iervolino A, Scudiero I, Ferravante A, D'Andrea LE, Mazzone P, Zotti T, Leonardi A, Roberto L, Zannini M, et al: NF-κB essential modulator (NEMO) is critical for thyroid function. J Biol Chem. 291:5765–5773. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Nikkhah-Bodaghi M, Maleki I, Agah S and Hekmatdoost A: Zingiber officinale and oxidative stress in patients with ulcerative colitis: A randomized, placebo-controlled, clinical trial. Complement Ther Med. 43:1–6. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Meurer MC, Mees M, Mariano LNB, Boeing T, Somensi LB, Mariott M, da Silva RCMVAF, Dos Santos AC, Longo B, Santos França TC, et al: Hydroalcoholic extract of Tagetes erecta L. flowers, rich in the carotenoid lutein, attenuates inflammatory cytokine secretion and improves the oxidative stress in an animal model of ulcerative colitis. Nutr Res. 66:95–106. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Kang JE, Kim HD, Park SY, Pan JG, Kim JH and Yum DY: Dietary supplementation with a Bacillus superoxide dismutase protects against γ-radiation-induced oxidative stress and ameliorates dextran sulphate sodium-induced ulcerative colitis in mice. J Crohn's Colitis. 12:860–869. 2018. View Article : Google Scholar

67 

Rana SV, Sharma S, Prasad KK, Sinha SK and Singh K: Role of oxidative stress & antioxidant defence in ulcerative colitis patients from north India. Indian J Med Res. 139:568–571. 2014.PubMed/NCBI

68 

Almeer RS, Mahmoud SM, Amin HK and Abdel Moneim AE: Ziziphus spina-christi fruit extract suppresses oxidative stress and p38 MAPK expression in ulcerative colitis in rats via induction of Nrf2 and HO-1 expression. Food Chem Toxicol. 115:49–62. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Sabzevary-Ghahfarokhi M, Shohan M, Shirzad H, Rahimian G, Soltani A, Ghatreh-Samani M, Deris F, Bagheri N, Shafigh M and Tahmasbi K: The regulatory role of Nrf2 in antioxidants phase2 enzymes and IL-17A expression in patients with ulcerative colitis. Pathol Res Pract. 214:1149–1155. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Liu D, Huo X, Gao L, Zhang J, Ni H and Cao L: NF-κB and Nrf2 pathways contribute to the protective effect of Licochalcone A on dextran sulphate sodium-induced ulcerative colitis in mice. Biomed Pharmacother. 102:922–929. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Li J, Wang H, Zheng Z, Luo L, Wang P, Liu K, Namani A, Jiang Z, Wang XJ and Tang X: Mkp-1 cross-talks with Nrf2/Ho-1 pathway protecting against intestinal inflammation. Free Radic Biol Med. 124:541–549. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Saber S, Khalil RM, Abdo WS, Nassif D and El-Ahwany E: Olmesartan ameliorates chemically-induced ulcerative colitis in rats via modulating NFκB and Nrf-2/HO-1 signaling crosstalk. Toxicol Appl Pharmacol. 364:120–132. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 22 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li X, Liu X, Zhang Y, Zhang Y, Liu S, Zhang N, Li Y and Wang D: Protective effect of Gloeostereum incarnatum on ulcerative colitis via modulation of Nrf2/NF‑κB signaling in C57BL/6 mice . Mol Med Rep 22: 3418-3428, 2020
APA
Li, X., Liu, X., Zhang, Y., Zhang, Y., Liu, S., Zhang, N. ... Wang, D. (2020). Protective effect of Gloeostereum incarnatum on ulcerative colitis via modulation of Nrf2/NF‑κB signaling in C57BL/6 mice . Molecular Medicine Reports, 22, 3418-3428. https://doi.org/10.3892/mmr.2020.11420
MLA
Li, X., Liu, X., Zhang, Y., Zhang, Y., Liu, S., Zhang, N., Li, Y., Wang, D."Protective effect of Gloeostereum incarnatum on ulcerative colitis via modulation of Nrf2/NF‑κB signaling in C57BL/6 mice ". Molecular Medicine Reports 22.4 (2020): 3418-3428.
Chicago
Li, X., Liu, X., Zhang, Y., Zhang, Y., Liu, S., Zhang, N., Li, Y., Wang, D."Protective effect of Gloeostereum incarnatum on ulcerative colitis via modulation of Nrf2/NF‑κB signaling in C57BL/6 mice ". Molecular Medicine Reports 22, no. 4 (2020): 3418-3428. https://doi.org/10.3892/mmr.2020.11420