Pharmacophore an International Research Journal
Pharmacophore
Submit Manuscript
Open Access | Published: 2022 - Issue 2

A COMPREHENSIVE REVIEW OF IRRITABLE BOWEL SYNDROME SCREENING MODELS FOR DRUG RESEARCH AND DEVELOPMENT

Ankita Wal1*, Pranay Wal1, Neha Verma1, Ashish Srivastava1, Awani K Rai1, Saurabh Kosey2

 

  1. Department of Pharmacy, Ranveer Singh Institute of Technology (Pharmacy), Kanpur, India.
  2. Department of Pharmacy Practice, ISF College Moga, Punjab, India.

ABSTRACT

IBS is a common digestive disorder (IBS). It is marked by visceral, hypersensitivity, and an alter in bowel habits. It is mostly caused by smoking, stress, variations in gut flora, and genetic variants. Because there is currently no particular treatment to cure IBS, it is critical to evaluate the benefits and drawbacks of existing IBS animal models, utilize these models, and construct better models for drug research and development.  The main goal of this paper is to study different models of irritable bowel syndrome to recreate IBS symptoms and find the molecular mechanisms that cause the disorder and utilize them in the development of medications that have the potential to be useful in the treatment of IBS. The report's material was generated from review articles and research papers from 1981 to 2021 using keywords like Stress, Brain-gut axis, Trinitrobenzene sulfonic acid, Acetic acid and so on. IBS has no recognized cause or treatment. so, using these models to build successful IBS medication. The pathogenesis of IBS is still poorly understood and psychosocial stress of various origins has been assigned a significant impact. As psychosocial stressors, animal models such as neonatal mother separation, water avoidance stress, and wrap restraint stress have been created to mimic IBS symptomatology and discover the biological pathways associated with the disease. In addition, other models such as antidiarrheal and anti-inflammatory are also used. The investigation of these models has resulted in the development of medications that may be efficacious in IBS management.

Keywords: Irritable bowel syndrome, Acetic acid, Brain-gut axis, Stress


Introduction

Irritable bowel syndrome (IBS) is an illness that occurs in the stomach and intestine that causes stomach discomfort, stool irregularity, and retention of fluid and gas in the stomach. IBS pervasiveness estimates range from 1.1 percent and 45 percent. it is hugely disruptive to a sufferer's regular lifestyle [1]. Symptoms may range from minor to highly debilitating, and women are 1.5 to 2 times more likely than males. Although there has been no reported death with IBS [2] Nowadays, the fundamental pathophysiological processes of IBS are yet unknown. However, greater epithelial permeability, swelling, visceral sensitivity, and changes in brain-gut connection perform a vital in the development of irritable bowel syndrome [3]. The origin and pathogenesis are yet unknown, reflected in the scarcity and ineffectiveness of present medical interventions. Furthermore, in recent generations, FGIDs were categorized as diseases of brain-gut association, (Figure 1) That emphasizing the bidirectional interaction among central and peripheral systems, and offering up novel research opportunities in FGID using anxiety and depression rodent models [4]. It has been shown that various stimuli have a vital impact on the onset and progression of IBS. Animal models mimicking the pathophysiology and signs of IBS are significant for IBS studies and may support the implementation of novel therapies. The majority of IBS animal models are produced using various stress stimuli. IBS animal models are classified into three categories based on the action locations of the stimulators: central stimulus produced animal models, peripheral stimulus produced animal models, and combined central and peripheral stimulus produced complicated animal models. Central stimulation alters brain activity and, as a result, impacts gut function via the brain-gut axis. IBS-like symptoms have been induced by peripheral stimulation via the intestinal nervous system (Figure 2).

Figure 1. Stress alters brain-gut function and plays a vital role in the development of irritable bowel syndrome

 

Figure 2. The different Animal models used in the development of irritable bowel syndrome

Stress Related-Model of IBS

Water Avoidance Stress (WAS) Induced Model

Irritable bowel syndrome is commonly caused by mental stress. Prolonged or severe stress likely causes long-term alterations in the central nervous system (CNS), triggering IBS problems [5].

The most common method to construct this model is to put an animal in a Plexiglas tank with a block attached to the ground's center. The tank is full of clean, room-temperature liquid. And the block is 1 cm higher than the water level. The animal is put upon this block for a continuous 1 h daily for a consecutive ten-day. Various studies have found that a 10-day WAS can enhance an animal's visceral hypersensitivity, which is a common symptom of IBS [6-8] (Figure 3).

Brandesi et al. [9] introduced this method, which exhibited a brief somatic antinociceptive reaction in conjunction with prolonged visceral hyperalgesia

Da Silva et al. [10] improved the methodology for establishing the WAS model by putting the animal on the block for 4 days in a row for 4 hours each day. They discovered that CWAS might cause visceral hypersensitivity and modify gut flora by limiting the growth of Lactobacillus farciminis, as seen in IBS patients. Myers examined rats under 1 hour of stress every day for 7 days in a row and discovered that chronic stress can cause prolonged visceral hypersensitivity, which can be alleviated by glucocorticoid receptor blockers and mineralocorticoid receptor antagonists [11].

 

Figure 3. Water avoidance stress (WAS) induced model

Restraint Stress-Induced Animal Model

Williams [12] presented the first restraint stress model, which confined the upper portion of the rat's body for 24 hours. This model demonstrated a reduction of intestinal transport as well as an increase in feces without the development of ulcers. It is regarded as a typical IBS model. Improved restraint stress models are now commonly employed (Figure 4).

Another study performed by, Lv et al. gave anesthesia using ether and tied the shoulder, upper arms, and chest of animals with paper tape to avoid itching the head and face for 1 hour, but animals' other behaviors were not restricted. This model demonstrated enhanced colonic movement, higher defecation with loose stools, and visceral sensitivity, indicating that it can be employed for mobility and visceral sensitivity research.

According to Liu et al. [13], short-term restraint stress can cause a transitory elevation in reactivity to nociceptive stress but not affect specific muscle cell contraction. Restraint stress can cause certain variations in the gastrointestinal system that are commonly associated with IBS, but it is not suitable for lengthy treatment since it might cause somatic harm.

 

Figure 4. Restraint stress-induced animal model

Neonatal Maternal Separation (NMS) Induced Animal Model

In this method, new-born rats were isolated from their moms for 3 hours every day on postnatal days 2-14. NMS has been shown to cause visceral hypersensitivity as well as alterations in the HPA-axis [14-17]. According to Barouei et al. [15], NMS causes higher Adrenocorticotropic Hormone (ACTH) concentrations in plasma and fecal counts of aerobes, anaerobes, enterococci, clostridia, and Escherichia (E. coli), but decreased plasma IgA concentrations.

Another study conducted during postnatal days 2 to 14, by Miquelet et al. [16] isolated wild-type C57Bl/AJ babies from their mom for three hours each day, which harmed the fecal Faecalibacterium prausnitzii number. According to Zhou's findings, the amount of Fusobacterium, which can diminish the extent of visceral hypersensitivity, was substantially reduced in C57Bl/AJ mice that received NMS [17].

 

Colonic Irritation Model of IBS

Castor-Oil Induced Diarrhea

Rats (150-250gm) of any such sex were starved for 18 hours. These are classified into four categories (n=6). Normal saline (2 mL/kg) was given orally to the first category, which served as the control. The standard drug, Loperamide (2 mg/kg) was treated orally as a preparation for the second category. The test drug was administered orally as a suspension to the third and fourth category at doses of 100 mg/kg and 200 mg/kg, respectively. All rats in each group were given 1ml of castor oil orally following 60 minutes of treatment, and the liquid fecal contents and frequency of bowel movement were recorded for up to 4 hours in the transparent boxes with filter paper at the base. Mass of paper before and after defecation was observed [18] (Figure 5).

 

Figure 5. Castor-oil induces colonic irritation that leads to the diarrheal condition

Zymosan-Induced Generalized Inflammation (zigi) Model

Zymosan is a compound generated from the yeast Saccharomyces cerevisiae's cell wall. It is made up of polysaccharide chains of varying molecular masses, with roughly 73% polysaccharides, 15% proteins, and 7% lipids and inorganic components [19]. When injected into animals, it causes inflammation by activating a variety of inflammatory processes [20].

In rodents, a strong zymosan dosage (0.8-1.0 mg/g body mass) administered intraperitoneally causes a three-phase sickness. The rats suffer acute peritonitis after receiving zymosan injections. During the first two days, they are quite unwell, as seen by ruffled fur, frequent bowel movements, sluggish nature, and a decrease in muscle weight. The rodents are leukopenic during this phase, with high oxygen demand [21], myeloperoxidase concentrations (showing neutrophil growth) in the lungs and peritoneum [22, 23], and endothelial permeability [24].

Inflammatory Model of IBS

Nonsteroidal anti-inflammatory medications (NSAIDS) are routinely used to treat inflammatory conditions. However, these medications have a number of side effects, including stomach irritation, ulceration that leads to gastrointestinal bleeding, and perforation. As a result, in recent years, there has been a significant surge in the search for phytochemicals and natural sources with anti-inflammatory characteristics [25]. For example, poncirin administration effectively decreased pain behaviours in all inflammatory pain experimental models, indicating poncirin's promising analgesic potential in inflammatory pain conditions [26].

 

Acetic Acid-Induced Colitis

Acetic acid-produced colitis is a widely used and conveniently induced model [27-29]. Acetic acid-induced colitis is a model of Irritable bowel syndrome that, in terms of etiology, histological characteristics, and inflammatory mediator profile, is very similar to clinical IBD [30-35] Intrarectal administration of a dilute acetic acid solution causes a non-transmural inflammatory process marked by high neutrophil infiltration into the intestinal tissue, massive cell death of the mucosal and submucosal layers, vascular dilation, edema, and submucosal ulceration, all of which are symptoms of human colitis [33, 35-38] (Figure 6).

It is expected that Protons are generated within the intracellular space by the protonated form of the acid, potentially causing enormous intracellular acidification and severe epithelium damage. To induce colitis with acetic acid, the rodents are anaesthetized by ether and then fasted for 24 hours. Following that, 12 ml of (3-4% acetic acid) is infused 6-5 cm proximal to the anus edge using a medical-grade polyurethane tube for enteral feeding (external diameter 2 mm). After 15-30 seconds of administration, the liquid is removed, and the rodents are slaughtered, then both hemoglobin and colons are taken 24-48 hours following colitis initiation for several histological and biochemical studies [37, 39-43]. Other researchers have shown that intracolonic treatment of 4 ml of 4% acetic acid at a dosage of 5 ml/kg induces colitis in rats [44-48].

 

Figure 6. Acetic acid produces inflammation and gastric mucosal damage

Acetic Acid–Induced Writhing Test

The acetic acid–induced abdominal writhing test was performed in mice to assess the peripheral pain response. This approach depends on acetic acid injection to contract the abdominal cavity. 45 minutes after the last medication delivery, 10 mL/kg of 0.6 percent acetic acid solution was administered intravenously. Following the 5-minute waiting time, the number of writhing was recorded for 10 minutes [49].

2, 4, 6-Trinitrobenzene Sulfonic Acid (TNBS)

TNBS promotes transmural swelling in the stomach and evokes cell-mediated immune responses identical to that seen in IBD sufferers [50, 51].

One of the most widely used technique for inducing colitis in rodents includes the use of 10 mg. TNBS was blended with 0.25 mL of 50% ethanol before being infused in male/female Wistar rats through a medical-grade polyurethane catheter (external diameter 2 mm) for enteral feeding at 8 cm proximal to the anal margin. Face-down posture is kept for 13 minutes after hapten installation to minimize leaking and to ensure uniform dispersion of the hapten. Decapitation of rats is performed after 2-6 days of colitis development to measure the extent of colonic inflammation employing several histological and immunohistochemical methods [51, 52]. Various studies have changed the approach by altering the quantity and dosage of TNBS solution, as well as the alcohol content, to produce varying degrees of IBD in Sprague Dawley or Wistar rats of either gender.

Miscellaneous Models of Irritable Bowel Syndrome

Many combined animal models have been produced by exposing animals to various stimuli in order to prevent the chance of rats habituating to a single repeating stimulus and to simulate the numerous pathogenetic aspects of IBS. To develop an IBS model, Zhuang et al. [53] coupled acetic acid with restraint stress. They discovered that this model exhibited gut visceral hypersensitivity, elevated concentrations of IL-4 and IL-9 in blood and intestinal mucosa, and enhanced mast cell degranulation.

Spread bury et al. [54] discovered that persistent WAS stress mixed with C. Rodentium infection increased DRG (dorsal root ganglion) excitability significantly. Because of the complicated etiology and various processes of IBS that are still unknown, it is increasingly important and practical to build more new IBS animal models utilizing integrated technical methods and to employ these novel IBS models for pathophysiologic research and therapeutic development.

Non-Rodent Models of Irritable Bowel Syndrome

Although the majority of the study has focused on rodent models, various species have been employed to examine the pathophysiology of Functional gastrointestinal disorders (FGID), such as irritable bowel syndrome (IBS).

Guinea pigs are an excellent model for studying intestinal motion and the enteric nervous system. The models employed are identical to those reported earlier in rodents, with the addition of stress models such as water avoidance and CRH injection [55, 56]. Various pharmacological techniques, such as gavage with mustard oil and serotonin or TRH injection, have been employed in guinea pigs as a model of altered GI transit. Mustard oil, when given orally, causes an increase in transit time in the upper GI (oesophagus) and a drop-in transit time in the lower GI (colon) [57]. Ricin oleic acid-induced guinea pig defecation is inhibited by a particular tachykinin receptor NK2 antagonist [58].

In rabbits, intracolonic Zymosan infusion causes intestinal irritation that is alleviated by a tachykinin NK2 receptor antagonist [59].

Pigs have a similar GI system to humans, with similarly sized anatomy, growth, and diet choice, which are obviously totally distinct in rodents [60]. Furthermore, as compared to rats, the enteric nervous system phenotype is comparable to the human equivalent, with more complex inter-neuronal connections and plexi [61]. Pigs have a more well-developed CNS with a complicated behavioral reaction to psychosocial stimuli [62], making them a better model for the GI tract's response to early life stress in humans. In pigs, weaning is regarded as a very stressful (both psychological and physical) experience that promotes a gut barrier dysfunction [63].

Behavioural Testing

The rodents were put through psychological tests in the sequence listed below: Y maze, elevated-plus maze, and forced swim test (Figure 7).

 

Figure 7. Psychological testing evaluated anxiety and depressive state in rodents

Y-MAZE

The maze test is utilized to examine short-term memory by analyzing the exploration activity of the three sections of the Y-shaped apparatus, as reported by the Kokkinidis group [64]. The maze utilized in this study has 3 sections (40 cm length, 8 cm diameter, and 15 cm tall, all joined at 120-degree angles) and equilateral triangular center space. For 8 minutes, the rat was put at the end of one section and permitted to travel in the maze openly. We used the spontaneous alternation indicator to test short-term memory.

Elevated-Plus Maze

To examine apprehensive behaviors, the elevated maze experiment (EPM) was used, which consisted of a cross-like four-section device set 50 cm above from floor having two sections surrounded with 30 cm high fences and the other two uncovered. The rat was put there at intersection of the open and closed section and given 5 minutes to travel in the maze. During this period, the Pellow group observed the entry and spending time in each section, as well as traveling sessions, for symptoms of anxiousness during a 5-minute test [65].

Forced Swim Test

An altered version of Porsolt's forced swim test (FST) for rodents was used to detect behavioral distress. The technique entails keeping the animals in a transparent cylindrical glass (30 cm wide, 59 cm tall) covered in liquid (15 cm, 26 °C). while the swimming movement of the escape behavior are evaluated. The rodents are subjected to test settings for six minutes, with the first two min dedicated to acclimatization and the final four minutes dedicated to evaluating a sequence of behavioral parameters that indicate depression: swimming, inactivity (floating), and striving behavior [66].

Conclusion

IBS's pathophysiology is complicated and not entirely understood because it is multifaceted. Human participants are forbidden from intervention research. Animal models help study the pathophysiology of IBS without the risks associated with human studies. Every model has advantages and disadvantages. The principal purpose of this review is to provide an overview of IBS models that investigators may employ for their gastrointestinal protection investigations. Following a review of IBS models, we discovered that the models mentioned above are frequently used based on pharmacological requirements, and the model chosen is also influenced by the model's outcome and time. The investigation of these models has resulted in the development of medications that may be efficacious in IBS management.

Acknowledgments: I am grateful to the pharmacy faculty at PSIT for motivating me to write this review article.

Conflict of interest: None

Financial support: None

Ethics statement: None

References

1.        Piche T, Ducrotté P, Sabate JM, Coffin B, Zerbib F, Dapoigny M, et al. Impact of functional bowel symptoms on quality of life and fatigue in quiescent Crohn disease and irritable bowel syndrome. Neurogastroenterol Motil. 2010;22(6):626-e174.

2.        Nageeb AN, Alsulami MS, Alshammari MT, Attar AA. An overview on irritable bowel syndrome diagnosis and management in primary health care centers. Pharmacophore. 2020;11(5):151-5.

3.        Wang Y, Bi Z, Wang E, Sun B, Zheng Y, Zhong LL, et al. Rodent model of irritable bowel syndrome. Int J Gastroenterol Disord Ther. 2017;4:131.

4.        Camilleri M, Buéno L, Andresen V, De Ponti F, Choi MG, Lembo A. Pharmacologic, pharmacokinetic, and pharmacogenomic aspects of functional gastrointestinal disorders. Gastroenterology. 2016;150(6):1319-31.

5.        Mertz H. Role of the brain and sensory pathways in gastrointestinal sensory disorders in humans. Gut. 2002;51(suppl 1):i29-33.

6.        Wang Z, Ocampo MA, Pang RD, Bota M, Bradesi S, Mayer EA, et al. Alterations in prefrontal-limbic functional activation and connectivity in chronic stress-induced visceral hyperalgesia. PLoS One. 2013;8(3):e59138.

7.        Shi HL, Liu CH, Ding LL, Zheng Y, Fei XY, Lu L, et al. Alterations in serotonin, transient receptor potential channels and protease-activated receptors in rats with irritable bowel syndrome attenuated by Shugan decoction. World J Gastroenterol. 2015;21(16):4852.

8.        Xu D, Gao J, Gillilland III M, Wu X, Song I, Kao JY, et al. Rifaximin alters intestinal bacteria and prevents stress-induced gut inflammation and visceral hyperalgesia in rats. Gastroenterology. 2014;146(2):484-96.

9.        Bradesi S, Schwetz I, Ennes HS, Lamy CM, Ohning G, Fanselow M, et al. Repeated exposure to water avoidance stress in rats: a new model for sustained visceral hyperalgesia. Am J Physiol Gastrointest Liver Physiol. 2005;289(1):G42-53.

10.     Da Silva S, Robbe-Masselot C, Raymond A, Mercade-Loubière M, Salvador-Cartier C, Ringot B, et al. Spatial localization and binding of the probiotic Lactobacillus farciminis to the rat intestinal mucosa: influence of chronic stress. PLoS One. 2015;10(9):e0136048.

11.     Myers B, Greenwood-Van Meerveld B. Differential involvement of amygdala corticosteroid receptors in visceral hyperalgesia following acute or repeated stress. Am J Physiol Gastrointest Liver Physiol. 2012;302(2):G260-6.

12.     Williams CL, Villar RG, Peterson JM, Burks TF. Stress-induced changes in intestinal transit in the rat: a model for irritable bowel syndrome. Gastroenterology. 1988;94(3):611-21.

13.     Sun Y, Liu FL, Song GQ, Qian W, Hou XH. Effects of acute and chronic restraint stress on visceral sensitivity and neuroendocrine hormones in rats. Chin J Dig Dis. 2006;7(3):149-55.

14.     Greenwood-Van Meerveld B, Prusator DK, Johnson AC. Animal models of gastrointestinal and liver diseases. Animal models of visceral pain: pathophysiology, translational relevance, and challenges. Am J Physiol Gastrointest Liver Physiol. 2015;308(11):G885-903.

15.     Barouei J, Moussavi M, Hodgson DM. Effect of maternal probiotic intervention on HPA axis, immunity and gut microbiota in a rat model of irritable bowel syndrome. PLoS One. 2012;7:e46051.

16.     Miquel S, Martin R, Lashermes A, Gillet M, Meleine M, Gelot A, et al. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep. 2016;6(1):1-8.

17.     Zhou XY, Li M, Li X, Long X, Zuo XL, Hou XH, et al. Visceral hypersensitive rats share common dysbiosis features with irritable bowel syndrome patients. World J Gastroenterol. 2016;22(22):5211.

18.     Teferi MY, Abdulwuhab M, Yesuf JS. Evaluation of in vivo antidiarrheal activity of 80% methanolic leaf extract of Osyris quadripartita Decne (Santalaceae) in Swiss Albino Mice. J Evid Based Integr Med. 2019;24:2515690X19833340.

19.     Fizpatrick FW, DiCarlo FJ. Zymosan. Ann N Y Acad Sci. 1964;118(4):235-61.

20.     Pillemer L, Ecker EE. Anti-complementary factor in fresh yeast. J Biol Chem. 1941;137(1):139-42.

21.     Goris RJ, Boekholtz WK, van Bebber IP, Nuytinck JK, Schillings PH. Multiple-organ failure and sepsis without bacteria: an experimental model. Arch Surg. 1986;121(8):897-901.

22.     Shayevitz JR, Miller C, Johnson KJ, Rodriguez JL. Multiple organ dysfunction syndrome: end organ and systemic inflammatory response in a mouse model of nonseptic origin. Shock. 1995;4(6):389-96.

23.     Rao TS, Currie JL, Shaffer AF, Isakson PC. In vivo characterization of zymosan-induced mouse peritoneal inflammation. J Pharmacol Exp Ther. 1994;269(3):917-25.

24.     Deng X, Wang X, Andersson R. Alterations in endothelial barrier permeability in multiple organs during overactivation of macrophages in rats. Shock. 1996;6(2):126-33.

25.     Sundar RD, Arunachalam S. Anti-inflammatory and antifungal activity of Dracaena victoria leaf extract. Bangladesh J Pharmacol. 2020;15(1):44-5.

26.     Afridi R, Khan AU, Khalid S, Shal B, Rasheed H, Ullah MZ, et al. Anti-hyperalgesic properties of a flavanone derivative Poncirin in acute and chronic inflammatory pain models in mice. BMC Pharmacol Toxicol. 2019;20(1):1-6.

27.     MacPherson BR, Pfeiffer CJ. Experimental production of diffuse colitis in rats. Digestion. 1978;17(2):135-50.

28.     Noa M, Más R, Carbajal D. Effect of D-002 on acetic acid-induced colitis in rats at single and repeated doses. Pharmacol Res. 2000;41(4):391-5.

29.     Sasaki S, Hirata I, Maemura K, Hamamoto N, Murano M, Toshina K, et al. 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(1):23-8.

30.     Elson CO, Sartor RB, Tennyson GS, Riddell RH. Experimental models of inflammatory bowel disease. Gastroenterology. 1995;109(4):1344-67.

31.     Gonzalez R, Rodriguez S, Romay C, González A, Armesto J, Remirez D, et al. Anti-inflammatory activity of phycocyanin extract in acetic acid-induced colitis in rats. Pharmacol Res. 1999;39(1):55-9.

32.     Gorgulu S, Yagci G, Kaymakcioglu N, Özkara M, Kurt B, Ozcan A, et al. Hyperbaric oxygen enhances the efficiency of 5-aminosalicylic acid in acetic acid–induced colitis in rats. Dig Dis Sci. 2006;51(3):480-7.

33.     Nakhai LA, Mohammadirad A, Yasa N, Minaie B, Nikfar S, Ghazanfari G, et al. Benefits of Zataria multiflora Boiss in experimental model of mouse inflammatory bowel disease. Evid Based Complement Alternat Med. 2007;4(1):43-50.

34.     Bitiren M, Karakilcik AZ, Zerin M, Ozardalı I, Selek S, Nazlıgül Y, et al. Protective effects of selenium and vitamin E combination on experimental colitis in blood plasma and colon of rats. Biol Trace Elem Res. 2010;136(1):87-95.

35.     Hartmann RM, 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(8):2038-44.

36.     Jurjus AR, Khoury NN, Reimund JM. Animal models of inflammatory bowel disease. J Pharmacol Toxicol Methods. 2004;50(2):81-92.

37.     Daneshmand A, Rahimian R, Mohammadi H, Ejtemaee-Mehr S, Tavangar SM, Kelishomi RB, et al. Protective effects of lithium on acetic acid-induced colitis in rats. Dig Dis Sci. 2009;54(9):1901-7.

38.     Closa D, Folch‐Puy E. Oxygen free radicals and the systemic inflammatory response. IUBMB life. 2004;56(4):185-91.

39.     Millar AD, Rampton DS, Chander CL, Claxson AW, Blades S, Coumbe A, et al. Evaluating the antioxidant potential of new treatments for inflammatory bowel disease using a rat model of colitis. Gut. 1996;39(3):407-15.

40.     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(1):69-77.

41.     Yalniz M, Demirel U, Orhan C, Bahcecioglu IH, Ozercan IH, Aygun C, et al. Nadroparin sodium activates Nrf2/HO-1 pathway in acetic acid-induced colitis in rats. Inflammation. 2012;35(3):1213-21.

42.     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(1):57-66.

43.     Iseri SO, Ersoy Y, Ercan F, Yuksel M, Atukeren P, Gumustas K, et al. The effect of sildenafil, a phosphodiesterase‐5 inhibitor, on acetic acid‐induced colonic inflammation in the rat. J Gastroenterol Hepatol. 2009;24(6):1142-8.

44.     Grisham MB, Granger DN. Neutrophil-mediated mucosal injury. Dig Dis Sci. 1988;33(3):6S-15S.

45.     Yamada T, Marshall S, Specian RD, Grisham MB. A comparative analysis of two models of colitis in rats. Gastroenterology. 1992;102(5):1524-34.

46.     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(3):800-11.

47.     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(1):469-75.

48.     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(9):4104-12.

49.     Hussein MC, Bektas N, Ozturk Y, Arslan R. Antinociception Induced by Moringa Stenopetela (Baker f.) Cufod. Leaves Extract and Possible Mechanisms of Action. Braz J Pharm Sci. 2022;58.

50.     De Almeida AB, Sanchez-Hidalgo M, Martín AR, Luiz-Ferreira A, Trigo JR, Vilegas W, et al. Anti-inflammatory intestinal activity of Arctium lappa L.(Asteraceae) in TNBS colitis model. J Ethnopharmacol. 2013;146(1):300-10.

51.     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(6):437-46.

52.     da Silva MS, Sánchez-Fidalgo S, Talero E, Cárdeno A, da Silva MA, Villegas W, et al. Anti-inflammatory intestinal activity of Abarema cochliacarpos (Gomes) Barneby & Grimes in TNBS colitis model. J Ethnopharmacol. 2010;128(2):467-75.

53.     Zhuang Z, Zhang L, Wang X, Tao L, Lv B. PDIA3 gene induces visceral hypersensitivity in rats with irritable bowel syndrome through the dendritic cell-mediated activation of T cells. Peer J. 2016;4:e2644.

54.     Spreadbury I, Ochoa‐Cortes F, Ibeakanma C, Martin N, Hurlbut D, Vanner SJ. Concurrent psychological stress and infectious colitis is key to sustaining enhanced peripheral sensory signaling. Neurogastroenterol Motil. 2015;27(3):347-55.

55.     Hussain Z, Da Hyun Jung YJ, Park H. The effect of trimebutine on the overlap syndrome model of Guinea pigs. J Neurogastroenterol Motil. 2018;24(4):669.

56.     Hussain Z, Kim HW, Huh CW, Lee YJ, Park H. The effect of peripheral CRF peptide and water avoidance stress on colonic and gastric transit in guinea pigs. Yonsei Med J. 2017;58(4):872-7.

57.     Park JJ, Chon NR, Lee YJ, Park H. The effects of an extract of Atractylodes Japonica rhizome, SKI3246 on gastrointestinal motility in guinea pigs. J Neurogastroenterol Motil. 2015;21(3):352.

58.     Tanaka T, Tanaka A, Nakamura A, Matsushita K, Imanishi A, Matsumoto-Okano S, et al. Effects of TAK-480, a Novel Tachykinin NK2–Receptor Antagonist, on Visceral Hypersensitivity in Rabbits and Ricinoleic Acid–Induced Defecation in Guinea Pigs. J Pharmacol Sci. 2012;120(1):15-25.

59.     Accarie A, Vanuytsel T. Animal Models for Functional Gastrointestinal Disorders. Front Psychiatry. 2020:1265.

60.     Brown DR, Timmermans JP. Lessons from the porcine enteric nervous system. Neurogastroenterol Motil. 2004;16:50-4.

61.     Timmermans JP, Hens J, Adriaensen D. Outer submucous plexus: an intrinsic nerve network involved in both secretory and motility processes in the intestine of large mammals and humans. Anat Rec. 2001;262(1):71-8.

62.     Gieling ET, Schuurman T, Nordquist RE, Staay F. The pig as a model animal for studying cognition and neurobehavioral disorders. Mol Funct Model Neuropsychiatry. 2011:359-83.

63.     Medland JE, Pohl CS, Edwards LL, Frandsen S, Bagley K, Li Y, et al. Early life adversity in piglets induces long‐term upregulation of the enteric cholinergic nervous system and heightened, sex‐specific secretomotor neuron responses. Neurogastroenterol Motil. 2016;28(9):1317-29.

64.     Kokkinidis L, Anisman H. Dissociation of the effects of scopolamine and d-amphetamine on a spontaneous alternation task. Pharmacol Biochem Behav. 1976;5(3):293-7.

65.     Pellow S, Chopin P, File SE, Briley M. Validation of open: closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods. 1985;14(3):149-67.

66.     Porsolt RD, Bertin A, Jalfre MJ. Behavioral despair in mice: a primary screening test for antidepressants. Arch Int Pharmacodyn Ther. 1977;229(2):327-36.

QR code:

Short Link:
Views: 222

Downloads: 31
Quick Access

Pharmacophore
ISSN: 2229-5402

Pharmacophore
© 2024 All rights reserved
Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.