• DOI: 10.1016/S0021-5198(19)41298-5
  • Corpus ID: 303501

Clinicopathologic study of dextran sulfate sodium experimental murine colitis.

  • K. Shichijo , Mutsumi Matuu , +3 authors I. Sekine
  • Published in Laboratory investigation; a… 1 August 1993
  • Laboratory investigation; a journal of technical methods and pathology

2,188 Citations

Clinical and histopathological features of dextran sulfate sodium induced acute and chronic colitis associated with dysplasia in rats.

  • Highly Influenced

Sodium arsenite reduces severity of dextran sulfate sodium-induced ulcerative colitis in rats

Histological analysis of murine colitis induced by dextran sulfate sodium of different molecular weights., dextran sulfate sodium (dss) colitis in rats (clinical, structural, and ultrastructural aspects), an experimental model of colitis induced by dextran sulfate sodium from acute progresses to chronicity in c57bl/6: correlation between conditions of mice and the environment, role of appendix and spleen in experimental colitis., dysplasia and cancer in the dextran sulfate sodium mouse colitis model. relevance to colitis-associated neoplasia in the human: a study of histopathology, b-catenin and p53 expression and the role of inflammation., acute colitis induced by dextran sulfate sodium progresses to chronicity in c57bl/6 but not in balb/c mice: correlation between symptoms and inflammation., dextran sulfate sodium-induced colitis occurs in severe combined immunodeficient mice., clinical assessment using disease activity index in experimental animal model of inflamatory bowel disease induced dextran sulfate sodium, related papers.

Showing 1 through 3 of 0 Related Papers

Clinicopathologic study of dextran sulfate sodium experimental murine colitis.

Full text links.

Add to Saved Papers

Get 1-tap access

Related Resources

Trending papers, for the best experience, use the read mobile app.

Mobile app image

Get seemless 1-tap access through your institution/university For the best experience, use the Read mobile app

Toggle icon

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Pascal and Francis Bibliographic Databases

Help

Selection :

  • Selected items ( 1 )

Clinicopathologic study of dextran sulfate sodium experimental murine colitis

Laboratory investigation . 1993, Vol 69, Num 2, pp 238-249 ; ref : 19 ref

Sauf mention contraire ci-dessus, le contenu de cette notice bibliographique peut être utilisé dans le cadre d’une licence CC BY 4.0 Inist-CNRS / Unless otherwise stated above, the content of this bibliographic record may be used under a CC BY 4.0 licence by Inist-CNRS / A menos que se haya señalado antes, el contenido de este registro bibliográfico puede ser utilizado al amparo de una licencia CC BY 4.0 Inist-CNRS

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Searching the Web

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Clinicopathologic study of dextran sulfate sodium experimental murine colitis.

展开 

BACKGROUND: We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and to discover the earliest histopathologic changes that lead to colitis. EXPERIMENTAL DESIGN: Acute colitis was induced in Swiss-Webster mice by 7 days of oral DSS with animals sacrificed daily. Chronic colitis was induced by: (a) 7 days of oral DSS followed by 7 days of H2O (for 1, 2, and 3 cycles) and (b) 7 days of oral DSS followed by 14 and 21 days of H2O. In each experimental group, the entire colons were examined histologically and correlated with clinical symptoms. RESULTS: Acute clinical symptoms (diarrhea and/or grossly bloody stool) were associated with the presence of erosions and inflammation. More importantly, the earliest histologic changes which predated clinical colitis was loss of the basal one-third of the crypt (day 3), which progressed with time to loss of the entire crypt resulting in erosions on day 5. The earliest changes were very focal and not associated with inflammation. Inflammation was a secondary phenomena and only became significant after erosions appeared. Animals treated with only 7 days of DSS followed by 14 and 21 days of H2O developed a chronic colitis with the following histologic features: areas of activity (erosions and inflammation), inactivity, crypt distortion, florid epithelial proliferation and possible dysplasia. These changes were similar to animals given 3 cycles of DSS. The clinical disease activity index correlated significantly with pathologic changes in both the acute and chronic phases of the disease. CONCLUSIONS: The mechanism of DSS colitis is presently unknown. However, the finding of crypt loss without proceeding or accompanying inflammation suggests that the initial insult is at the level of the epithelial cell with inflammation being a secondary phenomena. This may be a good model to study how early mucosal changes lead to inflammation and the biology of the colonic enterocyte. Chronic colitis induced after only 7 days of DSS may serve as a useful model to study the effects of pharmacologic agents in human inflammatory disease and mechanisms of perpetuation of inflammation. Finally, we believe that this model has the potential to study the dysplasia cancer sequence in inflammatory disease.

Colon Animals Mice, Inbred Strains Mice Colitis Dextran Sulfate Time Factors Female

clinicopathologic study of dextran sulfate sodium experimental murine colitis

通过 文献互助 平台发起求助,成功后即可免费获取论文全文。

我们已与文献出版商建立了直接购买合作。

你可以通过身份认证进行实名认证,认证成功后本次下载的费用将由您所在的图书馆支付

您可以直接购买此文献,1~5分钟即可下载全文,部分资源由于网络原因可能需要更长时间,请您耐心等待哦~

clinicopathologic study of dextran sulfate sodium experimental murine colitis

百度学术集成海量学术资源,融合人工智能、深度学习、大数据分析等技术,为科研工作者提供全面快捷的学术服务。在这里我们保持学习的态度,不忘初心,砥砺前行。 了解更多>>

百度云

©2024 Baidu 百度学术声明 使用百度前必读

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 01 September 2007

Dextran sulfate sodium-induced colitis-associated neoplasia: a promising model for the development of chemopreventive interventions

  • Margie Lee Clapper 1 ,
  • Harry Stanley Cooper 2 &
  • Wen-Chi Lee Chang 1  

Acta Pharmacologica Sinica volume  28 ,  pages 1450–1459 ( 2007 ) Cite this article

8781 Accesses

145 Citations

3 Altmetric

Metrics details

Individuals diagnosed with ulcerative colitis face a significantly increased risk of developing colorectal dysplasia and cancer during their lifetime. To date, little attention has been given to the development of a chemopreventive intervention for this high-risk population. The mouse model of dextran sulfate sodium (DSS) -induced colitis represents an excellent preclinical system in which to both characterize the molecular events required for tumor formation in the presence of inflammation and assess the ability of select agents to inhibit this process. Cyclic administration of DSS in drinking water results in the establishment of chronic colitis and the development of colorectal dysplasias and cancers with pathological features that resemble those of human colitis-associated neoplasia. The incidence and multiplicity of lesions observed varies depending on the mouse strain used (ie, Swiss Webster, C57BL/6J, CBA, ICR) and the dose (0.7%–5.0%) and schedule (1–15 cycles with or without a subsequent recovery period) of DSS. The incidence of neoplasia can be increased and its progression to invasive cancer accelerated significantly by administering DSS in combination with a known colon carcinogen (azoxymethane (AOM), 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP)) or iron. More recent induction of colitis-associated neoplasia in genetically defined mouse strains has provided new insight into the role of specific genes (ie, adenomatous polyposis coli ( Apc ), p53 , inducible nitric oxide synthase ( iNOS ), Msh2 ) in the development of colitis-associated neoplasias. Emerging data from chemopreventive intervention studies document the efficacy of several agents in inhibiting DSS-induced neoplasia and provide great promise that colitis-associated colorectal neoplasia is a preventable disease.

Similar content being viewed by others

clinicopathologic study of dextran sulfate sodium experimental murine colitis

The food additive EDTA aggravates colitis and colon carcinogenesis in mouse models

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Interactions between diet and gut microbiota in cancer

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Molecular mechanisms in colitis-associated colorectal cancer

Article pdf.

Eaden JA, Abrams KR, Mayberry JF . The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001; 48 : 526–35.

Article   CAS   Google Scholar  

Hardy RG, Meltzer SJ, Jankowski JA . ABC of colorectal cancer. Molecular basis for risk factors. BMJ 2000; 321 : 886–9.

Harpaz N, Talbot IC . Colorectal cancer in idiopathic inflammatory bowel disease. Sem Diagn Pathol 1996; 13 : 339–57.

CAS   Google Scholar  

Ekbom A, Helmick C, Zack M, Adami HO . Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med 1990; 323 : 1228–33.

Rutter MD, Saunders BP, Wilkinson KH, Rumbles S, Schofield G, Kamm MA, et al . Thirty-year analysis of a colonoscopic surveillance program for neoplasia in ulcerative colitis. Gastroenterology 2006; 130 : 1030–8.

Article   Google Scholar  

Farrell RJ, Peppercorn MA . Ulcerative colitis. Lancet 2002; 359 : 331–40.

Kiesslich R, Fritsch J, Holtmann M, Koehler HH, Stolte M, Kanzler S, et al . Methylene blue-aided chromoendoscopy for the detection of intraepithelial neoplasia and colon cancer in ulcer-ative colitis. Gastroenterology 2003; 124 : 880–8.

Kern SE, Redston M, Seymour AB, Caldas C, Powell SM, Kornacki S, et al . Molecular genetic profiles of colitis-associated neoplasms. Gastroenterology 1994; 107 : 420–8.

Aust DE, Terdiman J P, Willenbucher R F, Chang CG, Molinaro-Clark A, Baretton GB, et al . The APC/β-catenin pathway in ulcerative colitis-related colorectal carcinomas. A mutational analysis. Cancer 2002; 94 : 1421–7.

Hussain SP, Amstad P, Raja K, Ambs S, Nagashima M, Bennett WP, et al . Increased p 53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res 2000; 60 : 3333–7.

CAS   PubMed   Google Scholar  

Brentnall TA, Crispin DA, Bronner M P, Cherian S P, Hueffed M, Rabinovitch PS, et al . Microsatellite instability in nonneoplastic mucosa from patients with chronic ulcerative colitis. Cancer Res 1996; 56 : 1237–40.

Chang CL, Marra G, Chauhan DP, Ha HT, Chang DK, Ricciardiello L, et al . Oxidative stress inactivates the human DNA mismatch repair system. Am J Physiol Cell Physiol 2002; 283 : C148–54.

Fogt F, Vortmeyer AO, Goldman H, Giordano TJ, Merino MJ, Zhuang Z . Comparison of genetic alterations in colonic adenoma and ulcerative colitis-associated dysplasia and carcinoma. Hum Pathol 1998; 29 : 131–6.

Yashiro M, Carethers JM, Laghi L, Saito K, Slezak P, Jaramillo E, et al . Genetic pathways in the evolution of morphologically distinct colorectal neoplasms. Cancer Res 2001; 61 : 2676–83.

Wong NACS, Harrison DJ . Colorectal neoplasia in ulcerative colitis—recent advances. Histopathology 2001; 39 : 221–34.

Ohkusa T . Production of experimental ulcerative colitis in hamsters by dextran sulfate sodium and change in intestinal microflora. Jpn J Gastroenterol 1985; 82 : 1327–36.

Okayasu I, Hatekayama 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.

Cooper HS, Murthy SNS, Shah RS, Sedergran DJ . Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest 1993; 69 : 238–49.

Tanaka T, Kohno H, Suzuki R, Hata K, Sugie S, Niho N, et al . Dextran sodium sulfate strongly promotes colorectal carcinogenesis in Apc Min/+ mice: inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms. Int J Cancer 2006; 118 : 25–34.

Cooper HS, Murthy S, Kido K, Yoshitake H, Flanigan A . Dysplasia and cancer in the dextran sulfate sodium mouse colitis model. Relevance to colitis-associated neoplasia in the human: a study of histopathology, B-catenin and p53 expression and the role of inflammation. Carcinogenesis 2000; 21 : 757–68.

Nosho K, Yamamoto H, Adachi Y, Endo T, Hinoda Y, Imai K . Gene expression profiling of colorectal adenomas and early invasive carcinomas by cDNA array analysis. Br J Cancer 2005; 92 : 1193–200.

Cooper HS, Everley L, Chang W-C, Pfeiffer G, Lee B, Murthy S, et al . The role of mutant Apc in the development of dysplasia and cancer in the mouse model of dextran sulfate sodium-induced colitis. Gastroenterology 2001; 121 : 1407–16.

Coudry RA, Cooper HS, Gary M, Lubet RA, Chang WCL, Clapper ML . Correlation of inhibition of colitis-associated dysplasia by celecoxib with degree of inflammation in the mouse model of DSS-induced colitis. Proc Am Assoc Cancer Res 2004; 45 : 548.

Google Scholar  

Tanaka T, Kohno H, Suzuki R, Yamada Y, Sugie S, Mori H . A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci 2003; 94 : 965–73.

Suzuki R, Kohno H, Sugie S, Nakagama H, Tanaka T . Strain differences in the susceptibility to azoxymethane and dextran sodium sulfate-induced colon carcinogenesis in mice. Carcinogenesis 2006; 27 : 162–9.

Takahashi M, Nakatsugi S, Sugimura T, Wakabayashi K . Frequent mutations of the β- catenin gene in mouse colon tumors induced by azoxymethane. Carcinogenesis 2000; 21 : 1117–20.

Tanaka T, Suzuki R, Kohno H, Sugie S, Takahashi M, Wakabayashi K . Colonic adenocarcinomas rapidly induced by the combined treatment with 2-amino-1-methyl-6-phenylimidazo[4,5- b ]pyridine and dextran sodium sulfate in male ICR mice possess β- catenin gene mutations and increases immunoreactivity for β-catenin, cyclooxygenase-2 and inducible nitric oxide synthase. Carcinogenesis 2005; 26 : 229–38.

Aust DE, Terdiman JP, Willenbucher RF, Chew K, Ferrell L, Florendo C, et al . Altered distribution of β-catenin, and its binding proteins E-cadherin and APC, in ulcerative colitis-related colorectal cancers. Mod Pathol 2001; 14 : 29–39.

Souza RF, Yin J, Smolinski KN, Zou TT, Wang S, Shi YQ, et al . Frequent mutation of the E2F-4 cell cycle gene in primary human gastrointestinal tumors. Cancer Res 1997; 57 : 2350–3.

Sugimura T . Nutrition and dietary carcinogens. Carcinogenesis 2000; 21 : 387–95.

Nagao M . A new approach to risk estimation of food-borne carcinogens—heterocyclic amines—based on molecular information. Mutat Res 1999; 431 : 3–12.

Sugimura T . Overview of carcinogenic heterocyclic amines. Mutat Res 1997; 376 : 211–9.

Nishikawa A, Imazawa T, Kuroiwa Y, Kitamura Y, Kanki K, Ishii Y, et al . Induction of colon tumors in C57BL/6J mice fed MeIQx, IQ, or PhIP followed by dextran sulfate sodium treatment. Toxicol Sci 2005; 84 : 243–8.

Carrier J, Aghdassi E, Cullen J, Allard JP . Iron supplementation increases disease activity and vitamin E ameliorates the effect in rats with dextran sulfate sodium-induced colitis. J Nutr 2002; 132 : 3146–50.

Seril DN, Liao J, Ho KL, Warsi A, Yang CS, Yang GY . Dietary iron supplementation enhances DSS-induced colitis and associated colorectal carcinoma development in mice. Dig Dis Sci 2002; 47 : 1266–78.

Cooper HS, Chang WCL, Coudry R, Gary MA, Everley L, Spittle CS, et al . Generation of a unique strain of multiple intestinal neoplasia (Apc +/Min-FCCC ) mice with significantly increased numbers of colorectal adenomas. Mol Carcinog 2005; 44 : 31–41.

Burmer GC, Crispin DA, Kolli VR, Haggitt RC, Kulander BG, Rubin CE, et al . Frequent loss of a p53 allele in carcinomas and their precursors in ulcerative colitis. Cancer Commun 1991; 3 : 167–72.

Chang WC, Coudry RA, Clapper ML, Zhang X, Williams KL, Spittle CS, et al . Loss of p53 enhances the induction of colitis-associated neoplasia by dextran sulfate sodium. Carcinogenesis 2007 June 8; [Epub ahead of print].

Fujii S, Fujimori T, Kawamata H, Takeda J, Kitajima K, Omotehara F, et al . Development of colonic neoplasia in p53 deficient mice with experimental colitis induced by dextran sulphate sodium. Gut 2004; 53 : 710–6.

Wink DA, Vodovotz Y, Laval J, Laval F, Dewhirst MW, Mitchell JB . The multifaceted roles of nitric oxide in cancer. Carcinogenesis 1998; 19 : 711–21.

Jaiswal M, LaRusso NF, Gores GJ . Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol 2001; 281 : G626–G34.

Seril DN, Liao J, Yang GY . Colorectal carcinoma development in inducible nitric oxide synthase-deficient mice with dextran sulfate sodium-induced ulcerative colitis. Mol Carcinog 2007; 46 : 341–53.

Fleisher AS, Esteller M, Harpaz N, Leytin A, Rashid A, Xu Y, et al . Microsatellite instability in inflammatory bowel disease-associated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1 . Cancer Res 2000; 60 : 4864–8.

Cawkwell L, Sutherland F, Murgatroyd H, Jarvis P, Gray S, Cross D, et al . Defective hMSH2/hMLH1 protein expression is seen infrequently in ulcerative colitis associated colorectal cancers. Gut 2000; 46 : 367–9.

Kohonen-Corish MR, Daniel JJ, te Riele H, Buffinton GD, Dahlstrom JE . Susceptibility of Msh2-deficient mice to inflammation-associated colorectal tumors. Cancer Res 2002; 62 : 2092–7.

Oshima M, Dinchuk JE, Kargman SL, Oshima H, Hancock B, Kwong E, et al . Suppression of intestinal polyposis in Apc Δ716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell 1996; 87 : 803–9.

Steinbach G, Lynch P, Phillips RKS, Wallace MH, Hawk E, Gordon GB, et al . The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med 2000; 342 : 1946–52.

Mahadevan U, Loftus EV, Tremaine WJ, Sandborn WJ . Safety of selective cyclooxygenase-2 inhibitors in inflammatory bowel disease. Am J Gastroenterol 2002; 97 : 910–4.

Kapicioglu S, Baki AH, Sari M, Ozdemir F, Kavgaci H . Does nimesulide induce gastric mucosal damage? “A double-blind randomized placebo-controlled trial”. Hepatogastroenterology 2000; 47 : 1183–5.

Kohno H, Suzuki R, Sugie S, Tanaka T . Suppression of colitis-related mouse colon carcinogenesis by a COX-2 inhibitor and PPAR ligands. BMC Cancer 2005; 5 : 46.

Niho N, Takahashi M, Kitamura T, Shoji Y, Itoh M, Noda T, et al . Concomitant suppression of hyperlipidemia and intestinal polyp formation in Apc-deficient mice by peroxisome proliferator-activated receptor ligands. Cancer Res 2003; 63 : 6090–5.

Girnun GD, Smith WM, Drori S, Sarraf P, Mueller E, Eng C, et al . APC-dependent suppression of colon carcinogenesis by PPARγ. Proc Natl Acad Sci USA 2002; 99 : 13771–6.

Lefebvre AM, Chen I, Desreumaux P, Najib J, Fruchart JC, Geboes K, et al . Activation of the peroxisome proliferator-activated receptor γ promotes the development of colon tumors in C57BL/6J-APC Min/+ mice. Nat Med 1998; 4 : 1053–7.

Saez E, Tontonoz P, Nelson MC, Alvarez JG, Ming UT, Baird SM, et al . Activators of the nuclear receptor PPAR? enhance colon polyp formation. Nat Med 1998; 4 : 1058–61.

Tanaka T, Kohno H, Yoshitani S, Takashima S, Okumura A, Murakami A, et al . Ligands for peroxisome proliferator-activated receptors α and γ inhibit chemically induced colitis and formation of aberrant crypt foci in rats. Cancer Res 2001; 61 : 2424–8.

Gary MA, Coudry RA, Cooper HS, Litwin S, Concha AC, Lubet RA, et al . 5-Aminosalicylic acid inhibits colitis-associated dysplasia in the mouse model of AOM/DSS-induced colitis. Proc Am Assoc Cancer Res Third Annual International Conference on Frontiers in Cancer Prevention Research 2004; 45 : A81.

Grimm GR, Dirisina R, Lee G, Tang Y, Manjali J, Hanson NB, et al . 5-ASA suppressed induction of dysplasia on the AOM/DSS model: Correlation with inhibition of Wnt/beta-catenin transcriptional activity. Gastroenterology 2005; 128 ( Suppl 2): A200.

Seril DN, Liao J, Ho KL, Yang CS, Yang GY . Inhibition of chronic ulcerative colitis-associated colorectal adenocarcinoma development in a murine model by N -acetylcysteine. Carcinogenesis 2002; 23 : 993–1001.

Morris AJ, Malbon CC . Physiological regulation of G protein-linked signaling. Physiol Rev 1999; 79 : 1373–430.

Goldstein JL, Brown MS . Regulation of low-density lipoprotein receptors: implications for pathogenesis and therapy of hyper-cholesterolemia and atherosclerosis. Circulation 1987; 76 : 504–7.

Suzuki S, Tajima T, Sassa S, Kudo H, Okayasu I, Sakamoto S . Preventive effect of fluvastatin on ulcerative colitis-associated carcinogenesis in mice. Anticancer Res 2006; 26 : 4223–8.

Raboy V . Myo-Inositol-1,2,3,4,5,6-hexakisphosphate. Phytochemistry 2003; 64 : 1033–43.

Fox CH, Eberl M . Phytic acid (IP6), novel broad spectrum anti-neoplastic agent: a systematic review. Complement Ther Med 2002; 10 : 229–34.

Liao J, Seril DN, Yang AL, Lu GG, Yang GY . Inhibition of chronic ulcerative colitis associated adenocarcinoma development in mice by inositol compounds. Carcinogenesis 2007; 28 : 446–54.

Tanaka T, Kawabata K, Kakumoto M, Makita H, Hara A, Mori H, et al . Citrus auraptene inhibits chemically induced colonic aberrant crypt foci in male F344 rats. Carcinogenesis 1997; 18 : 2155–61.

Kohno H, Suzuki R, Curini M, Epifano F, Maltese F, Gonzales SP, et al . Dietary administration with prenyloxycoumarins, auraptene and collinin, inhibits colitis-related colon carcinogenesis in mice. Int J Cancer 2006; 118 : 2936–42.

Download references

Author information

Authors and affiliations.

Divisions of Population Science, Fox Chase Cancer Center, Philadelphia, 19111, PA, USA

Margie Lee Clapper & Wen-Chi Lee Chang

Divisions of Medical Science, Fox Chase Cancer Center, Philadelphia, 19111, PA, USA

Harry Stanley Cooper

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Margie Lee Clapper .

Additional information

This work was supported by USPHS grants CN05121, CA099122, and CA06927 from the National Cancer Institute, an ACS Institutional Research Grant, and by an appropriation from the Commonwealth of Pennsylvania. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the National Cancer Institute or the American Cancer Society.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Clapper, M., Cooper, H. & Chang, WC. Dextran sulfate sodium-induced colitis-associated neoplasia: a promising model for the development of chemopreventive interventions. Acta Pharmacol Sin 28 , 1450–1459 (2007). https://doi.org/10.1111/j.1745-7254.2007.00695.x

Download citation

Received : 31 May 2007

Accepted : 23 July 2007

Issue Date : 01 September 2007

DOI : https://doi.org/10.1111/j.1745-7254.2007.00695.x

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • colorectal colitis
  • chemoprevention
  • dextran sulfate sodium
  • mouse models

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • J Biomed Biotechnol
  • v.2012; 2012

Dextran Sodium Sulphate Colitis Mouse Model: Traps and Tricks

Inflammatory bowel disease (IBD) is a complex multifactorial disease of unknown etiology. Thus, dozens of different animal models of IBD have been developed in past decades. Animal models of IBD are valuable and indispensable tools that provide a wide range of options for investigating involvement of various factors into the pathogenesis of IBD and to evaluate different therapeutic options. However, the dextran sulphate sodium (DSS-) induced colitis model has some advantages when compared to other animal models of colitis. It is well appreciated and widely used model of inflammatory bowel disease because of its simplicity. It has many similarities to human IBD, which are mentioned in the paper. In spite of its simplicity and wide applicability, there are also traps that need to be taken into account when using DSS model. As demonstrated in the present paper, various factors may affect susceptibility to DSS-induced lesions and modify results.

1. Introduction

Inflammatory bowel disease (IBD) is a complex multifactorial disease [ 1 – 3 ]. It commonly refers to ulcerative colitis (UC) and Crohn's disease (CD), the two chronic conditions that involve inflammation of the intestine. IBD is common in developed countries, with up to 1 in 200 of individuals of Northern European region affected by these disease [ 4 ]. Patients with IBD present several clinically challenging problems for physicians. Despite recent advances in treatment, there remains a need for a safe, well-tolerated therapy with a rapid onset, and increased capacity for maintaining long-term remission [ 5 ].

In past decades, dozens of different animal models of IBD have been developed. These models can be broadly divided into spontaneous colitis models, inducible colitis models, genetically modified models, and adoptive transfer models [ 6 – 8 ]. Although these models do not represent the complexity of human disease, they are valuable and indispensable tools that provide a wide range of options for investigating involvement of various factors into the pathogenesis of IBD and evaluate different therapeutic options. Chemically induced murine models of intestinal inflammation are one of the most commonly used models because they are simple to induce, the onset, duration, and severity of inflammation are immediate and controllable. Both dextran sulphate sodium (DSS) and trinitrobenzene sulfonic acid (TNBS-) induced colitis are well-established animal models of mucosal inflammation that have been used for over 2 decades in the study of IBD pathogenesis and preclinical studies [ 6 – 8 ]. The DSS-induced colitis model has some advantages when compared to other animal models of colitis. For example, an acute, chronic, or relapsing model can be produced easily by changing the concentration of administration of DSS (and cycle in rats and other strains of mice). Moreover, dysplasia that resembles the clinical course of human UC occurs frequently in the chronic phase of DSS-induced colitis. DSS-induced model for studying colitis-associated carcinogenesis has been recently reviewed by others [ 9 , 10 ]. Furthermore, studies that validated DSS model by using different therapeutic agents for human IBD show that DSS-induced colitis can be used as a relevant model for the translation of mice data to human disease [ 11 ]. Thus, the aim of the present paper is to give a concise introduction of different factors that may be involved in the patogenesis of DSS-induced colitis and need to be taken into account when using this model. At the same time few aspects of applicability and further investigation of this model are mentioned.

2. Induction of DSS Colitis

First report on the use of DSS dates back in the year 1985, when Ohkusa et al. published their investigation on DSS-induced colitis in hamsters [ 12 ]. Thereafter, DSS colitis was induced also in mice [ 13 ]. Today there are numerous studies using DSS-induced colitis model to investigate pathogenesis of colitis and different factors affecting colitis. Colitis is induced by addition of DSS to drinking water. Depending on the concentration, the duration, and frequency of DSS administration, the animals may develop acute or chronic colitis or even colitis-induced dysplastic lesions. Mice show differential susceptibilities and responsiveness to DSS-induced colitis. The varying responses to DSS appear to be dependent on not only DSS (concentration, molecular weight, duration of DSS exposure, manufacturer, and batch) but also genetic (strain and substrain, gender) and microbiological (microbiological state and intestinal flora) factors of animal, which are disscussed in the present paper ( Figure 1 ). Colitis onset and severity may vary with many of these factors. Stress can be one of them [ 14 ]. Differences in the DSS susceptibility do not correlate with differences in the consumption of DSS-supplemented water [ 15 ]. However, there is a need to monitor DSS consumption, especially when animals are exposed to different therapeutic strategies that may lower consumption of DSS (increased fluid intake or thirst) [ 16 ].

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.001.jpg

Schematic simplified representation of various factors that can influence the susceptibility, onset, severity, and responsiveness to DSS-induced colitis.

3. The Molecular Weight of DSS

DSS is sulfated polysaccharide with a highly variable molecular weight, ranging from 5 kDa to up to 1400 kDa. It was found that the molecular weight of DSS is very important factor in the induction of colitis [ 17 ] or colitis-induced dysplastic lesions (carcinogenicity) [ 18 ]. The severity of colitis [ 17 ] and carcinogenic activity [ 18 ] differs with the administration of DSS at different molecular weights (i.e., 5 kDa, 40 kDa, and 500 kDa). The most severe colitis in BALB/c mice was observed when mice were treated with DSS of 40 kDa molecular weight, while mice treated with DSS of 5 kDa developed milder form of colitis. Mice treated with DSS of 500 kDa had no lesions in the large bowel [ 17 ]. Similarly, carcinogenic activity in colon was induced by DSS of 54 kDa, while DSS of larger (520 kDa) or smaller (9.5 kDa) molecular weights induced no carcinogenic activity [ 18 ]. Examination of uptake and tissue distribution of DSS by histochemical techniques showed that failure in the induction of colitis with 500 kDa DSS is due to its high molecular weight that prevents passage of the molecule through the mucosal membrane [ 17 ]. Molecular weight of DSS can affect location of colitis as well. Mice treated with 40 kDa DSS developed most severe diffuse colitis in the middle and distal third of the large bowel, while mice treated with 5 kDa DSS developed relatively patchy lesions mainly in the cecum and upper colon [ 17 ].

3.1. Uptake and Tissue Distribution of DSS

The histochemical analyses of uptake and tissue distribution of DSS at 40 kDa molecular weight demonstrated that DSS penetrates the mucosal membrane in the intestine. One day after DSS treatment small amounts of DSS were found in macrophages in large bowel and mesenteric lymph nodes and in the liver Kupffer cells. At day 3 DSS was noticed in a few macrophages in the spleen and 7 days after the start of 5 day DSS treatment in the kidney and in a few macrophages in the small intestine. DSS was seen in the liver Kupffer cells even 8 weeks after DSS removal [ 19 ]. During chronic phase of DSS colitis, considerable amounts of DSS were also found in the spleen [ 20 ]. In other organs and tissues (for instance brain, lung, heart, thymus, stomach, and duodenum), DSS was not observed [ 19 ]. Major excretion routes of DSS are urine and feces. Thus, presence of DSS in the epithelial cells of the proximal renal tubules after 7 days of DSS treatment is an indication of the excretion process of absorbed DSS [ 19 ]. DSS is resistant to degradation by intestinal microflora or the effects of different pH conditions (4.0–7.5) and anaerobic incubation [ 21 ].

4. Clinical and Histological Features of DSS-Induced Colitis

Clinical and histopathological features of DSS-colitis reflect those seen in human IBD. Acute colitis is usually induced by continuous administration of 2–5% DSS for short period (4–9 days). Chronic colitis may be induced by continuous treatment of low concentrations of DSS or cyclical administration of DSS. For instance, 4 cycles of DSS treatment for 7 days followed by 10 days of water ( Table 4 ). Clinical manifestation of DSS colitis in acute phase may include weight loss, diarrhea, occult blood in stools, piloerection, anaemia, and eventually death. However, clinical manifestations in chronic phase of colitis usually do not reflect severity of inflammation or histologic features found in large bowel.

Genetically engineered mouse models in the pathogenesis of DSS colitis.

GEM modelBackground (breeder)Age gender MSMW of DSSDSS treatmentEndSRef
TLR2
TLR4
TLR2/4 C57BL/10ScSn = 10–12nr40 kDa3.5% for 7 daysDay 8[ ]
5–8 wks
TLR4 C57BL/6JMale, female 2.5% for 5 daysDay 5
MyD88 (Jax) = 5-6nr36–50 kDa 2.5% for 5/7 days Day 12/14[ ]
TLR2
TLR4 F2
MyD88 129/SvJ × C57BL/6 SPF40 kDa2% for 7 daysDay 0–28[ ]
8 wks
Male 1.2% for 8 days
MyD88 nr = 10SPF50 kDa2.5% for 8 daysDay 8[ ]
10 wks
C57BL/6Male [ , ]
IL-18 (nr) = 20SPF50 kDa2% for 8 daysDay 12
3 cycles of
C57BL/67–9 wks 2% for 5 days +Day 5
IL-15 (Taconic) = 11nr36–50 kDa5 days of water Day 25[ ]
C57BL/6JMale, female
TNF- (Shimizu, Japan) = 15nr8 kDa4.5% for 7 daysDay 7[ ]
8–10 wks
Male
MRP1 FVB = 8SC40 kDa3% for 7daysDay 7[ ]
C57BL/68-9 wks Day 4, 7
Mtgr1 (nr) Male, femalenr40–50 kDa3% for 4days Wk 6, 10[ ]
7-8 wks
C57BL/6Male, female 3% for 5 days
SOCS-1 (nr) = 14SPF36–50 kDa(4% for 7 days)Day 14[ ]
5 cycles of
C57BL/6J8–12 wk 2% for 7 days +
IRF-1 (Jax) = 15nr40 kDa7 days of waterWk 10[ ]
8–10 wk
Female
PPAR nr = 20nr35–40 kDa2.5% for 7 daysDay 7[ ]
C57BL/6J
PPAR (nr) = 34nr36–44 kDa2.5% for 7 daysDay 0,2,7[ ]
C57BL/6
TPH1 (Taconic) = 10nr40 kDa5% for 5 daysDay 5[ ]
Bk2R C57BL/68–12 wk ns
C3 (Jax) = 8nr36–40 kDa3% for 10 daysDay 10[ ]
C57BL/6Male, female
iNOS (Jax) = 5nr40 kDa2.5% for 9 daysDay 3,9[ , ]
eNOS
nNOS 129/Sv × C57BL/6Male, female
e/nNOS (Jax) = 5nr40 kDa2.5% for 9 daysDay 3,9ns[ ]
5–10 wks
C57BL/6Female
APN (nr) = 5–7nr40 kDa2% for 5 daysDay 5,10[ ]
8–10 wks
C57BL/6JMale
APN (Clea Japan) = 11SPF36–50 kDa0.5% for 15 daysDay 15[ ]
Vhl Day 5
Hif-1 6–8 wk 2.5% or 5% for1 yearns
Arnt nrnrnr35–40 kDa5 days ns[ ]

GEM: genetically engineered mice, N : number of mice pre group, MS: microbiological state, MW: molecular weight, S: susceptibility to DSS in comparison to wild-type mice (mice of the same strain without deletion), ↓: significantly lower, SFP: specific pathogen-free conditions, SC: standard conditions, ns: nonsignificant, nr: not reported, ↑: significantly higher, wk: week, TLR: toll-like receptor, MyD88: myeloid differential protein, IL: interleukine, MRP: multi drug resistant protein, Mtgr: myeloid translocation gene related-1, SOCS: suppressor of cytokine signaling, IRF: Interferon regulatory factor, PPAR: peroxisome proliferator-activated receptor, TPH: tryptophan hydroxylase, Bk2R: bradykinin type 2 receptor, C3: complement, APN: adiponectine, Vhl: von Hippel-Lindau tumor suppressor protein, Hif: hypoxia-inducible factor, Arnt: aryl hydrocarbon nuclear translocator.

Histological changes in DSS-induced colitis can be classified as acute (early) and chronic (advanced). Typical histological changes of acute DSS-colitis are mucin depletion, epithelial degeneration, and necrosis leading to disappearance of epithelial cells ( Figure 2(a) ). The latter is accompanied by neutrophils infiltration of lamina propria and submucosa, cryptitis, crypt abscesses (Figures 3(b) and ​ and4), 4 ), and phlegmonous inflammation in mucosa and submucosa ( Figure 2(b) ). Usually also shallow erosions appear ( Figure 2(b) ). Rarely there is a vacuolar hydropic degeneration of cells ( Figure 3(a) ). Transepithelial migration of neutrophils into mucosal epithelium is termed cryptitis. Migration of numerous neutrophils through mucosal epithelium into crypt lumen results in the formation of crypt abscess. Cryptitis and crypt abscesses are common histological feature of human IBD but rarely reported in DSS-induced colitis using haematoxylin-eosin staining method [ 22 ]. In contrast, we found cryptitis and crypt abscesses in half of DSS-treated C57BL/6JOlaHsd mice, while in BALB/cAnNHsd mice cryptitis was rarely observed (unpublished data). However, we used Kreyberg-Jareg trichrom staining method that differentially stains acid mucopolysaccharides blue and thus contributes to better distinction between normal and aberrant mucosa. Interestingly, the presence of neutrophils in the epithelial lining of colon mucosa (cryptitis) and in the lumen of crypts (crypt abscess) was reported in DSS-treated C57BL/6OlaHsd mice when investigators used immunofluorescent staining against neutrophils [ 23 ].

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.002.jpg

(a) Disappearance of crypts. (b) Erosion and phlegmonous inflammation of mucosa and submucosa. Kreyberg trichrom stain (acid mucopolysaccharides are stained blue). C57BL/6JOlaHsd female mice are exposed to 3% DSS solution for 5 days followed by drinking water for 7 days.

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.003.jpg

(a) Vacuolar hydropic degeneration of cells. (b) Epithelial necrosis, cryptitis, and crypt abscesses. Kreyberg trichrom stain (acid mucopolysaccharides are stained blue). C57BL/6JOlaHsd female mice exposed to 3% DSS solution for 5 days followed by drinking water for 21 days.

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.004.jpg

Arrows denote crypt abscesses (neutrophils) in the lumen of crypts. Kreyberg trichrom stain (acid mucopolysaccharides are stained blue). C57BL/6JOlaHsd male mice are exposed to 3% DSS solution for 5 days followed by drinking water for (a) 7 days and (b) 28 days.

Chronic changes appear few weeks after DSS application. They consist of mononuclear leucocytes infiltration, crypt architectural disarray ( Figure 5(a) ), increasing the distance (widening of the gap) between crypt bases and muscularis mucosa, deep mucosal lymphocytosis ( Figure 4(a) ), and transmural inflammation ( Figure 6 ). It is widely believed that transmural inflammation is infrequent feature of DSS-induced colitis. In contrast, we observed transmural inflammation in C57BL/6JOlaHsd mice as well as in BALB/cAnNHsd mice, consonant with observation by Melgar et al. [ 22 ]. Moreover, in chronic colitis of C57BL/6JOlaHsd mice we even observed transmural inflammation with lymphoid follicles ( Figure 6(b) ), which is histological feature of Crohn's disease and was until now not observed in DSS colitis model.

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.005.jpg

(a) Mononuclear leucocytes infiltration, crypt architectural disarray, and deep mucosal lymphocytosis. (b) Focally there is a moderate epithelial regeneratory atypia simulating dysplasia (arrow). Kreyberg trichrom stain (acid mucopolysaccharides are stained blue). C57BL/6JOlaHsd male mice are exposed to 3% DSS solution for 5 days followed by drinking water for 28 days.

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.006.jpg

(a) Focal transmural chronic colitis (skip lesion). C57BL/6JOlaHsd female mice are exposed to 3% DSS solution for 5 days followed by drinking water for 28 days. (b) Transmural inflammation with lymphoid follicles in subserosa (arrows) and chronic erosion. C57BL/6JOlaHsd female mice are exposed to 3% DSS solution for 9 days followed by drinking water for 28 days. Kreyberg trichrom stain (acid mucopolysaccharides are stained blue).

Rarely reported characteristic change is also reepithelisation of rectal and distal colonic erosions by squamous epithelium that evidently originates from anal squamous mucosa ( Figure 7 ). In some slides islands of squamous epithelium are found surrounded by colonic mucosa proximally and distally, but in most cases with deeper sections there appeared a continuity of squamous epithelium distally, suggesting an irregular shape of a squamous cell regenerate. Focally sometimes there is a moderate epithelial regeneratory atypia simulating dysplasia ( Figure 5(b) ). Most frequently it is found at the edge of chronic erosions.

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.007.jpg

(a) and (b) reepithelisation of rectal and distal colonic erosion by squamous epithelium (arrows). C57BL/6JOlaHsd male mice are exposed to 3% DSS solution for 5 days followed by drinking water for 7 days. Kreyberg trichrom stain (acid mucopolysaccharides are stained blue).

We observed reepithelisation of distal part of colon as early as day 5 of DSS treatment in C57BL/6JOlaHsd but not in BALB/cAnNHsd mice. According to our experience reepithelisation of distal colon with squamous epithelium is frequent observation in C57BL/6JOlaHsd mice 1–4 weeks after DSS removal. The distance of reepithelisation with squamous epithelium that we measured under microscope ranged from 0.5–6.5 mm of colon length.

Histological changes seen in mouse DSS-induced colitis are the features of IBD in man, some of them of ulcerative colitis (regular rectal localization) and some of Crohn's disease (transmural inflammation with disseminated lymphoid follicles, focal lesions) [ 24 , 25 ].

Besides all in Figure 1 , mentioned factors that affect features of DSS-induced colitis preparation of tissues for histological examination may also be one of the reasons for discrepancy among studies. According to our experience as well as recommendation of other investigators [ 15 ] longitudinal sections as well as at least few sections of the same slide at intermediate distance of not less than 0.1 mm may better reflect the actual damage of DSS colitis than cross sections because of the patchy nature of DSS-induced lesions. In addition, the choice of staining method may also affect histological observations. For instance, cryptitis, crypt abscesses as well as squamous reepithelisation are easier to diagnose in sections stained with Kreyberg-Jareg method than hematoxylin-eosin method.

5. The Role of Intestinal Flora in the Pathogenesis of DSS-Induced Colitis

Intestinal microflora and their products have been implicated in the pathogenesis of human IBD [ 2 , 26 – 28 ] and in several animal models [ 29 ]. The importance of the intestinal flora is directly supported by studies of some murine models where colitis is not observed when they are maintained in a gnotobiotic state but rapidly emerges when they are reconstituted with bacteria that are considered normal constituents of luminal flora (explained in detail by Nell et al. [ 29 ]). It has been demonstrated that intestinal flora is implicated in the pathogenesis of DSS colitis in mice as well. First who suggested contribution of colonic bacteria or their products in the development of colitis in this model were Okayasu et al. [ 13 ]. They observed increased numbers of Enterobacteriaceae, Bacteroidaceae, and Clostridium spp. in the colons of mice affected by DSS colitis [ 13 ]. Another suggestion in this direction was given by Hans et al. [ 30 ], who reported that treatment with antibiotics reduced infiltration of granulocytes in the mucosa and improved histological signs of DSS colitis in acute phase [ 30 ]. The role of commensal bacteria and innate immunity in the development of intestinal inflammation has been further demonstrated by Hudcovic et al. [ 31 ]. When DSS-treated immunocompetent BALB/c and immunodeficient SCID mice (lacking T and B lymphocytes) were maintained under conventional conditions, both strains developed substantial changes in the colon mucosa. BALB/c mice showed complete loss of the surface epithelium and severe infiltration of inflammatory cells. To evaluate susceptibility to DSS colitis in gnotobiotic conditions BALB/c and SCID mice were transferred into isolators for germ-free rearing by special gnotobiological techniques. Interestingly, BALB/c and SCID mice reared in germ-free conditions (lacking any intestinal microflora) developed only minor signs of mucosal inflammation after DSS treatment [ 31 ]. This finding indicates that the presence of microflora facilitates the inflammation in DSS-induced colitis. Similar findings were observed by Kitajima et al. [ 32 ]. They used IQI/Jic mice maintained in germ-free conditions and their littermates that were conventionalized with feces obtained from SPF BALB/c mice three weeks before DSS administration. Histopathological findings revealed that IQI/Jic mice under germ-free conditions had no lesions indicative of colitis in the large intestine 3 days after 5% DSS treatment, while IQI/Jic mice in conventional conditions developed focal erosions in the large bowel, mostly in the cecum and proximal colon. Interestingly, when both groups of mice were treated with 1% DSS for 14 days, mice under germ-free conditions developed slight inflammatory cell infiltration and edema in the lamina propria of the cecum and proximal colon and sever ulcerations, hemorrhages with frequent thrombi, and slight inflammatory cell infiltration in the distal colon. In the large bowel of IQI/Jic mice in conventional conditions only focal lesions of slight inflammatory cells infiltration and edema in mucosa along the whole large bowel were observed [ 32 ]. These findings demonstrate firstly that mucosal destruction caused by DSS occurs without the involvement of intestinal microflora and secondly that intestinal microflora may play an important modifying role in the susceptibility and responsiveness to DSS-induced damage of epithelial cells. The role of intestinal microflora in DSS-induced colitis is further investigated in mice deficient for different toll-like receptors (TLR), which function as sensors of microbial infection and are critical for the initiation of inflammatory and immune defence responses [ 33 ].

6. The Role of Genetic Factors in the Pathogenesis of DSS-Induced Colitis

Similar to IBD in humans [ 3 ], genetic factors play important role in DSS-induced colitis. Differences in susceptibility and responsiveness to DSS-induced colitis among inbred strains and substrains of mice have been identified. A quantitative histological analysis of DSS-induced colitis in nine mouse strains using a standardized protocol (3.5% DSS of 36–45 kDa molecular weight for 5 days) demonstrated major differences in DSS responsiveness among strains [ 15 ]. C3H/HeJ, NOD/Ltj, and NOD-scid inbred strains are very susceptible to DSS-induced lesions, which develop in severe form mostly in the cecum. 129/SvPas and DBA/2J inbred mice are less susceptible to DSS-induced lesions and show various degrees of susceptibility to DSS, depending upon anatomical site as schematically demonstrated in the Table 1 . Interestingly, severity of DSS-induced lesions in most inbred strains increased from proximal to the distal colon. A greater susceptibility to DSS-induced colonic but not cecal lesions was observed in male mice [ 15 ].

Susceptibility to DSS-induced colitis among inbred strains (adapted from [ 15 ]).

CecumProximal colonMiddle colonDistal colon
DBA/2JBCAA
NON/LtJABAA
NON.H2 ABABAB
129/SvPasAABB
NOD-scidDAABBC
C57BL/6JBCABBC
C3H/HeJDAABC
NOD/LtJCDBBC
C3H/HeJBirDAABC

Strains that do not share the same letter in a column have significat differences in histological score for particular part of the colon after DSS administration. Letter A denotes the lowest histological score, while D denotes the highest histological score observed among the strains. — denotes no changes found in the colon (normal mucosa).

Stevceva et al. [ 35 ] demonstrated that C3H mice are more susceptible to DSS colitis than CBA/H and BALB/c. C3H mice developed severe colitis with severe inflammatory response predominantly involving ascending (proximal) colon and cecum, while CBA/H and BALB/c mice developed severe colitis mostly in distal colon. BALB/c mice, which are frequently used strain in DSS-colitis, are also less susceptible to DSS as IQI/Jic [ 32 ] or C57BL/6 [ 22 ]. The differences in the susceptibility to DSS-induced lesions were either due to genetic differences in the ability of the mucosa to withstand inflammatory damage, differences in the ability to limit the inflammatory response, or both. Melgar et al. [ 22 ] demonstrated differences in the progression of DSS colitis between two commonly used strains. After DSS withdrawal (one cycle), BALB/c mice recovered of DSS colitis, while in C57BL/6 mice colitis progressed into chronic phase [ 22 ]. The genetic factors contributing to DSS susceptibility in mice are unknown. Interestingly, some strains of mice are able to limit and eliminate DSS-induced inflammatory response in colon, while inflammatory process in C57BL/6 mice can not be repared but progresses into chronic form of colitis [ 22 ]. This indicates that genetic factors are importantly involved into regulations of inflammatory response, which is of great interest for further research.

7. Pathogenesis of DSS-Induced Colitis

It is widely accepted that DSS is toxic to colonic epithelial cells and causes defects in the epithelial barrier integrity, whereby increasing the colonic mucosal permeability to allow permeation of large molecules such as DSS. The mechanism of how DSS passes through the mucosal epithelial cells (transcellularly or paracellularly mediated via tight junctions) remains uncertain. First changes related to DSS were observed after 1 day of DSS treatment. These changes were loss of one of the components of tight junction complex [ 36 , 37 ] zonula occludens-1 (ZO-1) [ 38 ] and significantly increased expression of proinflammatory cytokines (TNF- α , IL-1 β , IFN- γ , IL-10, and IL-12) in the colon [ 39 ]. By day 3 of DSS treatment significant increase in permeability to Evan's blue was observed [ 38 ]. At this time first histological changes in the colonic mucosa in the form of basal crypts loss and increased inflammatory cells infiltration can be seen. In acute phase of DSS colitis the impairment of epithelial barrier function is associated with loss and redistribution of the tight junction proteins such as occludin, ZO-1 [ 38 ], claudin-1, -3, -4, and -5 and an increased epithelial apoptotic ratio [ 40 ]. Altered expression of tight junction proteins and increased epithelial apoptosis were reported also in human IBD. It was proposed that imbalance between apoptosis and proliferation causes relevant leaks in the epithelial barrier. This is supported by the finding that both increased apoptosis and decreased proliferation of the epithelium take place in the acute phase of DSS colitis [ 41 ]. The single layered intestinal epithelium is a physical and immunological barrier that prevents direct contact of the intestinal mucosa with the luminal microbiota.

As evidenced by the amelioration of inflammation in germ-free animals and in mice treated with antibiotics [ 27 , 30 , 31 ], DSS-induced breakdown of mucosal epithelial barrier function allows the entry of luminal antigens and microorganisms into the mucosa resulting in overwhelming inflammatory response.

Microarray analysis of the gene expression revealed that 173 genes were differentially and significantly expressed in the colon of DSS-treated mice by a factor of two or more when compared to control mice. Fifteen were previously associated with IBD in humans (shown in Table 2 ) [ 34 ]. Intestinal Na + -related transporters/channels and their regulatory proteins (NHE1,3, β -ENaC, and NHERF1,2) have been found to be downregulated in mouse colon in acute phase of DSS colitis and in mucosal biopsies from IBD patients (UC or CD) in active phase of disease [ 44 ].

Differentially expressed genes in acute phase of DSS colitis that had been previously associated with human IBD [ 34 ].

GeneNameExpressionGene      Name Expression
Interleukine-6↑ 2.6        Stromelysin 1 ↑ 22.5
Interleukine-16↓ 2.7     Membrane type 1 MMP ↑ 2.1
Interleukine-22↑ 2.4     Tissue inhibitor of metalloproteinase 1 ↑ 3.6
JE (human:MCP-1)↑ 1.8     Regenerating islet-derived 3  ↑ 14.0
MIP-1 ↑ 3.3      Pancreatitis-associated protein ↑ 79.1
Eotaxin↑ 3.3     S-100 calcium-binding protein A9 (calgranulin B) ↑ 14.0
KC (hu: GRO )↑ 2.9
LIX (hu: ENA-78)↑ 6.6      Prostaglandin-endoperoxidase synthase 2 (COX-2) ↑ 4.1

Colitis was induced in 8 wk female BALB/c mice that received 3% DSS for 7 days and were sacrificed on day 7. ↑: significantly increased expression by denoted factor, ↓: significantly decreased expression by denoted factor.

7.1. Inflammatory Response and Mediators Involved in the Pathogenesis of DSS Colitis

Numerous inflammatory mediators have been implicated in the pathogenesis of human IBD. These include cytokines, eicosanoids, reactive oxygen species, nitric oxide, and complement system activation products [ 45 – 48 ]. Similarly, DSS-induced colitis is associated with the upregulation of different cytokines, chemokines, nitric oxide [ 49 , 50 ], and inducible nitric oxide synthase (iNOS) [ 51 ]. Changes in production of inflammatory mediators in DSS-treated mice were investigated during different phases of colitis, in the serum and/or colon and by different methods (shown in Table 3 ). Increased expression of different inflammatory mediators (TNF- α , IL-1 β , IFN- γ , IL-10, and IL-12) was observed as early as the first day of DSS treatment [ 39 ]. The production of these inflammatory mediators increased progressively during DSS treatment. Different profile of inflammatory mediators in acute and chronic phase of DSS colitis was demonstrated. Acute inflammation in DSS colitis converts to a predominant Th2-mediated inflammatory response in the chronic state (lower levels of TNF- α , IL-17, and KC and elevated levels of IL-6, IFN- γ , and IL-4, IL-10) [ 43 ]. Progressive upregulation in the transcripts for Th1 cytokines (IL-12, IFN- γ , IL-1, and TNF- α ) was observed with increasing dosage of DSS [ 42 ]. Different cytokine profile in chronic phase of DSS colitis was found between BALB/c and C57BL/6J mice, which reflected the severity of inflammation or infiltration of inflammatory cells in the colon found histologically [ 22 ].

Profile of inflammatory mediators in different phases of DSS-induced colitis in mice.

Tissue/methodAnalytes measuredacute DSS colitischronic DSS colitisMouse strainref.
Colon/RT PCRIL-1, IL-4, IL-10, IL-12, IFN , TNF ↑ IL-1, IL-10, IL-12,
↑ IFN- , TNF
BALB/c[ ]
Colon/ELISA, MSIIL-1 , IL-1 , IL-12 p40, IL-12 p70, IL-17, IL-6, G-CSF, IL-18↑ IL-1 , IL-1 , IL-12 p40,
↑ IL-12 p70, IL-17, IL-6,
↑ G-CSF
↑ IL-1 , IL-1 , IL-12 p40,
↑ IL-12 p70, IL-17, IL-6,
↑ G-CSF
C57BL/6J[ ]
↑ IL-1 , IL-1 , IL-6,
↑ IL-18, G-CSF
↑ IL-12 p40BALB/c[ ]
Colon/RT PCRTNF- , IL-1 , IL-6, IL-10, IL-12, IFN- , KC, MIP-2↑ TNF- , IL-1 , IL-6,
↑ IL-10, IL-12, IFN- ,
↑ KC, MIP-2
↑ TNF- , IL-1 , IL-6, IL-10,
↑ IL-12, IFN- , KC, MIP-2
C57BL/6J[ ]
Serum/MSIIL-1 , IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, IL-17, IFN- , TNF- , GM-CSF, IP-10, KC, MCP-1, MIG, MIP-1 ↑ IL-6, IL-17, TNF- , KC↑ IL-6, IFN- , IL-4, IL-10 [ ]
Colon/Western IL-6, IL-12, IL-23, IL-17, IFN- ↑ IL-6, IFN- , IL-17↑ IL-6C57BL/6J[ ]

RT PCR: real-time PCR; MSI: multiplex sandwich immunoassay; IFN- γ : interferone gama; IL: interleukine; GM-CSF: granulocyte-macrophage colony-stimulating factor; IP-10: interferon-inducible protein; KC: keratinocyte-derived chemokine; MCP-1: monocyte chemoattractant protein; MIG: monokine induced by IFN- γ ; MIP-1 α : macrophage inflammatory protein; TNF- α : tumor necrosis factor alpha.

These inflammatory mediators not only play a role in the pathogenesis of DSS-induced colitis but are important as intervention targets against colitis as excellently described by Kawada et al. [ 52 ]. Cytokine profile in DSS colitis correlates with clinical and histological parameters as well as barrier properties. Different expression depending on strain and phase of colitis provides this model as a useful tool to dissect the role of these cytokines in the induction of inflammation and recovery from it.

Recently comprehensive study of mucosal and systemic immune responses in C57BL/6OlaHsd mice exposed to 3% DSS for 6 days has been performed using immunofluorescent staining and flow cytometry analyses. It has been shown that adaptive immune responses in this mouse strain are induced during both acute and chronic phase of colitis in all organs tested, that are, colon, spleen, and mesenteric lymph nodes as early as day 1 of DSS treatment until the end (i.e., day 25) [ 23 ]. Interplay of neutrophils, dendritic cells, macrophages, and T and B cells among spleen, mesenteric lymph nodes, and colon in DSS-treated C57BL/6OlaHsd mice in temporal fashion is schematically represented in Figure 8 .

An external file that holds a picture, illustration, etc.
Object name is JBB2012-718617.008.jpg

Schematic representation of mucosal (colon) and systemic (spleen, MLN) immune responses in C57BL/6OlaHsd mice exposed to 3% DSS for 6 days followed by water for 19 days. Measurements were made on day 0, 1, 3, 5, 8, 12, and 25 as denoted by numbers. Schematic representation is based on results obtained by Hall et al. [ 23 ]. MLN: mesenteric lymph nodes, N: neutrophils, M: macrophages, DC: dendritic cells, T: T cells, B: B cells.

8. Geneticaly Modified Mice and DSS Treatment

DSS-induced colitis is nowadays frequently used in genetically engineered mice (GEM), to study basic immunologic mechanisms of IBD and to elucidate the role of particular deleted ( −/− ) or inserted (Tg) gene in the pathogenesis of the colitis.

Toll-like receptors (TLR) are critical receptors and signal transducers for structurally conserved pathogen-associated molecular patterns of bacterial cell components (such as lipopolysaccharide) that provoke innate immune responses by stimulating macrophages/dendritic cells to produce proinflammatory cytokines. It has been shown that mice deficient for different TLR (TLR2, TLR4) or other molecules, which are implicated in signaling via TLR such as myeloid differential protein (MyD88), are very susceptible to DSS colitis, indicating important protective role of these molecules in colitis [ 33 , 53 – 55 ]. Overproduction of IL-18 or deletion of TNF- α has been shown to exacerbate DSS colitis [ 51 , 56 ]. On the other hand mice deficient for IL-15 showed reduced susceptibility to DSS colitis [ 58 ]. Mice deficit for IL-12 developed mild DSS colitis, while mice deficit for IL-18 developed severe colitis associated with high lethality [ 69 ]. Deletion of suppressor of cytokine signaling-1 (SOCS-1), which is a negative feedback molecule for cytokine signaling, increased susceptibility to DSS colitis, suggesting that SOCS-1 plays preventive role in the development of DSS-induced colitis in mice by inhibiting IFN- γ /STAT1 signaling [ 61 ]. Interferon regulatory factor-1 (IRF-1) is a transcription factor stimulated by IFN- γ and TNF- α that binds to the promoter region of inflammation-related genes, such as IFN- α , IFN- β , and iNOS. It is expressed in lymphocytes, monocytes, and a wide variety of other cell types, including enterocytes. Mice lacking IRF-1 transcription factor developed significantly increased DSS-induced colonic dysplasia [ 62 ]. Tryptophan hydroxylase-1 (TPH1) catalyzes the rate-limiting step in the synthesis of 5-hydroxytryptamine (5-HT) from tryptophan. It is an important enteric mucosal signaling molecule influencing gut physiology (motor and secretory function) following inflammation and is considered important in maintaining intestinal homeostasis. However, mice lacking TPH1 had significantly reduced susceptibility to DSS colitis [ 65 ].

Taken together, GEM treated with DSS are valuable model to test different molecules, which are involved in the mediation of inflammation such as cytokines (IL-12, IL-15, IL-18, TNF- α ), nitric oxides (iNOS, eNOS, and nNOS), complement system activation products or other molecules involved in signaling of inflammation (SOCS-1, IRF-1), to further identify bacterial factors involved in maintaining intestinal homeostasis (TLR, MyD88), to investigate the role of particular factor when totally deleted or deleted only in particular types of cells such as intestinal, (PPAR γ ΔIEpC ; Vhl ΔF/F ; Hif-1 α ΔF/F ; Arnt ΔF/F ) or CD4+ T cells (PPAR γ ΔCD4+ ) and to test therapeutic interventions based upon inhibition of particular gene expression strategies (TPH1, APN, and MRP1). In the Table 4 susceptibility to DSS-induced colitis in mice deficient for a particular gene is shown. Mouse background, gender, microbiological state, molecular weight of DSS, and DSS treatment, factors that importantly affect DSS colitis and need to be taken into account in designing investigation or evaluating results, are also stated.

9. Conclusion

DSS-induced model is simple to induce and not expensive, which makes it one of the most commonly used model of IBD to study various aspects of IBD such as pathogenesis, genetic predisposition to IBD, immune mechanisms, and role of microflora in the pathogenesis of IBD as well as bowel malignancy secondary to IBD. As demonstrated above, various factors may affect susceptibility to DSS and modify results. Representative examples of how important can be all mentioned details are studies where two groups of researchers investigated the role of adiponectine in DSS colitis. Both groups used in their studies the same mice model (mice with APN deletion) exposed to DSS treatment. They observed quite opposite results [ 66 , 67 ]. It is thus advisable to state and describe all details and conditions that may affect the DSS susceptibility.

Acknowledgment

This work was in part supported by ARRS (Slovenian Research Agency, Program P3-0054).

Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin

  • Original Articles
  • Published: September 1993
  • Volume 38 , pages 1722–1734, ( 1993 )

Cite this article

clinicopathologic study of dextran sulfate sodium experimental murine colitis

  • S. N. S. Murthy PhD 1 , 2 ,
  • Harry S. Cooper MD 1 , 2 ,
  • Helen Shim MD 1 , 2 ,
  • Rupal S. Shah BS 1 , 2 ,
  • Samih A. Ibrahim MS 1 , 2 &
  • Deborah J. Sedergran MT (ASCP) 1 , 2  

1920 Accesses

480 Citations

9 Altmetric

Explore all metrics

The use of oral and intravenous cyclosporin represents a significant advance in the therapy of refractory inflammatory bowel diseases (IBD). However, oral administration of cyclosporin is fraught with improper delivery of cyclosporin to the colon for its topical action. Because of unpredictable metabolism by cytochrome P-450 IIIA, the targeted blood level for systemic effect is not reached at low doses. Furthermore, the doses that have been used for therapy of IBD have been shown to induce several adverse side effects. Thus, an alternate method of delivering cyclosporin to the colon is desirable. In this study, the effect of intracolonically administered cyclosporin was tested for its efficacy to heal mucosal erosions in dextran sulfate sodium (DSS)-induced colitis in mice. Both acute and chronic colitis was induced by feeding female Swiss-Webster mice with 5% DSS (30,000–40,000 mol wt) for five or seven days, respectively. Therapy was advocated prophylactically, prophylaxis plus therapy and therapeutically during the acute and chronic phase of the disease and therapeutically during the chronic phase of the disease. Intracolonic cyclosporin given prophylactically showed adverse effects by increasing the damage to the colonic mucosa. However, intracolonic cyclosporin given therapeutically in 2.5, 5, and 10 mg/kg after the induction of colitis resulted in dramatic responses in terms of reducing the disease activity and histologic scores, corroborated by complete histological resolution compared to oral cyclosporin given at identical doses. Intracolonic cyclosporin (5 mg/kg) was also very effective in reducing the chronic inflammation. The results of this study highlight the application of this animal model for therapeutic research. Furthermore, cyclosporin administered as an enema provides a new stratagem for the therapy of IBD because of its rapid onset of action at very low doses without the risk inherent in oral or systemic administration.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime

Price includes VAT (Russian Federation)

Instant access to the full article PDF.

Rent this article via DeepDyve

Institutional subscriptions

Similar content being viewed by others

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Uridine Ameliorates Dextran Sulfate Sodium (DSS)-Induced Colitis in Mice

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Dextran Sulfate Sodium (DSS)-Induced Acute Colitis in the Rat

clinicopathologic study of dextran sulfate sodium experimental murine colitis

Restoration of mucosal integrity and epithelial transport function by concomitant anti-TNFα treatment in chronic DSS-induced colitis

Riddell RH: Pathology of idiopathic inflammatory bowel diseases. In Inflammatory Bowel Diseases. JB Kirsner RG Shorter (ed). Philadelphia, Lea and Febiger, 1988, pp 329–350

Google Scholar  

Lauritsen K: Drug treatment and formation of eicosanoids in patients with chronic inflammatory bowel diseases. Danish Med Bull 36:378–393, 1989

Sutherland L, Singelton F, Sessions F, Hanauer S, Karwitt E, Ranking G, Summers R, Mekhjian H, Greenberger N, Kelly M, Levine L, Thomson A, Alpert E: Double blind placebo controlled trial of metronidazole in Crohn's disease. Gut 18:1071–1075, 1991

Present DH: 6-Mercaptopurine and other immunosuppressive agents in the treatment of Crohn's disease and ulcerative colitis. Gastroenterol Clin North Am 18:57–71, 1989

Stange EF, Fleig WE, Rekhlaw E, Ditschuneit H: Cyclosporin A treatment in inflammatory bowel diseases. Dig Dis Sci 34:1387–1392, 1989

Lichteger S, Present DH: Preliminary report: Cyclosporin in treatment of severe active ulcerative colitis. Lancet 336(8706):16–19, 1990

Porro GB, Panza E, Petrillo M: Cyclosporin A in acute ulcerative colitis. Lancet 2:1277–1278, 1984

Lobo AJ, Juby LD, Foster RN, Rothwell J, Smith AH, Axon ATR: Oral cyclosporin and renal function in Crohn's disease. Gut 30:A1480, 1989

Watkins PB: The role of cytochrome P-450 IIIA in cyclosporin metabolism. J Am Acad Dermatol 23:1309–1311, 1990

Sandborn WJ, Strong RM, Forland SC, Chase RL, Cutter RE: The pharmacokinetics and colonic tissue concentrations after IV, oral and enema administration. J Clin Pharmacol 31:76–80, 1991

Brynskov J, Freund L, Thomsen OO, Anderson CB, Rasmussen SN, Binder V: Treatment of refractory ulcerative colitis with cyclosporin enemas. Lancet 1:721–722, 1989

Sandborn WJ, Tremaine WJ, Schroeder KW, Batts KP, Lawson GM: Cyclosporine (CYA) enemas for treatment-resistant, ulcerative proctosignoiditis (UPS). Gastroenterology 102:A690, 1992

Winter T, Dalton HR, Merrett MN, Campbell A, Jewell DP: Cyclosporin A retention enemas in refractory distal ulcerative colitis: An open trial. Gastroenterology 102:A947, 1992

Ranzi T, Campanini MC, Velio P, Quarto di Palo F, Bianchi P: Treatment of chronic proctosigmoiditis with cyclosporin enemas. Lancet 2:97, 1989

Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R: Novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 98:694–702, 1990

Cooper HS, Murthy SNS, Shah RS, Sedergran DJ: Clinicopathological study of dextran sulfate sodium experimental murine colitis. Lab Invest (submitted)

Dunnet CW: Multiple comparison tests. Biometrica 26:139–141, 1970

Newman D: The distribution of range in sample from a normal population expressed in terms of independent estimate of standard deviation. Biometrica 31:20–30, 1939

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 96:795–803, 1989

Fitzpatric LR, Bostwick JS, Renzetti M, Pendleton RG, Dektor DL: Antiinflammatory effects of various drugs on acetic acid induced colitis in the rat. Agents Actions 30:399–411, 1990

Azad Khan AK, Piris J, Truelove SC: An experiment to determine the actual therapeutic moiety of sulphasalazine. Lancet 2:892–895, 1977

Meyers S, Sachar DB, Present DH, Janowitz HD: Olsalazine sodium in the treatment of ulcerative colitis among patients intolerant of sulfasalazine. A prospective, randomized placebo-controlled, double blind, dose ranging clinical trial. Gastroenterology 93:1255–1262, 1987

Robinson MG: New oral salicylates in the therapy of chronic idiopathic inflammatory bowel disease. Gastroenterol Clin North Am 18:43–49, 1989

Sutherland LR, Martin F, Greer S, Robinson MG, Greenberger N, Sarbid F, Martin T, Sparr J, Prokipchuk ED, Borgen L: 5-Aminosalicylic acid enemas in the treatment of distal ulcerative colitis, proctosigmoiditis and proctitis. Gastroenterology 92:1844–1848, 1987

Kolars JC, Duell EA, Schmiedlin-Ren P, Ellis CN, Voorhees JJ, Watkins PB: Cyclosporin metabolism by cytochrome P-450 IIIA in rat enterocytes: Another determinant of oral bioavailability. Gastroenterology 98:A192, 1990

Kronke M, Leonard WJ, Deeper JM, Green WC: Sequential expression of genes involved in human lymphocyte growth and differentiation. J Exp Med 161:1593–1598, 1985

Wagner H: Cyclosporin A: Mechanism action. Transplant Proc 15:523–526, 1983

Kubes P, Hunter J, Granger DN: Effects of cyclosporin A and FK 506 on ischemia/reperfusion-induced neutrophil infiltration in the cat. Dig Dis Sci 36:1469–1472, 1991

Thomson AW, Moon DK, Geczy CL, Nelson DS: Modification of delayed-hypersensitivity reactions to ovalbumin in cyclosporin A-treated guinea pigs. Immunology 48:301–308, 1983

Download references

Author information

Authors and affiliations.

Krancer Center for Inflammatory Bowel Disease Research, Division of Gastroenterology, Department of Medicine, Hahnemann University, Broad and Vine Streets, Mail Stop 131, 19102-1192, Philadelphia, Pennsylvania

S. N. S. Murthy PhD, Harry S. Cooper MD, Helen Shim MD, Rupal S. Shah BS, Samih A. Ibrahim MS & Deborah J. Sedergran MT (ASCP)

Department of Pathology and Laboratory Medicine, Hahnemann University, Broad and Vine Streets, 19102-1192, Philadelphia, Pennsylvania

You can also search for this author in PubMed   Google Scholar

Rights and permissions

Reprints and permissions

About this article

Murthy, S.N.S., Cooper, H.S., Shim, H. et al. Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin. Digest Dis Sci 38 , 1722–1734 (1993). https://doi.org/10.1007/BF01303184

Download citation

Received : 02 March 1992

Revised : 14 October 1992

Accepted : 20 October 1992

Issue Date : September 1993

DOI : https://doi.org/10.1007/BF01303184

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • cyclosporin enema
  • oral cyclosporin
  • ulcerative colitis in mice
  • cyclosporin therapy
  • inflammation
  • Find a journal
  • Publish with us
  • Track your research

OpenLB Open Library of Bioscience

Clinicopathologic study of dextran sulfate sodium experimental murine colitis..

  • H S Cooper: Department of Pathology and Laboratory Medicine, Hahnemann University, Philadelphia, Pennsylvania.

BACKGROUND: We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and to discover the earliest histopathologic changes that lead to colitis. EXPERIMENTAL DESIGN: Acute colitis was induced in Swiss-Webster mice by 7 days of oral DSS with animals sacrificed daily. Chronic colitis was induced by: (a) 7 days of oral DSS followed by 7 days of H2O (for 1, 2, and 3 cycles) and (b) 7 days of oral DSS followed by 14 and 21 days of H2O. In each experimental group, the entire colons were examined histologically and correlated with clinical symptoms. RESULTS: Acute clinical symptoms (diarrhea and/or grossly bloody stool) were associated with the presence of erosions and inflammation. More importantly, the earliest histologic changes which predated clinical colitis was loss of the basal one-third of the crypt (day 3), which progressed with time to loss of the entire crypt resulting in erosions on day 5. The earliest changes were very focal and not associated with inflammation. Inflammation was a secondary phenomena and only became significant after erosions appeared. Animals treated with only 7 days of DSS followed by 14 and 21 days of H2O developed a chronic colitis with the following histologic features: areas of activity (erosions and inflammation), inactivity, crypt distortion, florid epithelial proliferation and possible dysplasia. These changes were similar to animals given 3 cycles of DSS. The clinical disease activity index correlated significantly with pathologic changes in both the acute and chronic phases of the disease. CONCLUSIONS: The mechanism of DSS colitis is presently unknown. However, the finding of crypt loss without proceeding or accompanying inflammation suggests that the initial insult is at the level of the epithelial cell with inflammation being a secondary phenomena. This may be a good model to study how early mucosal changes lead to inflammation and the biology of the colonic enterocyte. Chronic colitis induced after only 7 days of DSS may serve as a useful model to study the effects of pharmacologic agents in human inflammatory disease and mechanisms of perpetuation of inflammation. Finally, we believe that this model has the potential to study the dysplasia cancer sequence in inflammatory disease.

Similar Articles

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Unveiling Colitis: A Journey through the Dextran Sodium Sulfate-induced Model

Affiliations.

  • 1 Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA, 30303, USA.
  • 2 Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA.
  • PMID: 38280217
  • PMCID: PMC11063560 (available on 2025-01-27 )
  • DOI: 10.1093/ibd/izad312

Animal models of inflammatory bowel disease (IBD) are valuable tools for investigating the factors involved in IBD pathogenesis and evaluating new therapeutic options. The dextran sodium sulfate (DSS)-induced model of colitis is arguably the most widely used animal model for studying the pathogenesis of and potential treatments for ulcerative colitis (UC), which is a primary form of IBD. This model offers several advantages as a research tool: it is highly reproducible, relatively easy to generate and maintain, and mimics many critical features of human IBD. Recently, it has also been used to study the role of gut microbiota in the development and progression of IBD and to investigate the effects of other factors, such as diet and genetics, on colitis severity. However, although DSS-induced colitis is the most popular and flexible model for preclinical IBD research, it is not an exact replica of human colitis, and some results obtained from this model cannot be directly applied to humans. This review aims to comprehensively discuss different factors that may be involved in the pathogenesis of DSS-induced colitis and the issues that should be considered when using this model for translational purposes.

Keywords: chemically induced colitis model; pathogenesis mechanism of action; preclinical research; sulfated glucan; ulcerative colitis.

Plain language summary

This review discusses different factors that may be involved in the pathogenesis of DSS-induced colitis and the issues that should be considered when using this model for translational purposes.

Published by Oxford University Press on behalf of Crohn’s & Colitis Foundation 2024.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Similar articles

  • Dextran sodium sulfate colitis murine model: An indispensable tool for advancing our understanding of inflammatory bowel diseases pathogenesis. Eichele DD, Kharbanda KK. Eichele DD, et al. World J Gastroenterol. 2017 Sep 7;23(33):6016-6029. doi: 10.3748/wjg.v23.i33.6016. World J Gastroenterol. 2017. PMID: 28970718 Free PMC article. Review.
  • 2,3,5,4'-Tetrahydroxystilbene-2-O-β-D-glucoside, a major bioactive component from Polygoni multiflori Radix (Heshouwu) suppresses DSS induced acute colitis in BALb/c mice by modulating gut microbiota. He X, Liu J, Long G, Xia XH, Liu M. He X, et al. Biomed Pharmacother. 2021 May;137:111420. doi: 10.1016/j.biopha.2021.111420. Epub 2021 Feb 23. Biomed Pharmacother. 2021. PMID: 33761623
  • Gut microbiota regulation and anti-inflammatory effect of β-carotene in dextran sulfate sodium-stimulated ulcerative colitis in rats. Zhu L, Song Y, Liu H, Wu M, Gong H, Lan H, Zheng X. Zhu L, et al. J Food Sci. 2021 May;86(5):2118-2130. doi: 10.1111/1750-3841.15684. Epub 2021 Apr 21. J Food Sci. 2021. PMID: 33884622
  • Spinal anesthesia alleviates dextran sodium sulfate-induced colitis by modulating the gut microbiota. Hong Y, Zhao J, Chen YR, Huang ZH, Hou LD, Shen B, Xin Y. Hong Y, et al. World J Gastroenterol. 2022 Mar 28;28(12):1239-1256. doi: 10.3748/wjg.v28.i12.1239. World J Gastroenterol. 2022. PMID: 35431512 Free PMC article.
  • Dextran sodium sulphate colitis mouse model: traps and tricks. Perše M, Cerar A. Perše M, et al. J Biomed Biotechnol. 2012;2012:718617. doi: 10.1155/2012/718617. Epub 2012 May 14. J Biomed Biotechnol. 2012. PMID: 22665990 Free PMC article. Review.
  • Exploring the relationship between ulcerative colitis, colorectal cancer, and prostate cancer. Kura Y, De Velasco MA, Sakai K, Uemura H, Fujita K, Nishio K. Kura Y, et al. Hum Cell. 2024 Aug 20. doi: 10.1007/s13577-024-01118-2. Online ahead of print. Hum Cell. 2024. PMID: 39162974
  • The Role of TRP Channels in Sepsis and Colitis. Dvornikova KA, Platonova ON, Bystrova EY. Dvornikova KA, et al. Int J Mol Sci. 2024 Apr 27;25(9):4784. doi: 10.3390/ijms25094784. Int J Mol Sci. 2024. PMID: 38731999 Free PMC article. Review.
  • Serum Galectin-3 as a Non-Invasive Marker for Primary Sclerosing Cholangitis. Bajraktari G, Elger T, Huss M, Loibl J, Albert A, Kandulski A, Müller M, Tews HC, Buechler C. Bajraktari G, et al. Int J Mol Sci. 2024 Apr 27;25(9):4765. doi: 10.3390/ijms25094765. Int J Mol Sci. 2024. PMID: 38731984 Free PMC article.
  • Zhao M, Gönczi L, Lakatos PL, Burisch J.. The burden of inflammatory bowel disease in Europe in 2020. J Crohns Colitis. 2021;15(9):1573-1587. - PubMed
  • Kappelman MD, Moore KR, Allen JK, Cook SF.. Recent trends in the prevalence of Crohn’s disease and ulcerative colitis in a commercially insured US population. Dig Dis Sci. 2013;58(2):519-525. - PMC - PubMed
  • Ng SC. Emerging trends of inflammatory bowel disease in Asia. Gastroenterol Hepatol. 2016;12(3):193-196. - PMC - PubMed
  • Danese S, Schabel E, Ainsworth MA, Peyrin-Biroulet L.. Challenges and opportunities for IBD drug development: from early stage to regulatory approval. Gut. 2020;69(7):1157-1161. - PubMed
  • Sousa P, Noor NM. Revisiting drug development for older molecules in IBD. Inflamm Bowel Dis. 2022;29(6):1012-1013. - PubMed

Publication types

  • Search in MeSH

Related information

Grants and funding.

  • IK6 BX004476/BX/BLRD VA/United States
  • R01 DK116306/DK/NIDDK NIH HHS/United States
  • RO1-DK-116306/DK/NIDDK NIH HHS/United States
  • BX002526/VA/VA/United States

LinkOut - more resources

Full text sources.

  • Ovid Technologies, Inc.
  • Silverchair Information Systems

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

COMMENTS

  1. Clinicopathologic study of dextran sulfate sodium experimental murine

    The hypothesis that appendicular lymphoid tissue, but not the spleen, contributes to the development of colitis is supported by experimentally assessing the importance of secondary lymphoid tissues in inducing colitis in mice. Dysplasia and cancer in the dextran sulfate sodium mouse colitis model.

  2. Clinicopathologic study of dextran sulfate sodium experimental murine

    Background: We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and to discover the earliest histopathologic changes that lead to colitis. Experimental design: Acute colitis was induced in Swiss-Webster mice by 7 days of oral DSS ...

  3. (PDF) Clinicopathologic study of dextran sulfate sodium experimental

    Clinicopathologic study of dextran sulfate sodium experimental acute colitis January 1998 The Japanese Journal of Pharmacology 76:297 DOI: 10.1016/S0021-5198 (19)41298-5 License CC BY-NC-ND 4.0 ...

  4. An experimental model of colitis induced by dextran sulfate sodium from

    Background: Murine models are essential tools to understand IBD pathogenesis. Among different types of chemically induced colitis models, the dextran sulfate sodium (DSS)-induced colitis model is the most common model of IBD, due to its simplicity.

  5. Dextran sodium sulfate colitis murine model: An indispensable tool for

    The most widely used experimental model employs dextran sodium sulfate (DSS) to induce epithelial damage. The DSS colitis model in IBD research has advantages over other various chemically induced experimental models due to its rapidity, simplicity, reproducibility and controllability.

  6. Clinicopathologic study of dextran sulfate sodium experimental murine

    Clinicopathologic study of dextran sulfate sodium experimental murine colitis. H S Cooper, S N Murthy, R S Shah, D J Sedergran Laboratory Investigation; a Journal of Technical Methods and Pathology 1993 August BACKGROUND: We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and ...

  7. PAM3 protects against DSS-induced colitis by altering the M2 ...

    Inflammation of the gastrointestinal tract contributes to the development of inflammatory bowel disease (IBD). Human IBD is modeled by administering dextran sulfate sodium (DSS) to mice. In humans ...

  8. Clinicopathologic study of dextran sulfate sodium experimental murine

    A laboratory investigation article from 1993 that compares the clinical and pathologic features of acute and chronic colitis induced by dextran sulfate sodium in mice. The article provides keywords, abstract, authors, source, publication details and classification information.

  9. Treatment of dextran sulfate sodium-induced murine colitis by ...

    In this study, the effect of intracolonically administered cyclosporin was tested for its efficacy to heal mucosal erosions in dextran sulfate sodium (DSS)-induced colitis in mice. Both acute and chronic colitis was induced by feeding female Swiss-Webster mice with 5% DSS (30,000-40,000 mol wt) for five or seven days, respectively.

  10. Clinicopathologic study of dextran sulfate sodium experimental murine

    A study of the clinical and histopathologic features of acute and chronic colitis induced by oral DSS in mice. The earliest histologic change was crypt loss without inflammation, suggesting a primary epithelial injury and a secondary inflammatory response.

  11. Differential susceptibility of inbred mouse strains to dextran sulfate

    Abstract Dextran sulfate sodium (DSS)-induced murine colitis represents an experimental model for human inflammatory bowel disease. The aim of this study was to screen various inbred strains of mice for genetically determined differences in susceptibility to DSS-induced colitis.

  12. Cryptotanshinone ameliorates dextran sulfate sodium-induced murine

    Experimental study showed that Danshen had protective effect on acute colitis induced by dextran sulfate sodium (DSS) [11]. Cryptotanshinone (CTS) is one of the main active components of Danshen, with anti-inflammation, anti-oxidation, anticancer and other pharmacological activities [12], [13], [14], [15].

  13. The dextran sulphate sodium (DSS) model of colitis: an overview

    The dextran sulphate sodium model of colitis has demonstrated several correlations with human inflammatory bowel disease and is deemed suitable for investigating pathogenesis, therapeutic options and the dysplasia-adenocarcinoma sequence of inflammatory bowel disease. It is widely applicable to mice, rats, hamsters and guinea pigs.

  14. Europe PMC

    Background We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and to discover the earliest histopathologic changes that lead to colitis.

  15. HSPA9 reduction exacerbates symptoms and cell death in DSS ...

    In a dextran sodium sulfate-induced colitis mouse model, the downregulated HSPA9 exacerbates colitis symptoms, including increased immune cell infiltration, elevated proinflammatory cytokines ...

  16. Dextran sulfate sodium-induced colitis-associated neoplasia: a ...

    The mouse model of dextran sulfate sodium (DSS) -induced colitis represents an excellent preclinical system in which to both characterize the molecular events required for tumor formation in the ...

  17. Europe PMC

    About Europe PMC Preprints in Europe PMC Funders Become a funder Governance Roadmap Outreach Tools overview Article status monitor ORCID article claiming Journal list Grant finder External links service RSS feeds Annotations Annotations submission service Developer resources Articles RESTful API Grants RESTful API API case studies SOAP web ...

  18. Dextran Sodium Sulphate Colitis Mouse Model: Traps and Tricks

    Both dextran sulphate sodium (DSS) and trinitrobenzene sulfonic acid (TNBS-) induced colitis are well-established animal models of mucosal inflammation that have been used for over 2 decades in the study of IBD pathogenesis and preclinical studies [ 6 - 8 ].

  19. Treatment of dextran sulfate sodium-induced murine colitis by

    In this study, the effect of intracolonically administered cyclosporin was tested for its efficacy to heal mucosal erosions in dextran sulfate sodium (DSS)-induced colitis in mice. Both acute and chronic colitis was induced by feeding female Swiss-Webster mice with 5% DSS (30,000-40,000 mol wt) for five or seven days, respectively.

  20. Clinicopathologic study of dextran sulfate sodium experimental murine

    BACKGROUND: We undertook this study in order to fully characterize the clinical and histopathology features of the dextran sulfate sodium (DSS) model of experimental murine colitis and to discover the earliest histopathologic changes that lead to colitis. EXPERIMENTAL DESIGN: Acute colitis was induced in Swiss-Webster mice by 7 days of oral DSS ...

  21. PDF Clinicopathologic study of dextran sulfate sodium experimental acute

    KW-5617 nay be a useful drug for the treatment of diarrhea in terns of less side effect concerning constipation. P-659 Clinicopathologic study of dextran sulfate sodium experimental acute colitis.

  22. Unveiling Colitis: A Journey through the Dextran Sodium Sulfate-induced

    The dextran sodium sulfate (DSS)-induced model of colitis is arguably the most widely used animal model for studying the pathogenesis of and potential treatments for ulcerative colitis (UC), which is a primary form of IBD. This model offers several advantages as a research tool: it is highly reproducible, relatively easy to generate and ...