Journal List > Korean J Physiol Pharmacol > v.18(4) > 1025952

Randhawa, Singh, Singh, and Jaggi: A Review on Chemical-Induced Inflammatory Bowel Disease Models in Rodents

Abstract

Ulcerative colitis and Crohn's disease are a set of chronic, idiopathic, immunological and relapsing inflammatory disorders of the gastrointestinal tract referred to as inflammatory bowel disorder (IBD). Although the etiological factors involved in the perpetuation of IBD remain uncertain, development of various animal models provides new insights to unveil the onset and the progression of IBD. Various chemical-induced colitis models are widely used on laboratory scale. Furthermore, these models closely mimic morphological, histopathological and symptomatical features of human IBD. Among the chemical-induced colitis models, trinitrobenzene sulfonic acid (TNBS)-induced colitis, oxazolone induced-colitis and dextran sulphate sodium (DSS)-induced colitis models are most widely used. TNBS elicits Th-1 driven immune response, whereas oxazolone predominantly exhibits immune response of Th-2 phenotype. DSS-induced colitis model also induces changes in Th-1/Th-2 cytokine profile. The present review discusses the methodology and rationale of using various chemical-induced colitis models for evaluating the pathogenesis of IBD.

ABBREVIATIONS

IBD

inflammatory bowel disease

TNBS

2, 4, 6-trinitrobenzene sulfonic acid

DSS

dextran sodium sulphate

NSAIDs

nonsteroidal anti-inflammatory drugs

PGPS

peptidoglycan-polysaccharide

DAI

disease activity index

PG-APS

peptidoglycan polysaccharide from group A streptococci

NF-κB

nuclear factor-κB

ICAM-1

intercellular adhesion molecule 1

TNF-α

tumour necrosis factor-α

ppm

parts per million

INTRODUCTION

Inflammatory bowel disease (IBD) is a chronic, remitting and relapsing inflammatory disorder encompassing ulcerative colitis and Crohn's disease. Now IBD has become almost a global disease affecting masses of almost all ages including the pediatric population [1,2]. Although, the etiology of IBD is not yet fully clarified, there is now general consensus that etiologic factors of IBD possibly include genetic predisposition, immunologic abnormalities (disturbances in the innate and adaptive immune responses), and environmental influences that eventually result in colonic inflammation [3,4,5]. Both disorders have certain distinct and overlapping immunological, histopathological and clinical features. Ulcerative colitis is featured by inflammatory lesions usually affecting the large intestine, involving the rectum (proctitis), extending proximally to cover the sigmoid colon (proctosigmoiditis), descending colon (leftsided colitis), and the entire colon (pancolitis) [6]. Conversely, Crohn's disease can affect any part of gastrointestinal tract, but the most commonly affected regions include terminal ileum or the perianal region. Clinically, IBD is characterized by severe diarrhea, bleeding, abdominal pain, fluid and electrolyte loss reflecting the underlying inflammatory process. Crohn's disease is characterized by increased IL-12, IFN-γ, TNF-α, IL-2 production which indicates predominant activation of immune response of Th1-phenotype [7,8,9,10]. On the other hand, ulcerative colitis portrays Th-2 driven immune response as indicated by marked increase in IL-4, IL-5, IL-10 and IL-13 levels [7,11,12,13,14].
Furthermore, ulcerative colitis is characterized by significant thickening and dense infiltration of neutrophils, monocytes, macrophages, T cells typically on the mucosal layer of the bowel wall. On the contrary, Crohn's disease is featured by significant thickening as well as infiltration of macrophages, monocytes and T cells on the submucosal layer of the bowel wall. Besides, the presence of paracolonic lymph nodes is a unique feature to distinguish Crohn's disease from ulcerative colitis [15].
A convenient approach to study the pathogenesis and complexity of human IBD is to induce IBD in animals. Animal models of IBD are indispensible for the proper understanding of histopathological and morphological changes in the intestinal tract. Consequently, animal models play pivotal role in the development of novel therapeutic drug to cure IBD and dissect the possible mechanism of action of a particular drug. Various animal models have been established to study IBD; these broadly include chemical-induced colitis model, adoptive transfer model, model of spontaneous colitis, genetically engineered/transgenic animal model. Among these, the widely used models include chemical-induced colitis model that is sub-categorized into trinitrobenzene sulfonic acid (TNBS)-induced colitis [16,17,18], dextran sodium sulphate (DSS)-inducedcolitis [19,20,21], oxazolone-induced colitis [22,23,24], acetic acid-induced colitis [25,26,27,28], nonsteroidal anti-inflammatory drugs (NSAIDs)-induced colitis [29,30], carrageenan-induced colitis model [31,32], peptidoglycan-polysaccharide (PGPS)-induced colitis [33,34] (Table 1). Amongst the various models, chemical models are extensively used, as they offer reproducibility and ease of development. The present review discusses the methodology and rationale of using various chemical-induced colitis models for evaluating the pathogenesis of IBD.

CHEMICAL INDUCED MODELS OF CHRONIC INTESTINAL INFLAMMATION

2, 4, 6-Trinitrobenzene sulfonic acid (TNBS)

TNBS elicits cell-mediated immune responses and induces transmural inflammation in the gut with morphological and histopathological features similar to those of human IBD [18,35]. TNBS induces diffuse colonic inflammation, which is characterized by increased leukocyte infiltration, edema, and ulceration [36]. It is very well reported that administration of TNBS is associated with predominant activation of Th1-mediated immune response manifested by dense infiltration of local CD4+T cells. Thus, based on inclination towards Th1 immune response which involves IL-12 and TNF-α as effector cytokines, this model has been specifically related to Crohn's Disease in humans [16,37,38,39,40]. Hence, it is a very useful model and is frequently used in studying many aspects of gut inflammation including cytokine secretion pattern, cell adhesion and immuno-therapy. However, studies with IFN-γ-/- mice on a BALB/C background showed that in these mice TNBS colitis may be associated with a TH2-mediated colonic patch hypertrophy [41]. Furthermore, TNBS-induced colitis model is also frequently used as post inflammatory irritable bowel syndrome (PI-IBS) model as the major features of PI-IBS such as visceral hypersensitivity, dysfunction of motility, and alteration in permeability or secretion are also exhibited in TNBS model [42]. TNBS results in intestinal inflammation which is not the consequence of this chemical per se, but rather from delayed hypersensitivity as TNBS haptenizes colonic autologous/microbial proteins rendering them immunogenic to host immune system. In fact, TNBS-induced colitis is a hapten-induced colitis model in which Th1-mediated immune response involving various cytokines including IL-12 and TNF-α serving as effector cytokine which result in transmural infiltration and inflammation [17,43,44].
Experimentally, TNBS is dissolved in alcohol and is delivered intrarectally in rodents to induce colitis. Alcohol not only serves as a solvent or carrier, but also aids in inducing gut inflammation by breaking the mucosal barrier [39,43]. Various researchers have demonstrated that a 12~24 hr fast should precede induction of anesthesia which may be induced using ether/halothane/chloral hydrate etc. The most common method adopted for the induction of colitis in rats involves usage of 10 mg TNBS mingled in 0.25 ml of 50% ethanol and thereby, instillation in male/female Wistar rats using a medical-grade polyurethane catheter (external diameter 2 mm) for enteral feeding approximately 8 cm proximal to anal verge. After instillation of the hapten, head down position is maintained in order to prevent leakage and for even distribution of the hapten for 1~3 minutes. After 2~6 days of colitis induction, decapitation of rats is carried out for assessing degree of colonic inflammation using various histological and immunohistochemical techniques [17,18,45]. Different researchers have modified the procedure by varying the dose and volume of TNBS solution along with change in alcohol concentration in order to induce IBD of varying degree in Sprague Dawley or Wistar rats of either sex. These include intracolonic administration of 0.8 ml of 5% TNBS in 50% (v/v) ethanol [46]; 1 ml of 3% TNBS in 40% ethanol [36] and 0.5 ml of TNBS (100 mg/kg) [47]. Additionally, Qin et al compared the influence of variation in TNBS dose, ethanol concentration, depth of the site of instillation in TNBS induces PI-IBS. TNBS administered to rats at different doses (5, 10, 20 mg/0.8 ml per rat) gave an indication that TNBS induces acute inflammation and damage in a dose dependent manner with high and median doses being more effective. On varying alcohol concentration between 25% and 50% alcoholic concentration, it was found that the pathological score i.e. inflammation and visceral hypergelsia was more significant in TNBS-50% ethanol treated rats. Furthermore, it was revealed that instillation of 5 mg/kg dose of TNBS at a depth of 8 cm/4 cm produced similar pattern and severity of colonic inflammation [42]. Study conducted by Yang et al stated that recurrent ulcerative colitis model can also be induced in Wistar rats by instilling TNBS (100 mg/kg) into the colon through an obtuse cannula. This is followed by a second instillation with TNBS at a dose of 37.5 mg/kg into the colon 14 days after the first induction of colitis leading to generation of recurrent model of ulcerative colitis [48].
In mice, TNBS colitis was initially described in SJL/J mice, which is a mouse strain with high susceptibility for the induction of colitis. But now days, various other mouse strains are also frequently used for development of colitis which include BALB/C, C57BL/6 etc. Generally, it involves rectal application of low dose (100 µl of 0.5 mg TNBS in 50% ethanol) for induction of colitis that may result in a chronic transmural colitis with severe diarrhea, weight loss, and rectal prolapse, an illness that mimics some characteristics of Crohn's disease in humans [39]. Moreover, TNBS also induces significant changes in the morphology, mechanical properties and pharmacological response of circular muscle layer of distal colon as compared to proximal counterpart in mice to mimic human ulcerative colitis [35]. The typical method adopted for inducing colitis in C57BL/6 mice involves intrarectal administration of 200 mg/kg TNBS dissolved in 30% ethanol via a catheter approximately 3~4 cm proximal to anus. After 3 days mice are sacrificed to carry out histological examination of colon tissues [44]. Another researcher reported that colitis can be induced by instilling 20 mg of TNBS dissolved in 0.4 ml of 50% ethanol aqueous solution into the colon lumen [35]. TNBS results in infiltration of inflammatory cells within 2 hrs after administration, but typical signs of chronic inflammation develop after 48 hrs [35].
The exact mechanisms responsible for TNBS-induced IBD are poorly understood. Various scientists proposed different mechanisms for explaining the pathophysiological features of TNBS induced IBD. It is reported that L-type Ca2+ channel currents are down-regulated, whereas adenosine triphosphate (ATP)-sensitive K+ (KATP) channels are up-regulated in gastrointestinal smooth muscle cells after administering TNBS to mice, which induces hyper-polarization of the gastrointestinal smooth muscle cells and thus results in reduced colonic contractility [35].

Dextran Sodium Sulphate (DSS)

Dextran sulphate sodium (DSS)-induced colitis is a reproducible model that morphologically and symptomatically resembles ulcerative colitis in humans [49,50]. Additionally, DSS model of colitis is one of the widely used models as it can be easily developed owing to the wide availability and low cost of DSS. Some researchers have suggested that DSS mainly affects the large intestine i.e. middle and distal third of large intestine [51,52,53]. However, certain studies have reported that DSS also affects the distal small intestine i.e. ileum [54,55]. DSS causes erosions with complete loss of surface epithelium because of its direct toxic effect on epithelial cells. It causes deformity in the epithelial integrity, thereby increases the colonic mucosal permeability allowing permeation of large molecules such as DSS with molecular weight upto 50,000 Da [20,56,57]. Therefore, DSS notably causes acute colitis which is morphologically and macroscopically characterized by hyperemia, ulcerations, moderate to severe submucosal edema, lesions accompanied by histopathological changes which include infiltration of granulocytes, symptoms of which are ultimately manifested in the form of bloody diarrhea [20]. DSS significantly causes increase in the production of all proinflammatory cytokines in both mid and in distal colon, but DSS-induced ulcerative colitis appears to be more severe in the distal colon [58].
The elevation in TNF-α levels is the hallmark of DSS-induced colitis. Many researchers have reported that chronic pathological changes develop after induction of acute inflammation which involve changes in the morphology of crypts in conjunction with changes in Th1/Th2 cytokine profile and TCM (central memory T cells) [59,60,61,62]. TCM are differentiated mainly on the basis of expression of an adhesion molecule CD62L, which allows them to get in and stay in lymphoid tissues like colon patches [60]. DSS causes disturbance in the metabolism of phospholipids depicted by decreased levels of phosphocholine and glycerophosphocholine in the colon of mice [63]. Phosphocholine and glycerophosphocholine are the most important metabolites of choline and the major cellular constituents required for the assembly of biological membranes and disturbance in the metabolism suggests the possibility of distorted membrane integrity in the presence of DSS [64]. In addition to this, noticeable reduction in the levels of nucleotides, such as cytidine 5'-diphosphate and adenosine 5'-monophosphate, with analogous reduction in the levels of nucleobases and nucleosides, such as uracil and uridine is observed, in the colon of DSS-treated mice [63]. Likewise, loss of tight junction protein ZO-1 might also facilitate in increasing intestinal permeability [65].
Classically, 2~5% DSS (w/v) (molecular weight 50 kDa) is added to the drinking water for a period of 5~9 successive days to induce colitis in Sprague-Dawley or Wistar rats [19,20,21,57,66]. Animals are sacrificed on 9th day after the induction of colitis for the assessment of various parameters i.e. TNF-α (hallmark of DSS induced colitis), IL-10, IL-1β, TGFβ, mucin, TLR2/4 gene expression, MPO activity, number of filled goblet cells and number of empty goblet cells. Disease Activity Index (DAI) is carried out throughout 9 days [19,20,21,57]. The research tool most frequently used to assess severity of IBD is Disease Activity Index. Disease Activity Index is a cumulative index of ulcerative colitis to quantify body weight loss, rectal bleeding and stool consistency [67,68]. Another researcher reported that 1% DSS for 9 days and 2% DSS for 6 days minimum induces colitis in male wild type rats [69]. Some researchers have reported that chronic colitis is induced by 4% DSS for 6 days, followed by water for 6 days for three cycles in Sprague-Dawley rats [50].
DSS is a robust model of colitis in which colitis is inducible in all backgrounds of mice by administering DSS in the drinking water [67]. Typically, acute colitis is induced in male and female C57BL/6NTac, C57BL/6 and BALB/C mice by incorporating 3 to 5% DSS (w/v) in drinking water ad libitum for 5~8 days [58,63,70,71,72,73,74,75,76]. Afterwards, colons are dissected and examined for histological changes, tissue cytokine levels i.e. TNF-α levels, IL-1β, IL-6, IL-10, and IL-17 and MPO activity. Colitis can also be induced in ddY, C57BL/6J mice by adding DSS to their drinking water to a final concentration of 2~2.5% for 4~10 days [46,77].
Cooper et al reported the development of both acute and chronic colitis in Swiss-Webster mice by DSS. Acute colitis is induced by administering DSS orally for 6~10 consecutive days. On the other hand, chronic colitis is induced by administering 4~5 repeated cycles of DSS, each cycle involves administration of 5% DSS for 1 week followed by water for 10~14 days in BALB/C mice [51,78]. Giner et al reported that chronic colitis may be induced in female C57BL/6 mice after administering four successive cycles of DSS. 1% DSS is administered during the first and second cycle followed by 2% DSS during the third and fourth cycle. Each cycle comprises of incorporation of DSS in drinking water for 7 days followed by delivering water without DSS 7 days [79].
The severity of colitis i.e. mild, moderate, severe colitis may be varied by varying the DSS treatment period. Administering 3%, w/v DSS for 7 days and sacrificing on the 8th day induces mild colitis whereas moderate colitis is induced by administering 3% DSS for a period of 14 days i.e. from days 1 to 7 and 22 to 28 and then sacrificing animals on the 29th day. During the remission period i.e. from day 8~21, mice are administered normal drinking water. Likewise, severe colitis is induced by administering 3% DSS w/v for 21 days from days 1 to 7, 22 to 28 and 43 to 49 and then sacrificing the animals on the 50th day. During the remission period i.e. on days 8~21 and 29~42 mice are delivered normal drinking water [80].

Oxazolone

This haptenating agent is generally used to induce colitis in mice to study the pathological processes involved in the perpetuation of ulcerative colitis. It is speculated that it mediates Th2 driven immune response [81]. Therefore, based on the manifestations of mucosal inflammation, augumented epithelial microulcerations and histopathological changes in the distal colon, it has been described that oxazolone induces colitis in rodents that mimics human ulcerative colitis [81,82]. Different immunological responses have been observed in various strains of mice i.e. C57BL/6J, BALB/CJ and SJL/J and accordingly, different procedures have been adapted to induce colitis in different strains mice. C57BL/6J (C57/BL6 or C57/BL10) and SJL/J strains have a strong inclination towards Th1 immune response, whereas BALB/J strain has tendency to exhibit immune response of Th2 phenotype [83]. C57 strain mice pose resistance to colitis induction by oxazolone, therefore, induction of colitis in C57 strains requires a pre-sensitizing treatment. SJL/J mouse strain is less preferred for the induction of colitis as this strain is prone to a number of autoimmune diseases initiated by Th1 phenotype [83]. This strain also possesses a high mortality rate and moreover it is difficult to keep two or more SJL/J mice together because of their aggressive nature [40].
Rectal administration of the oxazolone (hapten) dissolved in ethanol induces severe colitis. In a typical procedure to induce colitis in rats, 300 µL of 5% oxazolone (in absolute alcohol) is initially applied transdermally on the exposed area for presensitization. This is supervened by intrarectal administration of 450 µl of 5% oxazolone in 50% ethanol solution into the colon on 5th and 7th day, 4~8 cm proximal to the anal verge of rats using a catheter (1 mm diameter). In order to ensure even distribution of the oxazolone solution throughout the entire colon and caecum, the animals are maintained in a vertical position for at least 45 s. Finally, rats are sacrificed after 7 to 21 days after rectal administration of hapten agent for histological grading and carrying out various estimations [84]. Likewise, another researcher reported that oxazolone solution with concentration of 7.5 mg/ml in 40% (v/v) in aqueous ethanol administered once at a dose of 1.1 ml/rat into the colon leads to induction of colitis in rats [85].
Similarly, colitis can also be induced in mice of various strains presenting histological picture that closely resembles human colitis. The oxazolone treated mice develops rapid-onset colitis [81]. Typically, colitis is induced in SLJ/J, BALB/C mice by presensitizing the shaved abdominal skin with 3% oxazolone in 100% ethanol ranging from 50~150 µl. Five to eight days after presensitization, mice are rechallenged by intrarectal administration of 70~150 µl of 0.75~1% oxazolone in 45-50% ethanol solution which leading to induction of colitis. Animals are sacrificed after 7~12 days after presensitization to assess severity of colitis [24,43,48,82]. Huang et al reported that colitis is induced in male BALB/C mice by presensitizing with transdermal application of 200 µl of a 3% (w/v) oxazolone solution in acetone/olive oil (4:1) on the shaven skin. Then, the presensitized mice are rechallenged on the 8th day by intrarectal administration of 100 µl of one percent oxazolone in 50% ethanol to induce colitis [86]. Administration of 100 ~150 µl of 1~2% oxazolone at a dose of 90 mg/kg, 7.5 mg/ml solution may also induce colitis in male BALB/C [81,87,88].

Acetic acid

Acetic acid induced colitis is commonly employed and easily inducible model [89,90,91]. Acetic acid-induced colitis is a model of IBD that bears close resemblance to human IBD in terms of pathogenesis, histopathological features and inflammatory mediator profile [37,92,93,94,95,96]. Intrarectal administration of dilute solution of acetic acid causes non-transmural inflammation characterized by increased neutrophil infiltration into the intestinal tissue, massive necrosis of mucosal and submucosal layers, vascular dilation, edema and submucosal ulceration that are noteworthy features of human colitis [49,94,96,97,98]. It has been anticipated that the protonated form of the acid liberates protons within the intracellular space that possibly causes massive intracellular acidification resulting in immense epithelial damage.
It is quite possible that oxidative stress might play critical role in initiation and progression of IBD [99,100,101,102]. Since production of reactive oxygen species which include superoxide anion, hydrogen peroxide, hypochlorous acid and hydroxyl radical and nitrogen species is increased in IBD patients, this ascertains that oxidative stress appears to be the crucial pathogenic factor in IBD [25,99,100,103]. Likewise, the characteristic feature of experimentally induced colitis using acetic acid in animals is an imbalance between oxidant and antioxidant substances [95,104,105]. It has been well documented that infiltration of neutrophils leads to production of superoxide anion and initiation of a cascade for the production of various reactive species. This may lead to generation of hydroxyl radicals and peroxides that significantly contribute to the progression of tissue necrosis and mucosal dysfunction [96,98,99]. Furthermore, neutrophils also release proteases and lipid mediators that additionally contribute to intestinal injury.
Typically, induction of colitis using acetic acid involves anaesthetizing the rats with ether anteceded by a 24 h fast. Subsequently, using a medical-grade polyurethane tube for enteral feeding (external diameter 2 mm), 1~2 ml of (3~4%) acetic acid is instilled 6~8 cm proximal to the anus verge. Eventually, after 15~30 s of exposure the fluid is withdrawn and animals are sacrificed and their blood and colons are collected 24~48 hrs after induction of colitis for carrying out various histopathological and biochemical investigations [25,26,89,97,106,107,108]. Other scientists have revealed that intracolonic administration of 4 ml of 4 % acetic acid at a dose 5 ml/kg also leads to induction of colitis in rats [99,109,110,111,112]. However, another group of researchers reported that confinement of 1~2 ml of 3~6% acetic acid solution (v/v) by maintaining horizontal position of rats for at least 2 min also leads to the induction of colitis. They emphasized that horizontal position of rats should be maintained for at least 2 min to prevent leakage of acetic acid immediately after administering acetic acid [27,28,113,114]. Different researchers have used different approaches to modify the procedure for achieving colitis like condition after administering acetic acid to the animals. These include confinement of acetic acid into colon [27,28,113,114]; injection of air (2 ml) after instillation of the enema so that acetic acid spreads completely in the colon [115] and rinsing the colon lumen with saline (5 ml) 20s after instilling the enema [116].
Mice have also been extensively used to create colitis model using acetic acid. In order to induce colitis in mice, catheter is used to instill 1 ml of acetic acid (4~5%, v/v) in 0.9% saline into the lumen of the colon approximately 4 cm proximal to the anus. Thereby, the animals are maintained in a supine trendelenburg position for 30 s in order to prevent the leakage of the intracolonic instill [39,117,118].

Peptidoglycan

Peptidoglycan-polysaccharide is a component of cell wall of bacterium and perhaps possesses immunogenic property when given to mice and may lead to activation of T-cell-mediated immune response. Peptidoglycan-polysaccharide (PGPS) model is an experimental rat model of chronic, spontaneously relapsing, granulomatous enterocolitis and closely mimics human Crohn's disease [34,119]. In this model, intestinal inflammation is induced by carrying out laparotomy followed by subsequent intramural injection of purified, sterile, poorly biodegradable bacterial cell wall component, peptidoglycan polysaccharide from group A streptococci (PG-APS) which is quantified as equivalents of rhamnose, a sugar component of PG-APS. Genetically susceptible Lewis rats are given intramural (subserosal) injections of PG-PS (12.5 µg rhamnose equivalents/g body wt) into a segment (5-cm) of distal colon. Symptoms of acute intestinal inflammation depicted by increased vascular permeability develop after 1~2 days, gradually decrease over the next 10 days, and then abruptly reactivate on 14th day and perpetuate into a chronic granulomatous inflammatory syndrome resembling Crohn's disease [33,34,119,120]. Animals are killed on day 2 and day 19 for carrying out histopathological evaluations of acute phase and chronic phase respectively [120].
It has been documented that peptidoglycan-polysaccharide when delivered to mice causes activation of kallikrein-kinin system; kallikrein thus released is chemotactic for neutrophils [121]. Moreover, activation of this system results in augumented release of bradykinin [122] which subsequently causes stimulation of inflammatory cytokines e.g. IL-1 [123] and consequently may mediate progression of inflammation in the intestine. Enhanced production of NO after PG-PS administration has also been reported [124].

Carrageenan

Carrageenan is a high molecular weight sulphated polysaccharide obtained from red algae. Based on the degree of sulphation and solubility it is categorized into three distinct sub types i.e. kappa, iota, lambda. Carrageenan itself isn't that much harmful but the products yielded after mild hydrolysis cause ulcerations in the colon of various animal species including rats. A lambda-carrageenan induced rat model of IBD allows sequential evaluation of histopathological and morphological changes in intestine over time and mimics human ulcerative colitis [31,32]. The lambda-carrageenan model of intestinal inflammation is chosen for its ease of preparation and lack of discomfort to the animals involved [31]. Intestinal lesions gradually develop within 2~4 weeks but are detectable only after 8 weeks in lambda-carrageenan fed rats and are found to be morphologically similar to those observed in human ulcerative colitis [125]. Colitis is induced by the addition of 2% lambda-carrageenan for 6 weeks into the drinking water of male Sprague-Dawley rats without prior sensitization to the compound [32]. Another experimental design involves prior parenteral sensitization by a 1.5 percent solution of lambda degraded carrageenan followed by oral administration of the same solution for 30 days [31].
It is quite possible that ulcerogenic potential of carrageenan might due to the presence of electronegative polyanions [126]. Although free radicals are involved [127], but overproduction of nitric oxide may be a possible mechanism for causing immunosuppression during chronic IBD [128]. In vitro studies have revealed that administration of degraded carrageenan causes induction of colitis via NF-κB mediated upregulation of ICAM-1, TNF-α secretion and expression [129].

NSAIDS

Administration of indomethacin solubilized in 100% alcohol and then diluted with 5% w/v sodium bicarbonate solution at a dose of 7.5 mg/kg s.c on two consecutive days is shown to induce colitis in rats [30]. Acute colitis can also be induced by administering a nonselective NSAID, piroxicam at a dose of 200 ppm for 2 weeks [29].

CONCLUSION

Among the various chemically induced colitis models, TNBS-induced-colitis, oxazolone induced-colitis and DSS-induced colitis models are the most widely used to induce IBD. These models symptomatically, morphologically and histopathologically resemble human IBD. Therefore, in the coming years utilizing an appropriate colitis model one can develop novel therapeutic strategy for the treatment of IBD. Moreover, the plausible mechanism of action of a particular drug can be illustrated using a suitable colitis model.

ACKNOWLEDGEMENT

The authors are thankful to Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India for supporting us.

ABBREVIATIONS

IBD

inflammatory bowel disease

TNBS

2, 4, 6-trinitrobenzene sulfonic acid

DSS

dextran sodium sulphate

NSAIDs

nonsteroidal anti-inflammatory drugs

PGPS

peptidoglycan-polysaccharide

DAI

disease activity index

PG-APS

peptidoglycan polysaccharide from group A streptococci

NF-κB

nuclear factor-κB

ICAM-1

intercellular adhesion molecule 1

TNF-α

tumour necrosis factor-α

ppm

parts per million

References

1. Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW, Kaplan GG. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012; 142:46–54. PMID: 22001864.
crossref
2. Büsch K, Ludvigsson JF, Ekström-Smedby K, Ekbom A, Askling J, Neovius M. Nationwide prevalence of inflammatory bowel disease in Sweden: a population-based register study. Aliment Pharmacol Ther. 2014; 39:57–68. PMID: 24127738.
crossref
3. Koch TR, Yuan LX, Stryker SJ, Ratliff P, Telford GL, Opara EC. Total antioxidant capacity of colon in patients with chronic ulcerative colitis. Dig Dis Sci. 2000; 45:1814–1819. PMID: 11052325.
4. Cetinkaya A, Bulbuloglu E, Kantarceken B, Ciralik H, Kurutas EB, Buyukbese MA, Gumusalan Y. Effects of L-carnitine on oxidant/antioxidant status in acetic acid-induced colitis. Dig Dis Sci. 2006; 51:488–494. PMID: 16614957.
crossref
5. Danese S, Angelucci E. New and emerging biologics in the treatment of inflammatory bowel disease: quo vadis? Gastroenterol Clin Biol. 2009; 33(Suppl 3):S217–S227. PMID: 20117345.
crossref
6. Carter MJ, Lobo AJ, Travis SP. IBD Section, British Society of Gastroenterology. Guidelines for the management of inflammatory bowel disease in adults. Gut. 2004; 53(Suppl 5):V1–V16. PMID: 15306569.
crossref
7. Fuss IJ, Neurath M, Boirivant M, Klein JS, de la Motte C, Strong SA, Fiocchi C, Strober W. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996; 157:1261–1270. PMID: 8757634.
8. Parronchi P, Romagnani P, Annunziato F, Sampognaro S, Becchio A, Giannarini L, Maggi E, Pupilli C, Tonelli F, Romagnani S. Type 1 T-helper cell predominance and interleukin-12 expression in the gut of patients with Crohn's disease. Am J Pathol. 1997; 150:823–832. PMID: 9060820.
9. Katz LH, Kopylov U, Fudim E, Yavzori M, Picard O, Ungar B, Eliakim R, Ben-Horin S, Chowers Y. Expression of IL-2, IL-17 and TNF-alpha in patients with Crohn's disease treated with anti-TNF antibodies. Clin Res Hepatol Gastroenterol. 2014; [Epub ahead of print].
crossref
10. Papaconstantinou I, Zeglinas C, Gazouli M, Nastos K, Yiallourou A, Papalois A, Tzathas C. The impact of perioperative anti-TNF treatment on anastomosis-related complications in Crohn's disease patients. A critical review. J Gastrointest Surg. 2014; 18:1216–1224. PMID: 24599780.
crossref
11. Camoglio L, Te Velde AA, Tigges AJ, Das PK, Van Deventer SJ. Altered expression of interferon-gamma and interleukin-4 in inflammatory bowel disease. Inflamm Bowel Dis. 1998; 4:285–290. PMID: 9836081.
12. Melgar S, Yeung MM, Bas A, Forsberg G, Suhr O, Oberg A, Hammarstrom S, Danielsson A, Hammarstrom ML. Overexpression of interleukin 10 in mucosal T cells of patients with active ulcerative colitis. Clin Exp Immunol. 2003; 134:127–137. PMID: 12974765.
crossref
13. Fuss IJ, Heller F, Boirivant M, Leon F, Yoshida M, Fichtner-Feigl S, Yang Z, Exley M, Kitani A, Blumberg RS, Mannon P, Strober W. Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. J Clin Invest. 2004; 113:1490–1497. PMID: 15146247.
crossref
14. Rana SV, Sharma S, Kaur J, Prasad KK, Sinha SK, Kochhar R, Malik A, Morya RK. Relationship of cytokines, oxidative stress and GI motility with bacterial overgrowth in ulcerative colitis patients. J Crohns Colitis. 2014; [Epub ahead of print].
crossref
15. Ellrichmann M, Wietzke-Braun P, Dhar S, Nikolaus S, Arlt A, Bethge J, Kuehbacher T, Wintermeyer L, Balschun K, Klapper W, Schreiber S, Fritscher-Ravens A. Endoscopic ultrasound of the colon for the differentiation of Crohn's disease and ulcerative colitis in comparison with healthy controls. Aliment Pharmacol Ther. 2014; 39:823–833. PMID: 24612000.
crossref
16. Morris GP, Beck PL, Herridge MS, Depew WT, Szewczuk MR, Wallace JL. Hapten-induced model of chronic inflammation and ulceration in the rat colon. Gastroenterology. 1989; 96:795–803. PMID: 2914642.
crossref
17. da Silva MS, Sánchez-Fidalgo S, Talero E, Cárdeno A, da Silva MA, Villegas W, Souza Brito AR, de La Lastra CA. Antiinflammatory intestinal activity of Abarema cochliacarpos (Gomes) Barneby & Grimes in TNBS colitis model. J Ethnopharmacol. 2010; 128:467–475. PMID: 20083187.
18. de Almeida AB, Sánchez-Hidalgo M, Martín AR, Luiz-Ferreira A, Trigo JR, Vilegas W, dos Santos LC, Souza-Brito AR, de la Lastra CA. Anti-inflammatory intestinal activity of Arctium lappa L. (Asteraceae) in TNBS colitis model. J Ethnopharmacol. 2013; 146:300–310. PMID: 23313393.
crossref
19. Moncada DM, Kammanadiminti SJ, Chadee K. Mucin and Toll-like receptors in host defense against intestinal parasites. Trends Parasitol. 2003; 19:305–311. PMID: 12855381.
crossref
20. Dharmani P, Leung P, Chadee K. Tumor necrosis factor-α and Muc2 mucin play major roles in disease onset and progression in dextran sodium sulphate-induced colitis. PLoS One. 2011; 6:e25058. PMID: 21949848.
crossref
21. Cheah KY, Bastian SE, Acott TM, Abimosleh SM, Lymn KA, Howarth GS. Grape seed extract reduces the severity of selected disease markers in the proximal colon of dextran sulphate sodium-induced colitis in rats. Dig Dis Sci. 2013; 58:970–977. PMID: 23143736.
crossref
22. Zhang HQ, Ding TT, Zhao JS, Yang X, Zhang HX, Zhang JJ, Cui YL. Therapeutic effects of Clostridium butyricum on experimental colitis induced by oxazolone in rats. World J Gastroenterol. 2009; 15:1821–1828. PMID: 19370778.
crossref
23. Yang J, Zhao J, Nakaguchi T, Gregersen H. Biomechanical changes in oxazolone-induced colitis in BALB/C mice. J Biomech. 2009; 42:811–817. PMID: 19264309.
crossref
24. Lee J, Yamamoto T, Kuramoto H, Kadowaki M. TRPV1 expressing extrinsic primary sensory neurons play a protective role in mouse oxazolone-induced colitis. Auton Neurosci. 2012; 166:72–76. PMID: 21855422.
crossref
25. 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. PMID: 8949646.
crossref
26. Hagar HH, El-Medany A, El-Eter E, Arafa M. Ameliorative effect of pyrrolidinedithiocarbamate on acetic acid-induced colitis in rats. Eur J Pharmacol. 2007; 554:69–77. PMID: 17112501.
crossref
27. Mousavizadeh K, Rahimian R, Fakhfouri G, Aslani FS, Ghafourifar P. Anti-inflammatory effects of 5-HT receptor antagonist, tropisetron on experimental colitis in rats. Eur J Clin Invest. 2009; 39:375–383. PMID: 19302562.
28. Al-Rejaie SS, Abuohashish HM, Al-Enazi MM, Al-Assaf AH, Parmar MY, Ahmed MM. Protective effect of naringenin on acetic acid-induced ulcerative colitis in rats. World J Gastroenterol. 2013; 19:5633–5644. PMID: 24039355.
crossref
29. Narushima S, Spitz DR, Oberley LW, Toyokuni S, Miyata T, Gunnett CA, Buettner GR, Zhang J, Ismail H, Lynch RG, Berg DJ. Evidence for oxidative stress in NSAID-induced colitis in IL10-/- mice. Free Radic Biol Med. 2003; 34:1153–1166. PMID: 12706496.
crossref
30. Pawar AT, Anap RM, Ghodasara JV, Kuchekar BS. Protective Effect of Hydroalcoholic Root Extract of Rubia cordifolia in Indomethacin-Induced Enterocolitis in Rats. Indian J Pharm Sci. 2011; 73:250–253. PMID: 22303076.
crossref
31. Moyana TN, Lalonde JM. Carrageenan-induced intestinal injury in the rat--a model for inflammatory bowel disease. Ann Clin Lab Sci. 1990; 20:420–426. PMID: 2073092.
32. Pricolo VE, Madhere SM, Finkelstein SD, Reichner JS. Effects of lambda-carrageenan induced experimental enterocolitis on splenocyte function and nitric oxide production. J Surg Res. 1996; 66:6–11. PMID: 8954824.
crossref
33. Fitzpatrick LR, Wang J, Le T. Gliotoxin, an inhibitor of nuclear factor-kappa B, attenuates peptidoglycan-polysaccharide-induced colitis in rats. Inflamm Bowel Dis. 2002; 8:159–167. PMID: 11979135.
crossref
34. Reingold L, Rahal K, Schmiedlin-Ren P, Rittershaus AC, Bender D, Owens SR, Adler J, Zimmermann EM. Development of a peptidoglycan-polysaccharide murine model of Crohn's disease: effect of genetic background. Inflamm Bowel Dis. 2013; 19:1238–1244. PMID: 23619717.
35. Cheon GJ, Cui Y, Yeon DS, Kwon SC, Park BG. Mechanisms of motility change on trinitrobenzenesulfonic Acid-induced colonic inflammation in mice. Korean J Physiol Pharmacol. 2012; 16:437–446. PMID: 23269907.
crossref
36. Isik F, Tunali Akbay T, Yarat A, Genc Z, Pisiriciler R, Caliskan-Ak E, Cetinel S, Altıntas A, Sener G. Protective effects of black cumin (Nigella sativa) oil on TNBS-induced experimental colitis in rats. Dig Dis Sci. 2011; 56:721–730. PMID: 20658190.
crossref
37. Elson CO, Sartor RB, Tennyson GS, Riddell RH. Experimental models of inflammatory bowel disease. Gastroenterology. 1995; 109:1344–1367. PMID: 7557106.
crossref
38. Yu Q, Zhu S, Zhou R, Yi F, Bing Y, Huang S, Wang Z, Wang C, Xia B. Effects of sinomenine on the expression of microRNA-155 in 2,4,6-trinitrobenzenesulfonic acid-induced colitis in mice. PLoS One. 2013; 8:e73757. PMID: 24066068.
crossref
39. Neurath MF, Fuss I, Kelsall BL, Stüber E, Strober W. Antibodies to interleukin 12 abrogate established experimental colitis in mice. J Exp Med. 1995; 182:1281–1290. PMID: 7595199.
crossref
40. Boirivant M, Fuss IJ, Chu A, Strober W. Oxazolone colitis: A murine model of T helper cell type 2 colitis treatable with antibodies to interleukin 4. J Exp Med. 1998; 188:1929–1939. PMID: 9815270.
crossref
41. Dohi T, Fujihashi K, Kiyono H, Elson CO, McGhee JR. Mice deficient in Th1- and Th2-type cytokines develop distinct forms of hapten-induced colitis. Gastroenterology. 2000; 119:724–733. PMID: 10982767.
crossref
42. Qin HY, Wu JC, Tong XD, Sung JJ, Xu HX, Bian ZX. Systematic review of animal models of post-infectious/post-inflammatory irritable bowel syndrome. J Gastroenterol. 2011; 46:164–174. PMID: 20848144.
crossref
43. Ikeda M, Takeshima F, Isomoto H, Shikuwa S, Mizuta Y, Ozono Y, Kohno S. Simvastatin attenuates trinitrobenzene sulfonic acid-induced colitis, but not oxazalone-induced colitis. Dig Dis Sci. 2008; 53:1869–1875. PMID: 18049901.
crossref
44. Takagi T, Naito Y, Mizushima K, Akagiri S, Suzuki T, Hirata I, Omatsu T, Handa O, Kokura S, Ichikawa H, Yoshikawa T. Inhalation of carbon monoxide ameliorates TNBS-induced colitis in mice through the inhibition of TNF-α expression. Dig Dis Sci. 2010; 55:2797–2804. PMID: 20094779.
crossref
45. Martínez-Moya P, Romero-Calvo I, Requena P, Hernández-Chirlaque C, Aranda CJ, González R, Zarzuelo A, Suárez MD, Martínez-Augustin O, Marín JJ, de Medina FS. Dose-dependent antiinflammatory effect of ursodeoxycholic acid in experimental colitis. Int Immunopharmacol. 2013; 15:372–380. PMID: 23246254.
crossref
46. Park SY, Ku SK, Lee ES, Kim JA. 1,3-Diphenylpropenone ameliorates TNBS-induced rat colitis through suppression of NF-κB activation and IL-8 induction. Chem Biol Interact. 2012; 196:39–49. PMID: 22410118.
crossref
47. Liu DY, Guan YM, Zhao HM, Yan DM, Tong WT, Wan PT, Zhu WF, Liu HN, Liang XL. The protective and healing effects of Si Shen Wan in trinitrobenzene sulphonic acid-induced colitis. J Ethnopharmacol. 2012; 143:435–440. PMID: 22751005.
crossref
48. Yang XL, Guo TK, Wang YH, Gao MT, Qin H, Wu YJ. Therapeutic effect of ginsenoside Rd in rats with TNBS-induced recurrent ulcerative colitis. Arch Pharm Res. 2012; 35:1231–1239. PMID: 22864746.
crossref
49. Jurjus AR, Khoury NN, Reimund JM. Animal models of inflammatory bowel disease. J Pharmacol Toxicol Methods. 2004; 50:81–92. PMID: 15385082.
crossref
50. Gaudio E, Taddei G, Vetuschi A, Sferra R, Frieri G, Ricciardi G, Caprilli R. Dextran sulfate sodium (DSS) colitis in rats: clinical, structural, and ultrastructural aspects. Dig Dis Sci. 1999; 44:1458–1475. PMID: 10489934.
51. Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 1990; 98:694–702. PMID: 1688816.
crossref
52. Tran CD, Ball JM, Sundar S, Coyle P, Howarth GS. The role of zinc and metallothionein in the dextran sulfate sodium-induced colitis mouse model. Dig Dis Sci. 2007; 52:2113–2121. PMID: 17410436.
crossref
53. Perše M, Cerar A. Dextran sodium sulphate colitis mouse model: traps and tricks. J Biomed Biotechnol. 2012; 2012:718617. PMID: 22665990.
crossref
54. Geier MS, Smith CL, Butler RN, Howarth GS. Small-intestinal manifestations of dextran sulfate sodium consumption in rats and assessment of the effects of Lactobacillus fermentum BR11. Dig Dis Sci. 2009; 54:1222–1228. PMID: 19005763.
crossref
55. Ohtsuka Y, Sanderson IR. Dextran sulfate sodium-induced inflammation is enhanced by intestinal epithelial cell chemokine expression in mice. Pediatr Res. 2003; 53:143–147. PMID: 12508094.
crossref
56. Ni J, Chen SF, Hollander D. Effects of dextran sulphate sodium on intestinal epithelial cells and intestinal lymphocytes. Gut. 1996; 39:234–241. PMID: 8991862.
crossref
57. Dai C, Zheng CQ, Meng FJ, Zhou Z, Sang LX, Jiang M. VSL#3 probiotics exerts the anti-inflammatory activity via PI3k/Akt and NF-κB pathway in rat model of DSS-induced colitis. Mol Cell Biochem. 2013; 374:1–11. PMID: 23271629.
crossref
58. Andújar I, Ríos JL, Giner RM, Miguel Cerdá J, Recio Mdel C. Beneficial effect of shikonin on experimental colitis induced by dextran sulfate sodium in BALB/c mice. Evid Based Complement Alternat Med. 2012; 2012:271606. PMID: 23346196.
crossref
59. Dieleman LA, Palmen MJ, Akol H, Bloemena E, Peña AS, Meuwissen SG, Van Rees EP. Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Exp Immunol. 1998; 114:385–391. PMID: 9844047.
crossref
60. Melgar S, Karlsson A, Michaëlsson E. Acute colitis induced by dextran sulfate sodium progresses to chronicity in C57BL/6 but not in BALB/c mice: correlation between symptoms and inflammation. Am J Physiol Gastrointest Liver Physiol. 2005; 288:G1328–G1338. PMID: 15637179.
crossref
61. Perše M, Cerar A. Dextran sodium sulphate colitis mouse model: traps and tricks. J Biomed Biotechnol. 2012; 2012:718617. PMID: 22665990.
crossref
62. Morgan ME, Zheng B, Koelink PJ, van de Kant HJ, Haazen LC, van Roest M, Garssen J, Folkerts G, Kraneveld AD. New perspective on dextran sodium sulfate colitis: antigen-specific T cell development during intestinal inflammation. PLoS One. 2013; 8:e69936. PMID: 23936123.
crossref
63. Dong F, Zhang L, Hao F, Tang H, Wang Y. Systemic responses of mice to dextran sulfate sodium-induced acute ulcerative colitis using 1H NMR spectroscopy. J Proteome Res. 2013; 12:2958–2966. PMID: 23651354.
crossref
64. Michel V, Yuan Z, Ramsubir S, Bakovic M. Choline transport for phospholipid synthesis. Exp Biol Med (Maywood). 2006; 231:490–504. PMID: 16636297.
crossref
65. Poritz LS, Garver KI, Green C, Fitzpatrick L, Ruggiero F, Koltun WA. Loss of the tight junction protein ZO-1 in dextran sulfate sodium induced colitis. J Surg Res. 2007; 140:12–19. PMID: 17418867.
crossref
66. Singh K, Jaggi AS, Singh N. Exploring the ameliorative potential of Punica granatum in dextran sulfate sodium induced ulcerative colitis in mice. Phytother Res. 2009; 23:1565–1574. PMID: 19367659.
67. Dieleman LA, Peña AS, Meuwissen SG, van Rees EP. Role of animal models for the pathogenesis and treatment of inflammatory bowel disease. Scand J Gastroenterol Suppl. 1997; 223:99–104. PMID: 9200314.
68. Rijcken EM, Laukoetter MG, Anthoni C, Meier S, Mennigen R, Spiegel HU, Bruewer M, Senninger N, Vestweber D, Krieglstein CF. Immunoblockade of PSGL-1 attenuates established experimental murine colitis by reduction of leukocyte rolling. Am J Physiol Gastrointest Liver Physiol. 2004; 287:G115–G124. PMID: 15001428.
crossref
69. Iwaya H, Fujii N, Hagio M, Hara H, Ishizuka S. Contribution of dipeptidyl peptidase IV to the severity of dextran sulfate sodium-induced colitis in the early phase. Biosci Biotechnol Biochem. 2013; 77:1461–1466. PMID: 23832365.
crossref
70. Kumar G K, Dhamotharan R, Kulkarni NM, Honnegowda S, Murugesan S. Embelin ameliorates dextran sodium sulfate-induced colitis in mice. Int Immunopharmacol. 2011; 11:724–731. PMID: 21296695.
71. Coburn LA, Gong X, Singh K, Asim M, Scull BP, Allaman MM, Williams CS, Rosen MJ, Washington MK, Barry DP, Piazuelo MB, Casero RA Jr, Chaturvedi R, Zhao Z, Wilson KT. L-arginine supplementation improves responses to injury and inflammation in dextran sulfate sodium colitis. PLoS One. 2012; 7:e33546. PMID: 22428068.
crossref
72. Laroui H, Ingersoll SA, Liu HC, Baker MT, Ayyadurai S, Charania MA, Laroui F, Yan Y, Sitaraman SV, Merlin D. Dextran sodium sulfate (DSS) induces colitis in mice by forming nano-lipocomplexes with medium-chain-length fatty acids in the colon. PLoS One. 2012; 7:e32084. PMID: 22427817.
crossref
73. Jadhav SR, Shandilya UK, Kansal VK. Exploring the ameliorative potential of probiotic Dahi containing Lactobacillus acidophilus and Bifidobacterium bifidum on dextran sodium sulphate induced colitis in mice. J Dairy Res. 2013; 80:21–27. PMID: 23317563.
74. Kleiveland CR, Hult LT, Spetalen S, Kaldhusdal M, Christofferesen TE, Bengtsson O, Romarheim OH, Jacobsen M, Lea T. The noncommensal bacterium Methylococcus capsulatus (Bath) ameliorates dextran sulfate (Sodium Salt)-Induced Ulcerative Colitis by influencing mechanisms essential for maintenance of the colonic barrier function. Appl Environ Microbiol. 2013; 79:48–56. PMID: 23064342.
crossref
75. Köhnke T, Gomolka B, Bilal S, Zhou X, Sun Y, Rothe M, Baumgart DC, Weylandt KH. Acetylsalicylic Acid reduces the severity of dextran sodium sulfate-induced colitis and increases the formation of anti-inflammatory lipid mediators. Biomed Res Int. 2013; 2013:748160. PMID: 24083240.
crossref
76. Liu L, Liu YL, Liu GX, Chen X, Yang K, Yang YX, Xie Q, Gan HK, Huang XL, Gan HT. Curcumin ameliorates dextran sulfate sodium-induced experimental colitis by blocking STAT3 signaling pathway. Int Immunopharmacol. 2013; 17:314–320. PMID: 23856612.
crossref
77. Kato E, Yamane S, Nomura R, Matsumoto K, Tashima K, Horie S, Saito T, Fujino H, Murayama T. Dysfunction of neurogenic VIP-mediated relaxation in mouse distal colon with dextran sulfate sodium-induced colitis. Pharmacol Res. 2012; 65:204–212. PMID: 21939768.
crossref
78. Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 1993; 69:238–249. PMID: 8350599.
79. Giner E, Recio MC, Ríos JL, Giner RM. Oleuropein protects against dextran sodium sulfate-induced chronic colitis in mice. J Nat Prod. 2013; 76:1113–1120. PMID: 23758110.
crossref
80. Trivedi PP, Jena GB. Dextran sulfate sodium-induced ulcerative colitis leads to increased hematopoiesis and induces both local as well as systemic genotoxicity in mice. Mutat Res. 2012; 744:172–183. PMID: 22414559.
crossref
81. Kojima R, Kuroda S, Ohkishi T, Nakamaru K, Hatakeyama S. Oxazolone-induced colitis in BALB/C mice: a new method to evaluate the efficacy of therapeutic agents for ulcerative colitis. J Pharmacol Sci. 2004; 96:307–313. PMID: 15539761.
crossref
82. Engel MA, Khalil M, Siklosi N, Mueller-Tribbensee SM, Neuhuber WL, Neurath MF, Becker C, Reeh PW. Opposite effects of substance P and calcitonin gene-related peptide in oxazolone colitis. Dig Liver Dis. 2012; 44:24–29. PMID: 22018693.
crossref
83. Charles PC, Weber KS, Cipriani B, Brosnan CF. Cytokine, chemokine and chemokine receptor mRNA expression in different strains of normal mice: implications for establishment of a Th1/Th2 bias. J Neuroimmunol. 1999; 100:64–73. PMID: 10695717.
crossref
84. Zhang HQ, Ding TT, Zhao JS, Yang X, Zhang HX, Zhang JJ, Cui YL. Therapeutic effects of Clostridium butyricum on experimental colitis induced by oxazolone in rats. World J Gastroenterol. 2009; 15:1821–1828. PMID: 19370778.
crossref
85. Abdin AA. Targeting sphingosine kinase 1 (SphK1) and apoptosis by colon-specific delivery formula of resveratrol in treatment of experimental ulcerative colitis in rats. Eur J Pharmacol. 2013; 718:145–153. PMID: 24055189.
crossref
86. Huang Z, Jiang Y, Yang Y, Shao J, Sun X, Chen J, Dong L, Zhang J. 3,3'-Diindolylmethane alleviates oxazolone-induced colitis through Th2/Th17 suppression and Treg induction. Mol Immunol. 2013; 53:335–344. PMID: 23085552.
crossref
87. Daniel C, Sartory NA, Zahn N, Schmidt R, Geisslinger G, Radeke HH, Stein JM. FTY720 ameliorates oxazolone colitis in mice by directly affecting T helper type 2 functions. Mol Immunol. 2007; 44:3305–3316. PMID: 17475330.
crossref
88. Ma Y, Semba S, Maemoto A, Takeuchi M, Kameshita I, Ishida A, Kato S, Katoh T, Liu Y, Taniguchi T. Oxazolone-induced over-expression of focal adhesion kinase in colonic epithelial cells of colitis mouse model. FEBS Lett. 2010; 584:3949–3954. PMID: 20682312.
crossref
89. MacPherson BR, Pfeiffer CJ. Experimental production of diffuse colitis in rats. Digestion. 1978; 17:135–150. PMID: 627326.
crossref
90. Noa M, Más R, Carbajal D, Valdés S. Effect of D-002 on acetic acid-induced colitis in rats at single and repeated doses. Pharmacol Res. 2000; 41:391–395. PMID: 10704261.
crossref
91. Sasaki S, Hirata I, Maemura K, Hamamoto N, Murano M, Toshina K, Katsu K. Prostaglandin E2 inhibits lesion formation in dextran sodium sulphate-induced colitis in rats and reduces the levels of mucosal inflammatory cytokines. Scand J Immunol. 2000; 51:23–28. PMID: 10632972.
crossref
92. González R, Rodríguez S, Romay C, Ancheta O, González A, Armesto J, Remirez D, Merino N. Anti-inflammatory activity of phycocyanin extract in acetic acid-induced colitis in rats. Pharmacol Res. 1999; 39:55–59. PMID: 10366332.
93. Gorgulu S, Yagci G, Kaymakcioglu N, Ozkara M, Kurt B, Ozcan A, Kaya O, Sadir S, Tufan T. Hyperbaric oxygen enhances the efficiency of 5-aminosalicylic acid in acetic acid-induced colitis in rats. Dig Dis Sci. 2006; 51:480–487. PMID: 16614956.
crossref
94. Nakhai LA, Mohammadirad A, Yasa N, Minaie B, Nikfar S, Ghazanfari G, Zamani MJ, Dehghan G, Jamshidi H, Boushehri VS, Khorasani R, Abdollahi M. Benefits of Zataria multiflora Boiss in experimental model of mouse inflammatory bowel disease. Evid Based Complement Alternat Med. 2007; 4:43–50. PMID: 17342240.
95. Bitiren M, Karakilcik AZ, Zerin M, Ozardali I, Selek S, Nazligul Y, Ozgonul A, Musa D, Uzunkoy A. Protective effects of selenium and vitamin E combination on experimental colitis in blood plasma and colon of rats. Biol Trace Elem Res. 2010; 136:87–95. PMID: 19774348.
crossref
96. Hartmann RM, Morgan Martins MI, Tieppo J, Fillmann HS, Marroni NP. Effect of Boswellia serrata on antioxidant status in an experimental model of colitis rats induced by acetic acid. Dig Dis Sci. 2012; 57:2038–2044. PMID: 22451119.
crossref
97. Daneshmand A, Rahimian R, Mohammadi H, Ejtemaee-Mehr S, Tavangar SM, Babaei Kelishomi R, Dehpour AR. Protective effects of lithium on acetic acid-induced colitis in rats. Dig Dis Sci. 2009; 54:1901–1907. PMID: 19082724.
crossref
98. Closa D, Folch-Puy E. Oxygen free radicals and the systemic inflammatory response. IUBMB Life. 2004; 56:185–191. PMID: 15230345.
crossref
99. Grisham MB, Granger DN. Neutrophil-mediated mucosal injury. Role of reactive oxygen metabolites. Dig Dis Sci. 1988; 33(3 Suppl):6S–15S. PMID: 2831016.
100. Kruidenier L, Verspaget HW. Review article: oxidative stress as a pathogenic factor in inflammatory bowel disease--radicals or ridiculous? Aliment Pharmacol Ther. 2002; 16:1997–2015. PMID: 12452933.
101. Wakefield AJ, Sawyerr AM, Dhillon AP, Pittilo RM, Rowles PM, Lewis AA, Pounder RE. Pathogenesis of Crohn's disease: multifocal gastrointestinal infarction. Lancet. 1989; 2:1057–1062. PMID: 2572794.
crossref
102. Dhillon AP, Anthony A, Sim R, Wakefield AJ, Sankey EA, Hudson M, Allison MC, Pounder RE. Mucosal capillary thrombi in rectal biopsies. Histopathology. 1992; 21:127–133. PMID: 1505929.
crossref
103. Joshi SV, Vyas BA, Shah PD, Shah DR, Shah SA, Gandhi TR. Protective effect of aqueous extract of Oroxylum indicum Linn. (root bark) against DNBS-induced colitis in rats. Indian J Pharmacol. 2011; 43:656–661. PMID: 22144769.
104. Keshavarzian A, Sedghi S, Kanofsky J, List T, Robinson C, Ibrahim C, Winship D. Excessive production of reactive oxygen metabolites by inflamed colon: analysis by chemiluminescence probe. Gastroenterology. 1992; 103:177–185. PMID: 1612325.
crossref
105. Dröge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002; 82:47–95. PMID: 11773609.
crossref
106. Yalniz M, Demirel U, Orhan C, Bahcecioglu IH, Ozercan IH, Aygun C, Tuzcu M, Sahin K. Nadroparin sodium activates Nrf2/HO-1 pathway in acetic acid-induced colitis in rats. Inflammation. 2012; 35:1213–1221. PMID: 22350949.
crossref
107. Kannan N, Guruvayoorappan C. Protective effect of Bauhinia tomentosa on acetic acid induced ulcerative colitis by regulating antioxidant and inflammatory mediators. Int Immunopharmacol. 2013; 16:57–66. PMID: 23538025.
crossref
108. Iseri SO, Ersoy Y, Ercan F, Yuksel M, Atukeren P, Gumustas K, Alican I. The effect of sildenafil, a phosphodiesterase-5 inhibitor, on acetic acid-induced colonic inflammation in the rat. J Gastroenterol Hepatol. 2009; 24:1142–1148. PMID: 19638092.
crossref
109. Yamada Y, Marshall S, Specian RD, Grisham MB. A comparative analysis of two models of colitis in rats. Gastroenterology. 1992; 102:1524–1534. PMID: 1314749.
crossref
110. Tannahill CL, Stevenot SA, Campbell-Thompson M, Nick HS, Valentine JF. Induction and immunolocalization of manganese superoxide dismutase in acute acetic acid-induced colitis in the rat. Gastroenterology. 1995; 109:800–811. PMID: 7657108.
crossref
111. Mascolo N, Izzo AA, Autore G, Maiello FM, Di Carlo G, Capasso F. Acetic acid-induced colitis in normal and essential fatty acid deficient rats. J Pharmacol Exp Ther. 1995; 272:469–475. PMID: 7815363.
112. Hassan GS, Soliman GA. Design, synthesis and anti-ulcerogenic effect of some of furo-salicylic acid derivatives on acetic acid-induced ulcerative colitis. Eur J Med Chem. 2010; 45:4104–4112. PMID: 20573425.
crossref
113. Kolgazi M, Uslu U, Yuksel M, Velioglu-Ogunc A, Ercan F, Alican I. The role of cholinergic anti-inflammatory pathway in acetic acid-induced colonic inflammation in the rat. Chem Biol Interact. 2013; 205:72–80. PMID: 23810507.
crossref
114. Thippeswamy BS, Mahendran S, Biradar MI, Raj P, Srivastava K, Badami S, Veerapur VP. Protective effect of embelin against acetic acid induced ulcerative colitis in rats. Eur J Pharmacol. 2011; 654:100–105. PMID: 21185828.
crossref
115. Tahan G, Aytac E, Aytekin H, Gunduz F, Dogusoy G, Aydin S, Tahan V, Uzun H. Vitamin E has a dual effect of antiinflammatory and antioxidant activities in acetic acid-induced ulcerative colitis in rats. Can J Surg. 2011; 54:333–338. PMID: 21933527.
crossref
116. Xing J, Sun J, You H, Lv J, Sun J, Dong Y. Anti-inflammatory effect of 3,4-oxo-isopropylidene-shikimic acid on acetic acid-induced colitis in rats. Inflammation. 2012; 35:1872–1879. PMID: 22829139.
crossref
117. Rachmilewitz D, Karmeli F, Takabayashi K, Hayashi T, Leider-Trejo L, Lee J, Leoni LM, Raz E. Immunostimulatory DNA ameliorates experimental and spontaneous murine colitis. Gastroenterology. 2002; 122:1428–1441. PMID: 11984528.
crossref
118. Niu X, Fan T, Li W, Huang H, Zhang Y, Xing W. Protective effect of sanguinarine against acetic acid-induced ulcerative colitis in mice. Toxicol Appl Pharmacol. 2013; 267:256–265. PMID: 23352506.
crossref
119. Fitzpatrick LR, Wang J, Le T. Caffeic acid phenethyl ester, an inhibitor of nuclear factor-kappaB, attenuates bacterial peptidoglycan polysaccharide-induced colitis in rats. J Pharmacol Exp Ther. 2001; 299:915–920. PMID: 11714876.
120. Stadnicki A, Sartor RB, Janardham R, Majluf-Cruz A, Kettner CA, Adam AA, Colman RW. Specific inhibition of plasma kallikrein modulates chronic granulomatous intestinal and systemic inflammation in genetically susceptible rats. FASEB J. 1998; 12:325–333. PMID: 9506476.
crossref
121. Kaplan AP, Kay AB, Austen KF. A prealbumin activator of prekallikrein. 3. Appearance of chemotactic activity for human neutrophils by the conversion of human prekallikrein to kallikrein. J Exp Med. 1972; 135:81–97. PMID: 5009705.
122. Stadnicki A, DeLa Cadena RA, Sartor RB, Bender D, Kettner CA, Rath HC, Adam A, Colman RW. Selective plasma kallikrein inhibitor attenuates acute intestinal inflammation in Lewis rat. Dig Dis Sci. 1996; 41:912–920. PMID: 8625762.
crossref
123. McCall RD, Haskill S, Zimmermann EM, Lund PK, Thompson RC, Sartor RB. Tissue interleukin 1 and interleukin-1 receptor antagonist expression in enterocolitis in resistant and susceptible rats. Gastroenterology. 1994; 106:960–972. PMID: 8144001.
crossref
124. Yamada T, Sartor RB, Marshall S, Specian RD, Grisham MB. Mucosal injury and inflammation in a model of chronic granulomatous colitis in rats. Gastroenterology. 1993; 104:759–771. PMID: 7680014.
crossref
125. Marcus R, Watt J. Colonic ulceration in young rats fed degraded carrageenan. Lancet. 1971; 2:765–766. PMID: 4106253.
crossref
126. Watt J, Marcus R. Experimental ulcerative disease of the colon in animals. Gut. 1973; 14:506–510. PMID: 4581171.
crossref
127. Moyana T, Lalonde JM. Carrageenan-induced intestinal injury: possible role of oxygen free radicals. Ann Clin Lab Sci. 1991; 21:258–263. PMID: 1859164.
128. Gross V, Arndt H, Andus T, Palitzsch KD, Schölmerich J. Free radicals in inflammatory bowel diseases pathophysiology and therapeutic implications. Hepatogastroenterology. 1994; 41:320–327. PMID: 7959565.
129. Benard C, Cultrone A, Michel C, Rosales C, Segain JP, Lahaye M, Galmiche JP, Cherbut C, Blottière HM. Degraded carrageenan causing colitis in rats induces TNF secretion and ICAM-1 upregulation in monocytes through NF-kappaB activation. PLoS One. 2010; 5:e8666. PMID: 20072622.
Table 1
Summarised methods for inducing experimental inflammatory bowel disease using chemical agents
kjpp-18-279-i001
TOOLS
Similar articles