Epigallocatechin‑3‑gallate‑induced inhibition of interleukin‑6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice

  • Authors:
    • Zhenglei Xu
    • Cheng Wei
    • Ru Zhang
    • Jun Yao
    • Dinguo Zhang
    • Lisheng Wang
  • View Affiliations

  • Published online on: October 23, 2015     https://doi.org/10.3892/etm.2015.2824
  • Pages: 2231-2238
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epigallocatechin‑3‑gallate (EGCG) has a promising therapeutic effect for ulcerative colitis (UC), but the treatment mechanism has yet to be fully elucidated. The aim of the present study was to investigate the mechanism of EGCG in the treatment of UC. Experimental colitis mouse models were prepared. The mice were randomly divided into four groups: Normal control, model (MD), 50 mg/kg/day EGCG treatment and 100 mg/kg/day EGCG treatment. The daily disease activity index (DAI) of the mice was recorded, changes in the organizational structure of the colon were observed and the spleen index (SI) was measured. In addition, levels of interleukin (IL)‑6, IL‑10, IL‑17 and transforming growth factor (TGF)‑β1 in the plasma and hypoxia‑inducible factor (HIF)‑1α and signal transducer and activator of transcription (STAT) 3 protein expression in colon tissues were evaluated. Compared with the MD group, the mice in the two EGCG treatment groups exhibited decreased DAIs and SIs and an attenuation in the colonic tissue erosion. EGCG could reduce the release of IL‑6 and IL‑17 and regulate the mouse splenic regulatory T‑cell (Treg)/T helper 17 cell (Th17) ratio, while increasing the plasma levels of IL‑10 and TGF‑β1 and decreasing the HIF‑1α and STAT3 protein expression in the colon. The experiments confirmed that EGCG treated mice with experimental colitis by inhibiting the release of IL‑6 and regulating the body Treg/Th17 balance.

Introduction

Ulcerative colitis (UC) is a chronic, non-specific inflammatory disease that involves the rectum and colon of the body; however, the etiology and pathogenesis of the disease have yet to be fully elucidated. Studies have suggested that UC is associated with a variety of factors, such as genetics, infection, the immune system, and environmental and intestinal dysbiosis, as well as damage to the intestinal tissues caused by immune cells and inflammatory cytokines in the inflammatory signaling cascade (1,2). In colonic mucosal lesions, inflammation, mucosal congestion, edema and ulcers may be visible, as well as the infiltration of immune cells (lymphocytes and neutrophils) in the acute exacerbation stage (3). Following the administration of 5% dextran sulfate sodium (C6H7Na3O14S3, DSS), mice gradually exhibit the symptoms of colitis, such as loose stools, bloody diarrhea and weight loss, thus forming an experimental colitis model similar to human UC (4).

Epigallocatechin-3-gallate (EGCG) is a type of natural compound that can be extracted from green tea. EGCG has no toxic effects but has a high biological activity and is known to exert antibacterial, anti-inflammatory, anti-tumor, antioxidation and anti-aging effects (58). Furthermore, it has been demonstrated that EGCG can modulate the immune system to treat autoimmune disease (911). EGCG is known to effectively treat experimental colitis in mice, but its therapeutic mechanism underlying this treatment is unclear (9,12). In a previous study it was found that an imbalance in the number and function of regulatory T cells (Tregs)/T helper 17 cells (Th17s) and the secretion of certain cytokines, such as interleukin (IL)-6, IL-10, IL-17 and transforming growth factor (TGF)-β1, had a crucial role in the development of UC (13). The aim of the present study was therefore to investigate the mechanism of EGCG in the treatment of UC by measuring the levels of proinflammatory [interleukin (IL)-6 and IL-17] and anti-inflammatory [IL-10 and transforming growth factor (TGF)-β1] cytokines in the plasma and the colonic protein expression of hypoxia-inducible factor (HIF)-1α and signal transducer and activator of transcription (STAT) 3 in a mouse model of colitis. The Treg/Th17 balance was also examined.

Materials and methods

Experimental animals

Forty male BALB/c mice (age, 6–7 weeks; weight, 22–26 g) of specific pathogen-free status were obtained from the China Experimental Animal Center of Southern Medical University (Guangzhou, China; certificate of conformity no. SCXK Guangdong 2011-0015) for use in the present study. The mice were housed in clean animal houses at a controlled temperature (22–25°C) and a relative humidity of 55%, and were subjected to a 12-h light/dark cycle. Food and drinking water were available ad libitum. This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The animal use protocol was reviewed and approved by the Institutional Animal Care and Use Committee of the Second Clinical Medical College of Jinan University (Shenzhen, China).

Treatment program

The 40 mice were fed with adaptability for one week, and then randomly divided into four groups (n=10/group): Normal control (NC), model (MD), EGCG (Qiyun Biotechnology Co., Ltd., Guangzhou, China) 50 mg/kg/day treatment (ELD) and EGCG 100 mg/kg/day treatment (EHD). The NC group was given free access to sterile distilled water for 14 days, while the MD and EGCG treatment groups received 5% DSS (Qiyun Biotechnology Co., Ltd.) continuously for seven days; the DSS was replaced by distilled water for seven days following the successful establishment of the model. On day 8 of the experiment, the mice in the NC and MD groups were given a daily gavage of 0.2 ml 0.5% ethanol for seven days, while the ELD and EHD groups were administered EGCG at doses of 50 and 100 mg/kg/day for each mouse, respectively. A total of 0.2 ml 0.5% ethanol was then used to dissolve the EGCG for gavage treatment for seven days. On the day 14 the mice were administered 100 mg/kg pentobarbital anesthesia, and eyeball blood was obtained following the sacrifice of the mice. Blood plasma and spleen and colon tissues were collected under sterile conditions.

Macroscopic and microscopic assessment

The disease activity index (DAI) and spleen index (SI) of the mice were measured and recorded daily, with reference to the Murthy scoring system (14). Details of the scoring system are shown in Table I. The DAI was calculated with the following formula: DAI = (weight loss rate score + stool consistency score + presence of blood in stools score)/3. The spleens of the mice were washed with saline, and then dried and weighed for the calculation of the SI using the following formula: SI = spleen weight (mg)/body weight (g).

Table I.

Murthy scoring system (10).

Table I.

Murthy scoring system (10).

ScoresWeight loss rate (%)Stool stickinessPresence of blood in stool
0(−)Normal (granular, with form)Normal
11–5
2   6–10Soft (mushy, not adhering to the anus)Occult blood (+)
311–15
4>15Diarrhea (watery, adhering to the anus)Bloody stools (+)
Specimen collection and pathology assessment

The mice were sacrificed following pentobarbital anesthetization and the abdominal cavity was opened. Measuring from the anus, a 10-cm section of colon was cut along the longitudinal axis of the mesentery and rinsed repeatedly with iced saline. The section was then cut vertically into two parts, taking the lesion as the center in the longitudinal axis. One colon sample was immediately placed in 10% formalin solution at room temperature and fixed for 48 h, prior to undergoing tissue dehydration, clearing, embedding and staining with hematoxylin and eosin. The colon tissue morphology was subsequently observed by microscopy. For the second section of the colon tissue, the lesion was excised, dried by filter paper, weighed and frozen at −70°C for tissue specimen tests.

Lymphocyte extraction from spleen tissue

Under sterile conditions, the spleen of each mouse was obtained for lymphocyte separation. A 5-ml volume of lymphocyte separation medium (Shenzhen Lvshiyuan Biotechnology, Co., Ltd., Shenzhen, China) was added to 35-mm petri dishes with homogenized spleen tissue. Following the separation of the suspension, spleen cell fluid was immediately transferred to a 15-ml centrifuge tube with 200–500 µl RPMI-1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Samples were centrifuged at 800 × g for 30 min at room temperature. The lymph cell layer was removed by suction and 10 ml RPMI-1640 medium was added for reverse washing. The samples were centrifuged at 250 × g for 10 min at room temperature, and the lymphocytes were subsequently collected. The number of lymphocytes was then adjusted to 2×106/reaction tube.

Flow cytometry

The lymphocyte samples (2×106/reaction tube) were analyzed using flow cytometry with a Th17/Treg kit (BD Biosciences, Franklin Lakes, NJ, USA) according to the manufacturer's instructions. A total of 50 ng/ml phorbol 12-myristate 13-acetate (for the lymphocytes), 1 µg/ml ionomycin and monensin were used to stimulate the cells for 5 h. After 5 h, the cells were collected, stained, fixed and lysed in strict accordance with the kit instructions, and 20 µl streaming antibodies were added to each tube in turn, mixed and incubated for 30 min. The main antibodies used in the flow cytometry included mouse cluster of differentiation 4 (CD4)-PerCP-Cyanine5.5, IL-17A-phycoerythrin (PE) and Alexa Fluor 647-forkhead box protein P3 (Foxp3). A parallel negative control group was set, and 0.2 mg/l PE-rat immunoglobulin (Ig)G1 and 0.2 mg/l Alexa Fluor 647-Rat IgG2b were added. The samples were loaded and the data were analyzed.

ELISA

Blood was obtained from the mice and treated with heparin to prevent coagulation, prior to the supernatant being centrifuged for 5 min at 1,000 × g (4°C). A total of 50 mg mouse colon tissue was weighed using an electronic analytical balance and cut into pieces as soon as possible with a small pair of ophthalmological scissors. Following cutting, 1,200 µl physiological saline at 0°C was added and the tissue was fully homogenized on ice using a glass homogenizer and centrifuged for 10 min using a refrigerated high-speed centrifuge (Sigma, St. Louis, MO, USA) at 4°C (1,000 × g). The supernatant was obtained, and protein quantification was then performed using a NanoDrop™ 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). According to the specifications of the ELISA kit (Dakota Biotech Co. Ltd., Beijing, China), the IL-6, IL-10, IL-17 and TGF-β1 concentrations in the plasma and colon tissues of the mice were detected.

Western blot analysis

A total of 100 mg mouse colon tissue was accurately weighed using an electronic analytical balance, and plasma and nuclear proteins were extracted from the cells using a nucleus and cytoplasm tissue extraction kit in accordance with the manufacturer's instructions (Thermo Fisher Scientific, Inc.). The protein samples were quantified using a NanoDrop 2000 spectrophotometer and the protease inhibitor phenylmethylsulfonyl fluoride was added. The proteins were then separated using SDS-PAGE (10%) and transferred to a nitrocellulose membrane (Beijing Bayer Di Biotechnology, Co., Ltd., Beijing, China). The membrane was blocked with Western blocking buffer (P0023B) and incubated with the primary and secondary antibodies, for 1 h each, and subsequently developed using enhanced chemiluminescence for analysis. Western washing liquid (P0023C) was used to wash the membrane between incubations. Anti-mouse polyclonal STAT3 (1:500; Bioworld Technology, Inc., St. Louis Park, MN, USA) and HIF-1α (1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA) primary antibodies were used to analyze the nuclear HIF-1α and cytoplasmic STAT3 protein levels. Goat anti-rabbit horseradish peroxidase secondary antibodies (1:2,000; Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA) were incubated with the membranes with phosphate-buffered saline, containing 0.1% Tween-20, 5% w/v skimmed milk powder and 2% BSA diluent/blocking solution, for 1 h at room temperature. In addition, levels where tested against lamin or β-actin (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA).

Statistical analysis

Experimental data are expressed as the mean ± standard deviation. The statistical analysis of the experimental data was performed using SPSS statistical software, version 20.0 (IBM SPSS, Armonk, NY, USA). The differences between the groups were compared using single-factor analysis of variance, and P<0.05 was considered to indicate a statistically significant difference.

Results

Assessment of DAI and SI

No animals died during the experiment. During the course of the experiment the activity, body weight, stool consistency and presence of blood in the stools of the mice were observed and recorded daily. The score of the NC group was 0. Blood, weight loss and significant decreases in activity were visible in the MD, ELD and EHD groups following the establishment of the model using 5% DSS for seven days. As shown in Fig. 1A, the presence of blood in the stools, the rate of weight loss and the DAI scores were significantly reduced in the ELD and EHD groups between days 10 and 14 compared with those of the MD group. Compared with the score in the ELD group, the DAI was decreased more significantly in the EHD group. The spleen in the MD group was enlarged compared with that in the NC group, while the spleen size and SI were significantly reduced in the ELD and EHD groups as compared with the MD group (Fig. 1B and C). The SI in the EHD group was decreased to a greater extent than that in the EDL group (Fig. 1C).

Pathological assessment

Compared with the NC group (Fig. 2A), the MD group (Fig. 2B) exhibited visible pathological manifestations, including acute inflammation accompanied by mucosal erosion, as well as edema, crypt reduction and the infiltration of inflammatory cells, such as neutrophils, in the muscularis propria and mucosa. Compared with the MD group, the colonic mucosal inflammatory cell infiltration, erosion and edema in the ELD (Fig. 2C) and EHD (Fig. 2D) groups were significantly improved.

Treg/Th17 ratio

Compared with the NC group, the CD4+IL-17+ (Th17)/CD4+ lymphocyte ratio was significantly elevated in the MD group, while the CD4+Foxp3+ (Treg)/CD4+ lymphocyte ratio and the Treg/Th17 ratio were decreased significantly (P<0.001) (Fig. 3). Compared with the MD group, the levels of Th17s in the ELD and EHD groups were decreased and the Treg/Th17 ratio was significantly increased (P<0.001). The difference in the Treg/Th17 ratio between the EHD and NC groups was not statistically significant (P=0.674), and a statistically significant difference was also found in the ratio between the ELD and NC groups (P=0.006). Compared with the ELD group, the Treg/Th17 ratio in the EHD group showed a significant increase (P<0.001).

Cytokines

Compared with the NC group, the IL-6 and IL-17 levels of the MD group were significantly increased, while the IL-10 and TGF-β1 levels were significantly decreased. Compared with the MD group, the IL-6 and IL-17 levels in the ELD and EHD groups were decreased while the IL-10 and TGF-β1 levels were significantly increased. Compared with the ELD group, the IL-6 and IL-17 levels in the EHD group were decreased to a greater extent, while the IL-10 and TGF-β1 levels were increased to a greater extent (Fig. 4).

Western blot analysis

As presented in Fig. 5, the nuclear HIF-1α and cytoplasmic STAT3 protein expression in the MD group was significantly increased compared with that in the NC group. Compared with the MD group, the expression of the proteins in the ELD and EHD treatment groups was decreased significantly, with a greater decrease observed in the EHD group.

Discussion

UC is a chronic inflammatory disease of the intestine. The exact pathogenesis of the disease is not fully understood, but it is believed to involve the abnormal activation of the innate and adaptive immune systems. The immune inflammatory response exhibits excessive hyperactivity, which negatively affects the self-limiting nature of the reaction (15,16). Immune cells and cell factors involved in intestinal tissue are imbalanced and lead to immune injury (15,16). It has previously been suggested that UC is mediated by Th2 cells (1721), while other studies have also indicated that the Thl/Th2 axis is fundamental to the pathogenesis of UC (2225); however, the identification of Th17s confirmed the immune bypass in UC and explained the anomalies of the traditional Th1/Th2 imbalance (2628). The rebalancing of the Treg/Th17 ratio is considered to be an important treatment strategy for UC (29,30). It is known that Treg/Th17 imbalance is common in a variety of autoimmune disorders, and that human inflammatory bowel disease (IBD) is associated with Tregs, Th17s and the secreted cytokines. The Treg/Th17 imbalance may therefore be a possible target for the treatment of IBD (2,31,32). IBD includes UC and Crohn's disease. The findings of the present study further confirmed the existence of a Treg/Th17 ratio imbalance in the mouse model of experimental colitis, a model reflecting human UC, and were consistent with the results of previous reports (3335). Previous studies have found that EGCG can treat UC, but the exact mechanism has yet to be fully elucidated. We hypothesized that EGCG could treat UC by mediating the rebalancing of the Treg/Th17 ratio, and attempted to explore the specific mechanisms underlying the EGCG-induced adjustments in the Treg/Th17 ratio.

Th17s are involved in the immune pathological process of UC, possibly by releasing IL-17, IL-21, IL-22 and other inflammatory cytokines (36). Tregs are a CD4+ T-cell subset with immunosuppressive activity. These cells inhibit the intestinal mucosal inflammation cascade and inflammatory reaction amplification effect by regulating the secretion of IL-10, TGF-β1 and other anti-inflammatory cytokines and maintain intestinal immune balance (37,38). Tregs act antagonistically with Thl7s to maintain the relative stability of the body's immune status, so that the body is held in a delicate and complex balance. Tregs undergo a similar differentiation process to Thl7s. It has been demonstrated that naive CD4+ T cells can differentiate into Tregs following stimulation from TGF-β1 alone, but, in the presence of IL-6, TGF-β1 and IL-6 act together to induce the expression of the transcription factor retinoic acid-related orphan receptor γt (RORγt), thus suppressing the production of Tregs and promoting the naive CD4+ T cells to differentiate into Th17s (3941) (Fig. 6). At low concentrations, TGF-β1 acts synergistically with IL-6 to induce RORγt generation; at high concentrations, TGF-β1 can regulate the expression of Foxp3. The balance between Foxp3 and RORγt could determine whether the naive T cells differentiate into Th17s or Tregs following stimulation by antigens (42). In the absence of IL-6, TGF-β1 does not induce the differentiation of the naive T cells into Th17s, but instead promotes the differentiation to Tregs (43). In the absence of TGF-β, Th17s can also be generated following stimulation by IL-6 or the combination of IL-23 and IL-1β (44). IL-6 can additionally inhibit the TGF-β-induced generation of Tregs (45).

It has previously been shown that two pathways, hypoxia-mammalian target of rapamycin-HIF-1α-Th17 and IL-6-STAT3-HIF-1α-Th17, have a crucial role in Treg/Th17 imbalance (26). In the first pathway, UC leads to a sustained hypoxic state in the intestine, and the local tissue hypoxia induces HIF-1α synthesis and promotes its entry into the nucleus to exert its biological functions, resulting in the upregulation of Th17 activity, the promotion of Foxp3 binding with ubiquitin for degradation, and then the downregulation and inhibition of Tregs (31,32,46). In the second pathway, UC causes increases in the levels of inflammatory cytokines, such as IL-6, leading to the upregulation and activation of HIF-1α by the IL-6/STAT3 signaling pathway. STAT3 can also inhibit the ubiquitination and degradation of HIF-1α and prolong its half-life, in addition to increasing the protein expression of HIF-1α; as a consequence, increased HIF-1α is available to activate Th17s (4749).

IL-6 promotes the differentiation of Th17s (39,5052), and the activated Thl7s secrete proinflammatory cytokines such as IL-17 (5355). IL-17 can further promote the release of cytokines, such as IL-6 and TNF, and the activation of other inflammatory cells (56). The waterfall-like inflammatory cell and cytokine cascade may be central to the disease progression of patients with UC (13). Reducing the concentration of IL-6 could reduce the induced expression of RORγt and inhibit the IL-6-STAT3-HIF-1α-Th17 pathway, thereby reducing the generation of Th17 cells (39,57). The waterfall-like effects could thus be interrupted and the inhibition of Treg generation could be reduced, thereby regulating the Treg/Th17 balance. EGCG has been found to reduce the generation of IL-6 in patients with IBD (58,59). Since IL-6 plays such an important role in the mechanism of Treg/Th17 imbalance, we speculated that EGCG could regulate the Treg/Th17 balance by reducing IL-6 levels in a mouse model of experimental colitis.

In the present study it was found that, compared with the MD group, the IL-6 levels in the ELD and EHD groups were significantly reduced, with a more significant decrease in the EHD group. This demonstrated that EGCG could significantly reduce the release of IL-6 in mice with experimental colitis. It could be concluded that EGCG reduced the induced expression of RORγt by downregulating the cytokine IL-6 and inhibiting STAT3 and HIF-1α protein expression, thus reducing the generation of Th17s and simultaneously reducing the inhibition of IL-6 on Tregs, and ultimately leading to the rebalancing of the Treg/Th17 ratio and treating mice with experimental colitis. This mechanism would explain the findings in the present study. The experiments also confirmed that the effect of EGCG treatment was dose-related, as the high-dose therapy produced superior results to the low-dose therapy. In conclusion, EGCG represents a natural medicine that exhibits potential for clinical application in the treatment of UC.

Acknowledgements

This study was supported by the Guangdong Provincial Natural Science Foundation of China (no. 10151802001000002) and the Shenzhen Key Science and Technology Projects (no. 200901003). The authors would like to thank the Southern Medical Cell and Microbiology Laboratory.

References

1 

Sartor RB: Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology. 126:1620–1633. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Lei X, Liu M, Yang Z, et al: Thymoquinone prevents and ameliorates dextran sulfate sodium-induced colitis in mice. Dig Dis Sci. 57:2296–2303. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Lichtenstein GR and Rutgeerts P: Importance of mucosal healing in ulcerative colitis. Inflamm Bowel Dis. 16:338–346. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Yao J, Wang JY, Lai MG, et al: Treatment of mice with dextran sulfate sodium-induced colitis with human interleukin 10 secreted by transformed Bifidobacterium longum. Mol Pharm. 8:488–497. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Zaveri NT: Green tea and its polyphenolic catechins: Medicinal uses in cancer and noncancer applications. Life Sci. 78:2073–2080. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Higdon JV and Frei B: Tea catechins and polyphenols: Health effects, metabolism, and antioxidant functions. Crit Rev Food Sci Nutr. 43:89–143. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Yang CS, Maliakal P and Meng X: Inhibition of carcinogenesis by tea. Annu Rev Pharmacol Toxicol. 42:25–54. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Caderni G, De Filippo C, Luceri C, Salvadori M, Giannini A, Biggeri A, Remy S, Cheynier V and Dolara P: Effects of black tea, green tea and wine extracts on intestinal carcinogenesis induced by azoxymethane in F344 rats. Carcinogenesis. 21:1965–1969. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Brückner M, Westphal S, Domschke W, Kucharzik T and Lügering A: Green tea polyphenol epigallocatechin-3-gallate shows therapeutic antioxidative effects in a murine model of colitis. J Crohns Colitis. 6:226–235. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Navarro-Perán E, Cabezas-Herrera J, Sánchez-Del-Campo L, García-Cánovas F and Rodríguez-López JN: The anti-inflammatory and anti-cancer properties of epigallocatechin-3-gallate are mediated by folate cycle disruption, adenosine release and NF-kappaB suppression. Inflamm Res. 57:472–478. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Miura Y, Chiba T, Tomita I, et al: Tea catechins prevent the development of atherosclerosis in apoprotein E-deficient mice. J Nutr. 131:27–32. 2001.PubMed/NCBI

12 

Ran ZH, Chen C and Xiao SD: Epigallocatechin-3-gallate ameliorates rats colitis induced by acetic acid. Biomed Pharmacother. 62:189–196. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Eastaff-Leung N, Mabarrack N, Barbour A, Cummins A and Barry S: Foxp3+ regulatory T Cells, Th17 effector cells, and cytokine environment in inflammatory bowel disease. J Clin Immunol. 30:80–89. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Cooper HS, Murthy SN, Shah RS and Sedergran DJ: Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 69:238–249. 1993.PubMed/NCBI

15 

Shen W and Durum SK: Synergy of IL-23 and Th17 cytokines: New light on inflammatory bowel disease. Neurochem Res. 35:940–946. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Hardenberg G, Steiner TS and Levings MK: Environmental influences on T regulatory cells in inflammatory bowel disease. Semin Immunol. 23:130–138. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Fuss IJ, Heller F, Boirivant M, Leon F, Yoshida M, Fichtner-Feigl S, Yang Z, Exley M, Kitani A, Blumberg RS, et al: Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. J Clin Invest. 113:1490–1497. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Heller F, Florian P, Bojarski C, Richter J, Christ M, Hillenbrand B, Mankertz J, Gitter AH, Bürgel N, Fromm M, et al: Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology. 129:550–564. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Hunter MM, Wang A and McKay DM: Helminth infection enhances disease in a murine TH2 model of colitis. Gastroenterology. 132:1320–1330. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Fuss IJ and Strober W: The role of IL-13 and NK T cells in experimental and human ulcerative colitis. Mucosal Immunol. 1(Suppl 1): S31–S33. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Matsumura K, Nakase H, Yamamoto S, Yoshino T, Takeda Y, Kasahara K, Ueno S, Uza N and Chiba T: Modulation of the Th1/Th2 balance by infliximab improves hyperthyroidism associated with a flare-up of ulcerative colitis. Inflamm Bowel Dis. 15:967–968. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Dohi T, Fujihashi K, Kiyono H, Elson CO and McGhee JR: Mice deficient in Th1- and Th2-type cytokines develop distinct forms of hapten-induced colitis. Gastroenterology. 119:724–733. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Sawa Y, Oshitani N, Adachi K, Higuchi K, Matsumoto T and Arakawa T: Comprehensive analysis of intestinal cytokine messenger RNA profile by real-time quantitative polymerase chain reaction in patients with inflammatory bowel disease. Int J Mol Med. 11:175–179. 2003.PubMed/NCBI

24 

Olsen T, Goll R, Cui G, Husebekk A, Vonen B, Birketvedt GS and Florholmen J: Tissue levels of tumor necrosis factor-alpha correlates with grade of inflammation in untreated ulcerative colitis. Scand J Gastroenterol. 42:1312–1320. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Alex P, Zachos NC, Nguyen T, Gonzales L, Chen TE, Conklin LS, Centola M and Li X: Distinct cytokine patterns identified from multiplex profiles of murine DSS and TNBS-induced colitis. Inflamm Bowel Dis. 15:341–352. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Fuss IJ: Is the Th1/Th2 paradigm of immune regulation applicable to IBD? Inflamm Bowel Dis. 14:S110–S112. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Bogaert S, Laukens D, Peeters H, et al: Differential mucosal expression of Th17-related genes between the inflamed colon and ileum of patients with inflammatory bowel disease. BMC Immunol. 11:612010. View Article : Google Scholar : PubMed/NCBI

28 

Liu ZJ, Yadav PK, Su JL, Wang JS and Fei K: Potential role of Th17 cells in the pathogenesis of inflammatory bowel disease. World J Gastroenterol. 15:5784–5788. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Heo YJ, Joo YB, Oh HJ, et al: IL-10 suppresses Th17 cells and promotes regulatory T cells in the CD4+ T cell population of rheumatoid arthritis patients. Immunol Lett. 127:150–156. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Kleczynska W, Jakiela B, Plutecka H, et al: Imbalance between Th17 and regulatory T-cells in systemic lupus erythematosus. Folia Histochem Cytobiol. 49:646–653. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Horiuchi A, Hayashi T, Kikuchi N, et al: Hypoxia upregulates ovarian cancer invasiveness via the binding of HIF-1α to a hypoxia-induced, methylation-free hypoxia response element of S100A4 gene. Int J Cancer. 131:1755–1767. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Chu W, Wan L, Zhao D, et al: Mild hypoxia-induced cardiomyocyte hypertrophy via up-regulation of HIF-1α-mediated TRPC signalling. J Cell Mol Med. 16:2022–2034. 2012. View Article : Google Scholar : PubMed/NCBI

33 

OGarra A and Vieira P: Regulatory T cells and mechanisms of immune system control. Nat Med. 10:801–805. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Veltkamp C, Ruhwald R, Giesem T, Autschbach F, Kaden I, Veltkamp R, Sartor RB and Stremmel W: CD4+CD25+ cell depletion from the normal CD4+ T cell pool prevents tolerance toward the intestinal flora and leads to chronic colitis in immunodeficient mice. Inflamm Bowel Dis. 12:437–446. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Oconnor W Jr, Kamanaka M, Booth CJ, Town T, Nakae S, Iwakura Y, Kolls JK and Flavell RA: A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol. 10:603–609. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Fiocchi C: Inflammatory bowel disease: evolutionary concepts in biology, epidemiology, mechanisms and therapy. Curr Opin Gastroenterol. 29:347–349. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Gupta S: Immune homeostasis: regulatory T cells (Treg) and molecules. J Clin Immunol. 28:617–618. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Unutmaz D and Pulendran B: The gut feeling of Treg cells: IL-10 is the silver lining during colitis. Nat Immunol. 10:1141–1143. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Bettelli E, Carrier Y, Cao W, et al: Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 441:235–238. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Lina C, Conghua W, Nan L and Ping Z: Combined treatment of etanercept and MTX reverses Th1/Th2, Th17/Treg imbalance in patients with rheumatoid arthritis. J Clin Immunol. 31:596–605. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Ben Aissa-Fennira F, Sassi A, Bouguerra A and Benammar-Elgaaied A: Immunoregulatory role for a public IgM idiotype in the induction of autoimmune diseases in Mycoplasma pneumoniae infection. Immunol Lett. 136:130–137. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Zhou L, Lopes JE, Chong MM, et al: TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 453:236–240. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Li MO and Flavell RA: TGF-beta: A master of all T cell trades. Cell. 134:392–404. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Ghoreschi K, Laurence A, Yang XP, et al: Generation of pathogenic T(H)17 cells in the absence of TGF-β signalling. Nature. 467:967–971. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Wang J, Pae M, Meydani SN and Wu D: Green tea epigallocatechin-3-gallate modulates differentiation of naïve CD4+ T cells into specific lineage effector cells. J Mol Med (Berl). 91:485–495. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Nutsch K and Hsieh C: When T cells run out of breath: the HIF-1α story. Cell. 146:673–674. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Jung JE, Kim HS, Lee CS, et al: STAT 3 inhibits the degradation of HIF-1alpha by pVHL-mediated ubiquitination. Exp Mol Med. 40:479–485. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Sun H, Zhang Y, Gao P, et al: Adiponectin reduces C-reactive protein expression and downregulates STAT3 phosphorylation induced by IL-6 in HepG2 cells. Mol Cell Biochem. 347:183–189. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Li Y, Yu C, Zhu WM, et al: Triptolide ameliorates IL-10-deficient mice colitis by mechanisms involving suppression of IL-6/STAT3 signaling pathway and down-regulation of IL-17. Mol Immunol. 47:2467–2474. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM and Stockinger B: TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 24:179–189. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Mangan PR, Harrington LE, OQuinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR and Weaver CT: Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 441:231–234. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Volpe E, Servant N, Zollinger R, Bogiatzi SI, Hupé P, Barillot E and Soumelis V: A critical function for transforming growth factor-beta, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses. Nat Immunol. 9:650–657. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM and Weaver CT: Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 6:1123–1132. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, Mazzinghi B, Parente E, Filì L, Ferri S, Frosali F, et al: Phenotypic and functional features of human Th17 cells. J Exp Med. 204:1849–1861. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Lubberts E: Th17 cytokines and arthritis. Semin Immunopathol. 32:43–53. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Komiyama Y, Nakae S, Matsuki T, Nambu A, Ishigame H, Kakuta S, Sudo K and Iwakura Y: IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol. 177:566–573. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS and Dong C: STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 282:9358–9363. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Dryden GW, Fernandez-Botran GR and Qazzaz HMH: EGCG reduces proinflammatory cytokine production and induces apoptosis in activated CD14+ macrophages, CD4+CD45+RO T cells, and mixed macrophage/T cell populations, but not CD4+CD45+RA T cells from IBD patients and controls. Gastroenterology. 140:S–838. 2011.

59 

Dryden GW, Lam A, Beatty K, Qazzaz HH and McClain CJ: A pilot study to evaluate the safety and efficacy of an oral dose of (−)-epigallocatechin-3-gallate-rich polyphenon E in patients with mild to moderate ulcerative colitis. Inflamm Bowel Dis. 19:1904–1912. 2013.PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 10 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu Z, Wei C, Zhang R, Yao J, Zhang D and Wang L: Epigallocatechin‑3‑gallate‑induced inhibition of interleukin‑6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice. Exp Ther Med 10: 2231-2238, 2015
APA
Xu, Z., Wei, C., Zhang, R., Yao, J., Zhang, D., & Wang, L. (2015). Epigallocatechin‑3‑gallate‑induced inhibition of interleukin‑6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice. Experimental and Therapeutic Medicine, 10, 2231-2238. https://doi.org/10.3892/etm.2015.2824
MLA
Xu, Z., Wei, C., Zhang, R., Yao, J., Zhang, D., Wang, L."Epigallocatechin‑3‑gallate‑induced inhibition of interleukin‑6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice". Experimental and Therapeutic Medicine 10.6 (2015): 2231-2238.
Chicago
Xu, Z., Wei, C., Zhang, R., Yao, J., Zhang, D., Wang, L."Epigallocatechin‑3‑gallate‑induced inhibition of interleukin‑6 release and adjustment of the regulatory T/T helper 17 cell balance in the treatment of colitis in mice". Experimental and Therapeutic Medicine 10, no. 6 (2015): 2231-2238. https://doi.org/10.3892/etm.2015.2824