Basic Research Open Access
Copyright ©The Author(s) 2003. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Nov 15, 2003; 9(11): 2533-2538
Published online Nov 15, 2003. doi: 10.3748/wjg.v9.i11.2533
Ameliorative effects of sodium ferulate on experimental colitis and their mechanisms in rats
Wei-Guo Dong, Shao-Ping Liu, Bao-Ping Yu, He-Sheng Luo, Jie-Ping Yu, Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, China
Dong-Fang Wu, Department of Pharmacy, Renmin Hospital of Wuhan University, 238 Jie-fang Road, Wuhan 430060, Hubei Province, China
Author contributions: All authors contributed equally to the work.
Supported by key Project in Scientific and Technological Researches of Hubei Province, No. 2001AA308B
Correspondence to: Professor Wei-Guo Dong, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, China. dongwg@public.wh.hb.cn
Telephone: +86-27-88054511
Received: May 13, 2003
Revised: May 23, 2003
Accepted: June 2, 2003
Published online: November 15, 2003

Abstract

AIM: To investigate the ameliorative effects of sodium ferulate (SF) on acetic acid-induced colitis and their mechanisms in rats.

METHODS: The colitis model of Sprague-Dawley rats was induced by intracolon enema with 8% (V/V) of acetic acid. The experimental animals were randomly divided into model control, 5-aminosalicylic acid therapy group and three dose of SF therapy groups. The 5 groups were treated intracolonically with normal saline, 5-aminosalicylic acid (100 mg•kg-1), and SF at the doses of 200, 400 and 800 mg·kg-1 respectively and daily (8: 00 am) for 7 d 24 h following the induction of colitis. A normal control group of rats clystered with normal saline instead of acetic acid was also included in the study. Pathological changes of the colonic mucosa were evaluated by the colon mucosa damage index (CMDI) and the histopathological score (HS). The insulted colonic mucosa was sampled for a variety of determinations at the end of experiment when the animals were sacrificed by decapitation. Colonic activities of myeloperoxidase (MPO) and superoxide dismutase (SOD), and levels of malondialdehyde (MDA) and nitric oxide (NO) were assayed with ultraviolet spectrophotometry. Colonic contents of prostaglandin E2 (PGE2) and thromboxane B2 (TXB2) were determined by radioimmunoassay. The expressions of inducible nitric oxide synthase (iNOS), cyclo-oxygenase-2 (COX-2) and nuclear factor kappa B (NF-κB) p65 proteins in the colonic tissue were detected with immunohistochemistry.

RESULTS: Enhanced colonic mucosal injury, inflammatory response and oxidative stress were observed in the animals clystered with acetic acid, which manifested as the significant increase of CMDI, HS, MPO activities, MDA and NO levels, PGE2 and TXB2 contents, as well as the expressions of iNOS, COX-2 and NF-κB p65 proteins in the colonic mucosa, although the colonic SOD activity was significantly decreased compared with the normal control (CMDI: 2.9 ± 0.6 vs 0.0 ± 0.0; HS: 4.3 ± 0.9 vs 0.7 ± 1.1; MPO: 98.1 ± 26.9 vs 24.8 ± 11.5; MDA: 57.53 ± 12.36 vs 9.21 ± 3.85; NO: 0.331 ± 0.092 vs 0.176 ± 0.045; PGE2: 186.2 ± 96.2 vs 42.8 ± 32.8; TXB2: 34.26 ± 13.51 vs 8.83 ± 3.75; iNOS: 0.365 ± 0.026 vs 0.053 ± 0.015; COX-2: 0.296 ± 0.028 vs 0.034 ± 0.013; NF-κB p65: 0.314 ± 0.026 vs 0.039 ± 0.012; SOD: 28.33 ± 1.17 vs 36.14 ± 1.91; P < 0.01). However, these parameters were found to be significantly ameliorated in rats treated locally with SF at the given dose protocols, especially at 400 mg·kg-1 and 800 mg·kg-1 doses (CMDI: 1.8 ± 0.8, 1.6 ± 0.9; HS: 3.3 ± 0.9, 3.1 ± 1.0; MPO: 63.8 ± 30.5, 36.2 ± 14.2; MDA: 41.84 ± 10.62, 37.34 ± 8.58; NO: 0.247 ± 0.042; 0.216 ± 0.033; PGE2: 77.2 ± 26.9, 58.4 ± 23.9; TXB2: 18.07 ± 14.83; 15.52 ± 8.62; iNOS:0.175 ± 0.018, 0.106 ± 0.019; COX-2: 0.064 ± 0.018, 0.056 ± 0.014; NF-κBp65: 0.215 ± 0.019, 0.189 ± 0.016; SOD: 32.15 ± 4.26, 33.24 ± 3.69; P < 0.05-0.01). Moreover, a therapeutic dose protocol of 800 mg·kg-1 SF was observed as effective as 100 mg·kg-1 of 5-ASA in the amelioration of colonic mucosal injury as evaluated by CMDI and HS.

CONCLUSION: Administration of SF intracolonically may have significant therapeutic effects on the rat model of colitis induced by acetic acid enema, which was probably due to the mechanism of antioxidation, inhibition of arachidonic acid metabolism and NF-κB expression.




INTRODUCTION

Inflammatory bowel disease (IBD) is a collection of chronic idiopathic inflammatory disorders of the intestine and/or colon. Although the pathophysiology of these disorders is not known with certainty, a growing body of experimental and clinical data suggested that this kind of chronic gut inflammation might be resulted from a maladjusted immune response to certain bacterial antigens[1-3]. This uncontrolled immune system activation results in a sustained overproduction of cytokines, eicosanoids, oxygen and nitrogen reactive species, which has been thought to be the major causative factors leading to intestinal and/or colonic injury and dysfunction in inflammatory bowel disease.

Based on this hypothesis, current therapeutic strategy for IBD has therefore focused on the use of anti-inflammatory agents, aminosalicylates and corticosteroids[1,4-9]. However, some notable limitations are found existing in the management of IBD patients and meanwhile some patients are refractory to these therapies. Long term use of glucocorticoids, for example, although effective in suppressing active inflammation, was associated with high rates of relapse and unacceptable toxicity[10-12]. On the other hand, 6-mercaptopurine and its prodrug azathioprine are effective in inducing and maintaining remission, however, a significant number of patients were resistant or intolerant to thiopurines[10]. All these suggest that seeking for more preferable drugs harboring better therapeutic results and less side-effects for the treatment of patients with IBD is urgently necessary.

Sodium ferulate (SF) is one of the effective components of Radix Angelica sinensis. Many researches have revealed that SF possesses plenty of beneficial pharmacological effects such as inhibiting the production of inflammatory mediators, improving endothelial function, relieving oxidative stress and safe in use[13-18]. We presumed that SF might be a possible candidate to be used for the treatment of IBD, which to our knowledge has not been investigated in previous studies. The present study was therefore conducted to confirm our hypothesis with an effort to demonstrate the therapeutic effects of sodium ferulate on experimental colonitis.

MATERIALS AND METHODS
Animals

Healthy Sprague-Dawley rats of both sexes, weighing 250 ± 30 g, from the Animal Center, Academy of Hubei preventive medical sciences were employed in the present study. The animals were fed on standard rat chow, allowed access to tap water ad libitum and acclimatized to the surroundings for one week prior to the experiments. The study protocol was in accordance with the guideline for animal research and was approved by the Ethical and Research Committee of the hospital.

Reagents

Acetic acid was obtained from Wuhan Chemical Corp. SF injection was provided by Pharmaceutical Factory of Zhongnan Hospital, Wuhan University (Lot. 20011219). 5-aminosalicylic acid (5-ASA) was purchased from Guoyi Pharmaceutical Ltd (Lot. 20029477). Polyclonal rabbit anti-rat-iNOS, COX-2 and NF-κB P65 were from Santa Cruz Company. S-P kits were obtained from Beijing Zhongshan Biological Technology CO Ltd. PGE2 and TXB2 radioimmunoassay kits were provided by Radioimmunity Institute of PLA General Hospital. MPO, SOD, MDA, NO detection kits were purchased from Nanjing Jiancheng Bioengineering Institute. Other reagents used in the present study were all of analytical grade.

Experimental protocol

The preparation of rat model of colitis was described in the literature[19,20]. Briefly, after rats were anesthetized with ether, a flexible plastic catheter with an outer diameter of 2 mm were inserted rectally into the colon with the aim to place the catheter tip at 8 cm proximal to the anus. Twenty seconds following enema with 2 mL of 8% (V/V) acetic acid through the catheter, colon lumen was rinsed twice with 5 mL of normal saline. The experimental animals were randomly divided into normal control, model control, 5-aminosalicylic acid therapy and three dose SF therapy groups, and treated intracolonically with normal saline, normal saline, 5-aminosalicylic acid (100 mg•kg-1), and SF 200, 400 and 800 mg•kg-1 respectively and daily (8: 00 am) for 7 d 24 h following the induction of colitis. Then the colon mucosa damage index (CMDI) and the histopathological score (HS) were evaluated and the colon mucosa was sampled for a variety of determinations and observations after the animals were sacrificed by decapitation.

Assessment of CMDI and HS

The colon segment taken from 10 cm proximal to anus of the sacrificed rats was excised longitudinally, rinsed by saline buffer and fixed on a wax block. CMDI and HS were assessed respectively by two independent researchers. The scoring formula for assessment of CMDI was reported in previous literature[20]. That is, 0: normal mucosa, no damage on the mucosal surface; 1: mild hyperemia and edema, no erosion or ulcer on the mucosal surface; 2: moderate hyperemia and edema, erosion appearing on the mucosal surface; 3: severe hyperemia and edema, necrosis and ulcer appearing on the mucosal surface with the major ulcerative area extending less than 1 cm; 4: severe hyperemia and edema, necrosis and ulcer appearing on the mucosal surface with the major ulcerative area extending more than 1 cm. For histopathological examination, the colonic samples were fixed in 4% neutral buffered paraformaldehyde overnight and 4-μm thick sections were routinely prepared and stained with haematoxylin and eosin. The colonic pathological changes were observed and evaluated by two independent researchers with a modified histopathological score formula[19] as: (1) Infiltration of acute inflammatory cells: 0, without; 1, mild; 2, severe; (2) Infiltration of chronic inflammatory cells: 0, without; 1, mild; 2, severe; (3) Fibrin deposition: 0, negative; 1, positive; (4) Submucosal edema: 0, without; 1, focal; 2, diffuse; (5) Necrosis of epithelial cells: 0, without; 1, focal; 2, diffuse and (6) Mucosal ulcer: 0, negative; 1, positive.

Determination of colonic MPO, SOD activities and MDA, NO contents

For determination of colonic MPO activity, the sampled tissues were homogenized (50 g•L-1) in 50 mmol•L-1 ice-cold potassium phosphate buffer (pH6.0) containing 0.5% of hexadecyltrimethylammoninm bromide. The homogenate was frozen and thawed thrice, then centrifuged at 4000 rpm for 20 min at 4 °C. The MPO activity in the supernatant was measured by the assay kit according to its provider’s instructions. The colonic samples for the determination of SOD activity, MDA and NO contents were homogenized in ice-cold PBS (pH7.4) and centrifuged at 3000 rpm for 10 min at 4 °C. The resulting supernatant was stored at 20 °C until analysis with corresponding assay kits according to the manufacturer’s guide.

Measurement of colonic PGE2 and TXB2 level

Colonic specimens used for the assay of arachidonic acid metabolites were prepared according to the protocol by Raab et al[21] and Taniguchi et al[22], and their levels of PGE2 and TXB2 were measured by using the corresponding radioimmunoassay kits following the manufacturer’s instructions.

Detection of colonic iNOS, COX-2 and NF-κB p65 expression

The immunohistochemical methods for formalin-fixed and paraffin-embedded sections were described previously[23]. The determinations of colonic iNOS, COX-2 and NF-κB p65 expression were all performed with S-P technique following the recommendations of assay kit providers. Briefly, polyclonal rabbit anti-rat-iNOS, COX-2 and NF-κB p65 were diluted with PBS to 1:150, 1:120, 1:200 respectively and used as the primary antibody in the corresponding detections. For each of these determinations, dewaxed sections were incubated first with the primary antibody overnight at 4 °C after antigen retrieval. The binding of antibodies to their antigenic sites in the tissue sections was further amplified with biotinylated goat anti-rabbit antibody and followed by reaction with 3,3’-diaminobenzidine. Sections prepared by substituting PBS for the primary antibody served as the negative control. iNOS, COX-2 or NF-κB p65 negatively expressed cells manifested as blue-stained nuclei while the positive cells as brown or dark brown cytoplasm and/or cell membrane. Expressions of these target proteins were semiquantitated respectively with automatic image analyzer (Nikon, Japan) and HPIAS-2000 image analyzing program, in which the average value of positive cell’s absorbance (A) in ten randomly selected high power fields (400 ×) for each section was used for the comparison of the target protein expressions.

Statistical analysis

Experimental results were expressed as ¯x±s. Statistical comparisons between groups were made by ANOVA followed by Student’s t test. P value less than 0.05 was considered statistically significant.

RESULTS
Inflammatory changes of the colonic mucosa

CMDI, HS and MPO activities were the main parameters for evaluating the degree of colonic inflammation in inflammatory bowel disease. Compared with the normal control, these parameters were all significantly increased in the colonic mucosa of the model control induced by acetic acid (P < 0.01). After the model rats were treated with SF (400, 800 mg•kg-1) or 5-ASA (100 mg•kg-1) as described in the experimental protocol, these elevated parameters were significantly ameliorated (P < 0.05 or 0.01) as shown in Table 1, in which a therapeutic dose protocol of 800 mg•kg-1 SF was observed as effective as 100 mg•kg-1 of 5-ASA in the treatment of this rat model of colitis.

Table 1 Effects of sodium ferulate on CMDI, HS and MPO activity in the colon tissue of acetic acid-induced rats colitis (n = 8, ¯x ± s).
GroupDose (mg•kg-1)CMDIHSMPO (U·g-1)
Normal-0.0 ± 0.00.7 ± 1.124.8 ± 11.5
Model-2.9 ± 0.6 d4.3 ± 0.9 d98.1 ± 26.9 d
5-ASA1001.6 ± 0.9b3.4 ± 0.5a29.6 ± 10.8b
SF2002.0 ± 0.8a3.6 ± 1.279.5 ± 38.4
SF4001.8 ± 0.8b3.3 ± 0.9a63.8 ± 30.5a
SF8001.6 ± 0.9b3.1 ± 1.0a36.2 ± 14.2b
Colonic oxidative alterations

Prominent oxidative stress in colonic mucosa was induced by enema of acetic acid in the model controls as shown by the significant elevation of colonic MDA and NO contents and decrease of colonic SOD activities (Table 2). These oxidative abnormalities in colonic mucosa were obviously ameliorated by the treatment with SF at the dose protocols of 200, 400, 800 mg•kg-1, which manifested as the significant reduction of colonic MDA content and the increase of SOD activity compared with the model control rats (P < 0.05-0.01). Furthermore, a significant improvement for the elevated colonic NO content was also noted in animals treated with SF at the dose protocol of 400 or 800 mg•kg-1 (P < 0.05-0.01) as shown in Table 2.

Table 2 Effects of sodium ferulate on SOD activity, MDA and NO contents in the colon tissue of acetic acid-induced rats colitis (n = 8, ¯x ± s).
GroupDose (mg·kg-1)SOD (kU·g-1)MDA (nmol·g-1)NO (nmol·g-1)
Normal-36.14 ± 1.919.21 ± 3.85176 ± 45
Model-28.33 ± 1.17 d57.53 ± 12.36 d331 ± 92 d
5-ASA10032.74 ± 3.52b31.85 ± 11.72b244 ± 51a
SF20031.63 ± 3.83a43.25 ± 13.47a256 ± 54
SF40032.15 ± 4.26a41.84 ± 10.62a247 ± 42a
SF80033.24 ± 3.69b37.34 ± 8.58b216 ± 33b
Effects of SF on the colonic PGE2, TXB2 levels

After the rats were insulted by enema of acetic acid, their colonic contents of PGE2 and TXB2 were significantly increased compared with the normal control (P < 0.01). A significant amelioration of PGE2 contents was observed in animals treated with SF at all dose protocols (P < 0.05-0.01). However, significant reduction of colonic TXB2 contents compared with the model control was only seen in rats treated with SF at the dose protocol of 400 and 800 mg•kg-1 (P < 0.05-0.01) as shown in Table 3.

Table 3 Effects of sodium ferulate on the content of PGE2 and TXB2 in the colon tissue of acetic acid-induced rats colitis (n = 8, ¯x ± s).
GroupDose (mg·kg-1)PGE2 (ng·g-1)TXB2 (ng·g-1)
Normal-42.8 ± 32.88.83 ± 3.75
Model-186.2 ± 96.2 d34.26 ± 13.51 d
5-ASA10067.0 ± 37.7b18.53 ± 14.26a
SF20090.7 ± 52.3a23.21 ± 12.46
SF40077.2 ± 26.9b18.07 ± 14.83a
SF80058.4 ± 23.9b15.52 ± 8.62b
SF reduces the expression of colonic NOS, COX-2 and NF-κB p65

As shown in Figure 1, Figure 2 and Figure 3, the expressions of colonic NOS, COX-2 and NF-κB p65 proteins were observed to be significantly increased in animals clystered with acetic acid compared with the normal controls (P < 0.01), which were ameliorated significantly in a dose-dependent manner as the animals treated with SF at the given doses (P < 0.05-0.01).

Figure 1
Figure 1 Effects of SF on the expressions of iNOS, COX-2 and NF-κB P65 in the colonic tissue of rats colitis induced by acetic acid (n = 8, ¯x ± s). bP < 0.01, vs control group; cP < 0.05, dP < 0.01, vs model group.
Figure 2
Figure 2 Expressions of iNOS (A), COX-2 (B) and NF-κB p65 (C) in the inflammatory areas of colonic tissue from rats with colitis induced by acetic acid enema, respectively. The strongly positive signal were found. SP stain × 400.
Figure 3
Figure 3 Expressions of iNOS (A), COX-2 (B) and NF-κB p65 (C) were significantly decreaed in the colonic tissue from rats with colitis induced by acetic acid enema after treasted with 800 mg•kg-1 ASP, respectively. The weakly positive staining could be found. SP stain × 400.
DISCUSSION

Induction of colitis by acetic acid (AA) in rats is the classical method to produce an experimental model of inflammatory bowel disease (IBD). Several major causative factors in the initiation of human IBD such as exorbitant oxidative stress, enhanced vasopermeability, prolonged neutrophils infiltration and increased production of inflammatory mediators were all involved in the induction of this animal model[19,20,24]. It is therefore acknowledged nowadays that this experimental model is suitable for the investigation of IBD pathogenesis and evaluation the therapeutic agents of this disease.

In the present study, we employed this rat model to make sure whether or not the ameliorative effects of SF on the experimental colitis existed as assessed by CMDI, HS and MPO activities that were usually regarded as the main parameters to evaluate the severity of colonic inflammation in inflammatory bowel disease[19,20]. Compared with the normal control, these parameters were all significantly increased in the colonic mucosa of the model control animals induced by acetic acid (P < 0.01). However, these elevated parameters were significantly ameliorated (P < 0.05 or 0.01) as shown in Table 1 after the model rats were treated with SF (400, 800 mg•kg-1) or 5-ASA (100 mg•kg-1) as described in the experimental protocol, in which a therapeutic dose protocol of 800 mg•kg-1 SF was observed as effective as 100 mg•kg-1 5-ASA in the treatment of this rat model of colitis. The results confirmed our previous speculation and strongly suggested that SF might serve as an alternative modality for the treatment of inflammatory bowel disease.

Although the pathogenesis is not known with certainty, some hypotheses have been proposed as the major causative factors contributing to inflammatory bowel disease. A large number of studies have revealed that the increase of oxidative stress and iNOS activity was a notable feature of IBD, which resulted in a pathological cascade of free radical reactions and further yielding more oxidative free radicals such as peroxynitrite (ONOO-) to impair the structure and function of cells[2,25-27]. Meanwhile, excessive NO could dilate vasculature and enhance vasopermeability, as well as inactivate the activity of antioxidases such as SOD, CAT, and GSH-Px by means of reacting with hydrosulfide group (-SH) in the enzymes. Some oxidants have been known to modulate the expression of a variety of genes that are involved in the immune and inflammatory responses, which lead to the apoptosis of intestinal epithelial cells, cascades of inflammatory response and the disruption of integrity and function of the intestinal mucosa[28-30].

Abnormal metabolism of arachidonic acid is another vital factor in the IBD pathogenesis. As the crucial synthetase in the arachidonic acid metabolic pathway, COX-2 could be activated to produce excessive PGE2 and TXB2, two important inflammatory mediators, in the inflammatory bowel disease, which contribute to the bowel hyperemia, edema and even dysfunction. In addition, TXB2 could also induce platelet aggregation, vasoconstriction and microthrombosis, aggravating the inflammatory reaction[31-35]. Administration of either COX-2 or thromboxane synthase inhibitors has been shown to be useful for the treatment of IBD[32-34].

Recently, plenty of literatures reported that NF-κB played a critical role in the early transcriptional changes of various immunoregulatory genes[28,36], whose activation and increased expression have been demonstrated to be involved in the pathogenesis of inflammatory bowel disease[37]. Activated NF-κB could promote the production of various inflammatory factors, chemotaxins, cytokines, and adhesive factors, and the expression of iNOS and COX-2, which interact further with each other and lead to an uncontrollable cascade of inflammatory response[38-41]. NF-κB could also activate anti-apoptotic genes including TNF receptor related genes (TRAF1, TRAF2), Bcl-2 homologens (A1/Bfl-1, IEX-1L), and repress the apoptosis of some inflammatory cells such as neutrophils and activated macrophages, thereby elongating and worsening tissue inflammatory injury[42-45]. In the NF-κB family, p65 is the major functional subunit, and an antisense oligonucleotide to NF-kappa B p65 has been shown to ameliorate inflammation even after induction of colitis[1].

To elucidate the mechanisms underlying the therapeutic effects of SF on this rat model of colitis, we observed simultaneously the changes of these oxidative and inflammatory variables mentioned above in the colonic tissue after SF therapy. The results revealed that the SOD activity, MDA and NO levels, PGE2 and TXB2 contents, as well as the expressions of iNOS, COX-2 and NF-κB p65 proteins in the colonic mucosa were significantly ameliorated in the rats treated locally with SF at the given dose protocols, especially at the 400 mg•kg-1 and 800 mg•kg-1 doses, compared with that in model control animals (P < 0.05-0.01). Taken together, these observations suggest the anti-oxidative stress and anti-inflammatory response are the fundamentals of SF action in the therapy of IBD, although the ameliorating effects of SF might be involved in other multiple mechanisms.

In summary, the results of this study show that intracolonic treatment with SF at 400 mg·kg-1 and 800 mg·kg-1 dose protocols can ameliorate the pathological changes of experimental colitis in rats, which suggests that SF may serve as an effective therapeutic agent for the treatment of IBD.

Footnotes

Edited by Zhu L and Wang XL

References
1.  Hibi T, Inoue N, Ogata H, Naganuma M. Introduction and overview: recent advances in the immunotherapy of inflammatory bowel disease. J Gastroenterol. 2003;38 Suppl 15:36-42.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Pavlick KP, Laroux FS, Fuseler J, Wolf RE, Gray L, Hoffman J, Grisham MB. Role of reactive metabolites of oxygen and nitrogen in inflammatory bowel disease. Free Radic Biol Med. 2002;33:311-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 296]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
3.  Lakatos L. Immunology of inflammatory bowel diseases. Acta Physiol Hung. 2000;87:355-372.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Travis S. Recent advances in immunomodulation in the treatment of inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2003;15:215-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
5.  Tuvlin JA, Kane SV. Novel therapies in the treatment of ulcerative colitis. Expert Opin Investig Drugs. 2003;12:483-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
6.  Goh J, O'Morain CA. Review article: nutrition and adult inflammatory bowel disease. Aliment Pharmacol Ther. 2003;17:307-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 148]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
7.  Hanauer SB. Update on medical management of inflammatory bowel disease: ulcerative colitis. Rev Gastroenterol Disord. 2001;1:169-176.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Kane SV. Novel therapies in the treatment of ulcerative colitis. Expert Opin Investig Drugs. 2001;10:1223-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.0]  [Reference Citation Analysis (0)]
9.  Das KM, Farag SA. Current medical therapy of inflammatory bowel disease. World J Gastroenterol. 2000;6:483-489.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Schröder O, Stein J. Low dose methotrexate in inflammatory bowel disease: current status and future directions. Am J Gastroenterol. 2003;98:530-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
11.  Holtmann MH, Galle PR, Neurath MF. Immunotherapeutic approaches to inflammatory bowel diseases. Expert Opin Biol Ther. 2001;1:455-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
12.  Rampton D. Management of difficult inflammatory bowel disease: where are we now. World J Gastroenterol. 2000;6:315-323.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Kikuzaki H, Hisamoto M, Hirose K, Akiyama K, Taniguchi H. Antioxidant properties of ferulic acid and its related compounds. J Agric Food Chem. 2002;50:2161-2168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 716]  [Cited by in F6Publishing: 617]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
14.  Ogiwara T, Satoh K, Kadoma Y, Murakami Y, Unten S, Atsumi T, Sakagami H, Fujisawa S. Radical scavenging activity and cytotoxicity of ferulic acid. Anticancer Res. 2002;22:2711-2717.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Zhang ZH, Yu SZ, Li GS, Zhao BT. Influence of sodium ferulate on human netrophil-derived oxygen metabolites. Zhongguo Yaolixue Tongbao. 2001;17:515-517.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Li YQ, Zhang JS, Cai HW. Influence of sodium ferulate on MDA, SOD, ET and NO during myocardial ischemia andreperfusion injury. Zhonghua Mazuixue Zazhi. 1998;18:688-690.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Xu LN, Yu WG, Tian JY, Liu QY. [Effect of sodium ferulate on arachidonic acid metabolism]. Yaoxue Xuebao. 1990;25:412-416.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Wang Z, Gao YH, Huang RS, Zhu GQ. [Sodium ferulate is an inhibitor of thromboxane A2 synthetase]. Zhongguo Yaoli Xuebao. 1988;9:430-433.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Millar AD, Rampton DS, Chander CL, Claxson AW, Blades S, Coumbe A, Panetta J, Morris CJ, Blake DR. Evaluating the antioxidant potential of new treatments for inflammatory bowel disease using a rat model of colitis. Gut. 1996;39:407-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 211]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
20.  Deng CH, Xia B, Chen DJ, Zhou Y, Gong TT, Guao ZQ. The mucosa protective effects of superoxide dismutase on ratscolitis induced by acetic acid. Zhongguo Bingli Shengli Zazhi. 1994;10:23-25.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Raab Y, Sundberg C, Hällgren R, Knutson L, Gerdin B. Mucosal synthesis and release of prostaglandin E2 from activated eosinophils and macrophages in ulcerative colitis. Am J Gastroenterol. 1995;90:614-620.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Taniguchi T, Tsukada H, Nakamura H, Kodama M, Fukuda K, Tominaga M, Seino Y. Effects of a thromboxane A2 receptor antagonist in an animal model of inflammatory bowel disease. Digestion. 1997;58:476-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
23.  Linnoila RI, Jensen SM, Steinberg SM, Mulshine JL, Eggleston JC, Gazdar AF. Peripheral airway cell marker expression in non-small cell lung carcinoma. Association with distinct clinicopathologic features. Am J Clin Pathol. 1992;97:233-243.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Li L, Wang ZL, Ke JT, Zhang M, Shao JF, Zhong CN. Select animal models for experimental colitis model. Shijie Huaren Xiaohua Zazhi. 2001;9:584-585.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Krieglstein CF, Cerwinka WH, Laroux FS, Salter JW, Russell JM, Schuermann G, Grisham MB, Ross CR, Granger DN. Regulation of murine intestinal inflammation by reactive metabolites of oxygen and nitrogen: divergent roles of superoxide and nitric oxide. J Exp Med. 2001;194:1207-1218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 195]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
26.  Zhou JF, Cai D, Zhu YG, Yang JL, Peng CH, Yu YH. A study on relationship of nitric oxide, oxidation, peroxidation, lipoperoxidation with chronic chole-cystitis. World J Gastroenterol. 2000;6:501-507.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Wang QG, He LY, Chen YW, Hu SL. Enzymohistochemical study on burn effect on rat intestinal NOS. World J Gastroenterol. 2000;6:421-423.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Jourd'heuil D, Morise Z, Conner EM, Grisham MB. Oxidants, transcription factors, and intestinal inflammation. J Clin Gastroenterol. 1997;25 Suppl 1:S61-S72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
29.  Banan A, Fields JZ, Zhang Y, Keshavarzian A. iNOS upregulation mediates oxidant-induced disruption of F-actin and barrier of intestinal monolayers. Am J Physiol Gastrointest Liver Physiol. 2001;280:G1234-G1246.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Huycke MM, Abrams V, Moore DR. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis. 2002;23:529-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 288]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
31.  McCowen KC, Ling PR, Bistrian BR. Arachidonic acid concentrations in patients with Crohn disease. Am J Clin Nutr. 2000;71:1008.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Carty E, Macey M, McCartney SA, Rampton DS. Ridogrel, a dual thromboxane synthase inhibitor and receptor antagonist: anti-inflammatory profile in inflammatory bowel disease. Aliment Pharmacol Ther. 2000;14:807-817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
33.  Karmeli F, Cohen P, Rachmilewitz D. Cyclo-oxygenase-2 inhibitors ameliorate the severity of experimental colitis in rats. Eur J Gastroenterol Hepatol. 2000;12:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
34.  Kankuri E, Vaali K, Korpela R, Paakkari I, Vapaatalo H, Moilanen E. Effects of a COX-2 preferential agent nimesulide on TNBS-induced acute inflammation in the gut. Inflammation. 2001;25:301-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
35.  Wu JX, Xu JY, Yuan YZ. Effect of emodin and sandostatin on metabolism of eicosanoids in acute necrotizing pancreatitis. World J Gastroenterol. 2000;6:293-294.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Shi XZ, Lindholm PF, Sarna SK. NF-kappa B activation by oxidative stress and inflammation suppresses contractility in colonic circular smooth muscle cells. Gastroenterology. 2003;124:1369-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 67]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
37.  Gan H, Ouyang Q, Chen Y, Xia Q. [Activation of nuclear factor-kappaB and effects of anti-inflammatory treatment thereon in intestinal mucosa of patients with ulcerative colitis]. Zhonghua Yixue Zazhi. 2002;82:384-388.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Surh YJ, Chun KS, Cha HH, Han SS, Keum YS, Park KK, Lee SS. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001;480-481:243-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1162]  [Cited by in F6Publishing: 1118]  [Article Influence: 48.6]  [Reference Citation Analysis (1)]
39.  Yamamoto Y, Gaynor RB. Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. J Clin Invest. 2001;107:135-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1137]  [Cited by in F6Publishing: 1119]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
40.  Jobin C, Sartor RB. NF-kappaB signaling proteins as therapeutic targets for inflammatory bowel diseases. Inflamm Bowel Dis. 2000;6:206-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 90]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
41.  Wulczyn FG, Krappmann D, Scheidereit C. The NF-kappa B/Rel and I kappa B gene families: mediators of immune response and inflammation. J Mol Med (Berl). 1996;74:749-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 212]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
42.  Sahnoun Z, Jamoussi K, Zeghal KM. [Free radicals and antioxidants: physiology, human pathology and therapeutic aspects (part II)]. Therapie. 1998;53:315-339.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Petranka J, Wright G, Forbes RA, Murphy E. Elevated calcium in preneoplastic cells activates NF-kappa B and confers resistance to apoptosis. J Biol Chem. 2001;276:37102-37108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
44.  Potoka DA, Upperman JS, Nadler EP, Wong CT, Zhou X, Zhang XR, Ford HR. NF-kappaB inhibition enhances peroxynitrite-induced enterocyte apoptosis. J Surg Res. 2002;106:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
45.  Bengoechea-Alonso MT, Pelacho B, Osés-Prieto JA, Santiago E, López-Moratalla N, López-Zabalza MJ. Regulation of NF-kappaB activation by protein phosphatase 2B and NO, via protein kinase A activity, in human monocytes. Nitric Oxide. 2003;8:65-74.  [PubMed]  [DOI]  [Cited in This Article: ]