J Pathol Transl Med.  2023 Jan;57(1):60-66. 10.4132/jptm.2022.11.02.

Inflammatory bowel disease–associated intestinal fibrosis

Affiliations
  • 1Department of Pathology, Keimyung University School of Medicine, Daegu, Korea
  • 2Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
  • 3Division of Colorectal Surgery, Department of Surgery, Keimyung University School of Medicine, Daegu, Korea

Abstract

Fibrosis is characterized by a proliferation of fibroblasts and excessive extracellular matrix following chronic inflammation, and this replacement of organ tissue with fibrotic tissue causes a loss of function. Inflammatory bowel disease (IBD) is a chronic inflammation of the gastrointestinal tract, and intestinal fibrosis is common in IBD patients, resulting in several complications that require surgery, such as a stricture or penetration. This review describes the pathogenesis and various factors involved in intestinal fibrosis in IBD, including cytokines, growth factors, epithelial-mesenchymal and endothelial-mesenchymal transitions, and gut microbiota. Furthermore, histopathologic findings and scoring systems used for stenosis in IBD are discussed, and differences in the fibrosis patterns of ulcerative colitis and Crohn’s disease are compared. Biomarkers and therapeutic agents targeting intestinal fibrosis are briefly mentioned at the end.

Keyword

Fibrosis; Inflammatory bowel disease; Crohn disease; Colitis; ulcerative

Figure

  • Fig. 1 Gross finding of resected large intestine specimen of 30-year-old patient with Crohn’s disease. Creeping fat covers more than 50% of the intestinal circumference surface with mesenteric proliferation. Cobblestone mucosa and proper muscle hyperplasia that result in intestinal stricture are also noted.

  • Fig. 2 Histologic findings of Crohn’s disease. (A) Hypertrophy and hyperplasia of the submucosa and muscularis propria are present. (B) Chronic inflammation with lymphoid aggregates and lymphoid follicles is dominant. (C) Although chronic inflammation is predominant, active inflammation that consists of extensive neutrophilic infiltrates is also noted. (D) Fibromuscular hyperplasia of damaged submucosal vessels causes slow blood flow. (E) Neural hypertrophy is also noted. (F) Trichrome stain reveals that dissection between the hyperplastic smooth muscle bundles is interspersed with fibrosis.


Reference

References

1. Leask A, Abraham DJ. TGF-beta signaling and the fibrotic response. FASEB J. 2004; 18:816–27.
2. Kumar V, Abbas AK, Aster JC. Robbins and Cotran pathologic basis of disease. Amsterdam: Elsevier;2020.
3. Jun JI, Lau LF. Resolution of organ fibrosis. J Clin Invest. 2018; 128:97–107.
Article
4. Di Gregorio J, Robuffo I, Spalletta S, et al. The epithelial-to-mesenchymal transition as a possible therapeutic target in fibrotic disorders. Front Cell Dev Biol. 2020; 8:607483.
Article
5. Rockey DC, Bell PD, Hill JA. Fibrosis: a common pathway to organ injury and failure. N Engl J Med. 2015; 372:1138–49.
Article
6. Zhang YZ, Li YY. Inflammatory bowel disease: pathogenesis. World J Gastroenterol. 2014; 20:91–9.
Article
7. Zidar N. Histopathology of fibrosis in Crohn’s disease: the importance of understanding its pathogenesis. Gastroenterology. 2020; 158:2313–4.
Article
8. Hanna MH, Kaiser AM. Update on the management of sigmoid diverticulitis. World J Gastroenterol. 2021; 27:760–81.
Article
9. Speca S, Giusti I, Rieder F, Latella G. Cellular and molecular mechanisms of intestinal fibrosis. World J Gastroenterol. 2012; 18:3635–61.
Article
10. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008; 214:199–210.
Article
11. Burke JP, Mulsow JJ, O’Keane C, Docherty NG, Watson RW, O’Connell PR. Fibrogenesis in Crohn’s disease. Am J Gastroenterol. 2007; 102:439–48.
Article
12. Franze E, Monteleone I, Laudisi F, et al. Cadherin-11 is a regulator of intestinal fibrosis. J Crohns Colitis. 2020; 14:406–17.
Article
13. Rieder F, Fiocchi C, Rogler G. Mechanisms, management, and treatment of fibrosis in patients with inflammatory bowel diseases. Gastroenterology. 2017; 152:340–50.
Article
14. Rieder F, Fiocchi C. Intestinal fibrosis in inflammatory bowel disease: current knowledge and future perspectives. J Crohns Colitis. 2008; 2:279–90.
15. Derkacz A, Olczyk P, Olczyk K, Komosinska-Vassev K. The role of extracellular matrix components in inflammatory bowel diseases. J Clin Med. 2021; 10:1122.
Article
16. Graham DB, Xavier RJ. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature. 2020; 578:527–39.
Article
17. Zhang H, Massey D, Tremelling M, Parkes M. Genetics of inflammatory bowel disease: clues to pathogenesis. Br Med Bull. 2008; 87:17–30.
Article
18. Jerala M, Hauptman N, Kojc N, Zidar N. Expression of fibrosis-related genes in liver and kidney fibrosis in comparison to inflammatory bowel diseases. Cells. 2022; 11:314.
Article
19. Gazouli M, Pachoula I, Panayotou I, et al. NOD2/CARD15, ATG16L1 and IL23R gene polymorphisms and childhood-onset of Crohn’s disease. World J Gastroenterol. 2010; 16:1753–8.
Article
20. Andriulli A, Annese V, Latiano A, et al. The frame-shift mutation of the NOD2/CARD15 gene is significantly increased in ulcerative colitis: an *IG-IBD study. Gastroenterology. 2004; 126:625–7.
Article
21. Wehkamp J, Harder J, Weichenthal M, et al. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut. 2004; 53:1658–64.
Article
22. Nimmo ER, Prendergast JG, Aldhous MC, et al. Genome-wide methylation profiling in Crohn’s disease identifies altered epigenetic regulation of key host defense mechanisms including the Th17 pathway. Inflamm Bowel Dis. 2012; 18:889–99.
Article
23. Chuang AY, Chuang JC, Zhai Z, Wu F, Kwon JH. NOD2 expression is regulated by microRNAs in colonic epithelial HCT116 cells. Inflamm Bowel Dis. 2014; 20:126–35.
Article
24. Rieder F, Lawrance IC, Leite A, Sans M. Predictors of fibrostenotic Crohn’s disease. Inflamm Bowel Dis. 2011; 17:2000–7.
Article
25. Henckaerts L, Van Steen K, Verstreken I, et al. Genetic risk profiling and prediction of disease course in Crohn’s disease patients. Clin Gastroenterol Hepatol. 2009; 7:972–80.
Article
26. Nijhuis A, Biancheri P, Lewis A, et al. In Crohn’s disease fibrosis-reduced expression of the miR-29 family enhances collagen expression in intestinal fibroblasts. Clin Sci (Lond). 2014; 127:341–50.
27. Duerr RH, Taylor KD, Brant SR, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006; 314:1461–3.
28. Glas J, Seiderer J, Wetzke M, et al. rs1004819 is the main disease-associated IL23R variant in German Crohn’s disease patients: combined analysis of IL23R, CARD15, and OCTN1/2 variants. PLoS One. 2007; 2:e819.
29. Cleynen I, Gonzalez JR, Figueroa C, et al. Genetic factors conferring an increased susceptibility to develop Crohn’s disease also influence disease phenotype: results from the IBDchip European Project. Gut. 2013; 62:1556–65.
Article
30. Brand S, Hofbauer K, Dambacher J, et al. Increased expression of the chemokine fractalkine in Crohn’s disease and association of the fractalkine receptor T280M polymorphism with a fibrostenosing disease Phenotype. Am J Gastroenterol. 2006; 101:99–106.
Article
31. Fowler EV, Doecke J, Simms LA, et al. ATG16L1 T300A shows strong associations with disease subgroups in a large Australian IBD population: further support for significant disease heterogeneity. Am J Gastroenterol. 2008; 103:2519–26.
32. Forni D, Cleynen I, Ferrante M, et al. ABO histo-blood group might modulate predisposition to Crohn’s disease and affect disease behavior. J Crohns Colitis. 2014; 8:489–94.
Article
33. Hume GE, Fowler EV, Lincoln D, et al. Angiotensinogen and transforming growth factor beta1: novel genes in the pathogenesis of Crohn’s disease. J Med Genet. 2006; 43:e51.
34. Adams AT, Kennedy NA, Hansen R, et al. Two-stage genome-wide methylation profiling in childhood-onset Crohn’s disease implicates epigenetic alterations at the VMP1/MIR21 and HLA loci. Inflamm Bowel Dis. 2014; 20:1784–93.
35. Meijer MJ, Mieremet-Ooms MA, van Hogezand RA, Lamers CB, Hommes DW, Verspaget HW. Role of matrix metalloproteinase, tissue inhibitor of metalloproteinase and tumor necrosis factor-alpha single nucleotide gene polymorphisms in inflammatory bowel disease. World J Gastroenterol. 2007; 13:2960–6.
Article
36. Alonso A, Domenech E, Julia A, et al. Identification of risk loci for Crohn’s disease phenotypes using a genome-wide association study. Gastroenterology. 2015; 148:794–805.
Article
37. Rieder F, Brenmoehl J, Leeb S, Scholmerich J, Rogler G. Wound healing and fibrosis in intestinal disease. Gut. 2007; 56:130–9.
Article
38. Rockey DC. Hepatic fibrosis, stellate cells, and portal hypertension. Clin Liver Dis. 2006; 10:459–79.
Article
39. Bamba S, Lee CY, Brittan M, et al. Bone marrow transplantation ameliorates pathology in interleukin-10 knockout colitic mice. J Pathol. 2006; 209:265–73.
Article
40. Brittan M, Wright NA. Gastrointestinal stem cells. J Pathol. 2002; 197:492–509.
Article
41. Mao R, Kurada S, Gordon IO, et al. The mesenteric fat and intestinal muscle interface: creeping fat influencing stricture formation in Crohn’s disease. Inflamm Bowel Dis. 2019; 25:421–6.
Article
42. Peyrin-Biroulet L, Gonzalez F, Dubuquoy L, et al. Mesenteric fat as a source of C reactive protein and as a target for bacterial translocation in Crohn’s disease. Gut. 2012; 61:78–85.
Article
43. Crohn BB, Ginzburg L, Oppenheimer GD. Regional ileitis: a pathologic and clinical entity. JAMA. 1932; 99:1323–9.
44. Goncalves P, Magro F, Martel F. Metabolic inflammation in inflammatory bowel disease: crosstalk between adipose tissue and bowel. Inflamm Bowel Dis. 2015; 21:453–67.
45. Desreumaux P, Ernst O, Geboes K, et al. Inflammatory alterations in mesenteric adipose tissue in Crohn’s disease. Gastroenterology. 1999; 117:73–81.
Article
46. Levine JA, Jensen MD, Eberhardt NL, O’Brien T. Adipocyte macrophage colony-stimulating factor is a mediator of adipose tissue growth. J Clin Invest. 1998; 101:1557–64.
Article
47. Chen W, Lu C, Hirota C, Iacucci M, Ghosh S, Gui X. Smooth muscle hyperplasia/hypertrophy is the most prominent histological change in Crohn’s fibrostenosing bowel strictures: a semiquantitative analysis by using a novel histological grading scheme. J Crohns Colitis. 2017; 11:92–104.
Article
48. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003; 112:1776–84.
Article
49. Zhang D, Wang S, Chen J, et al. Fibulin-4 promotes osteosarcoma invasion and metastasis by inducing epithelial to mesenchymal transition via the PI3K/Akt/mTOR pathway. Int J Oncol. 2017; 50:1513–30.
Article
50. Frid MG, Kale VA, Stenmark KR. Mature vascular endothelium can give rise to smooth muscle cells via endothelial-mesenchymal transdifferentiation: in vitro analysis. Circ Res. 2002; 90:1189–96.
51. Bates RC, Mercurio AM. Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Mol Biol Cell. 2003; 14:1790–800.
52. Strutz F, Zeisberg M, Ziyadeh FN, et al. Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int. 2002; 61:1714–28.
Article
53. Yang J, Dai C, Liu Y. A novel mechanism by which hepatocyte growth factor blocks tubular epithelial to mesenchymal transition. J Am Soc Nephrol. 2005; 16:68–78.
Article
54. Kagawa T, Takemura G, Kosai K, et al. Hepatocyte growth factor gene therapy slows down the progression of diabetic nephropathy in db/db mice. Nephron Physiol. 2006; 102:p92–102.
55. D’Alessio S, Ungaro F, Noviello D, Lovisa S, Peyrin-Biroulet L, Danese S. Revisiting fibrosis in inflammatory bowel disease: the gut thickens. Nat Rev Gastroenterol Hepatol. 2022; 19:169–84.
Article
56. Chu H, Khosravi A, Kusumawardhani IP, et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science. 2016; 352:1116–20.
Article
57. Qiu P, Ishimoto T, Fu L, Zhang J, Zhang Z, Liu Y. The gut microbiota in inflammatory bowel disease. Front Cell Infect Microbiol. 2022; 12:733992.
Article
58. Alfredsson J, Wick MJ. Mechanism of fibrosis and stricture formation in Crohn’s disease. Scand J Immunol. 2020; 92:e12990.
Article
59. Zhao Z, Cheng W, Qu W, Shao G, Liu S. Antibiotic alleviates radiation-induced intestinal injury by remodeling microbiota, reducing inflammation, and inhibiting fibrosis. ACS Omega. 2020; 5:2967–77.
Article
60. Zhao S, Dejanovic D, Yao P, et al. Selective deletion of MyD88 signaling in alpha-SMA positive cells ameliorates experimental intestinal fibrosis via post-transcriptional regulation. Mucosal Immunol. 2020; 13:665–78.
61. Porras AM, Zhou H, Shi Q, et al. Inflammatory bowel disease-associated gut commensals degrade components of the extracellular matrix. Preprint at: https://doi.org/10.1101/2022.08.09.503432 . 2022.
Article
62. Knowles CH, Farrugia G. Gastrointestinal neuromuscular pathology in chronic constipation. Best Pract Res Clin Gastroenterol. 2011; 25:43–57.
Article
63. Mosli MH, Parker CE, Nelson SA, et al. Histologic scoring indices for evaluation of disease activity in ulcerative colitis. Cochrane Database Syst Rev. 2017; 5:CD011256.
Article
64. Hutchings H, Alrubaiy L. Crohn’s disease activity index. Michalos AC, editor. Encyclopedia of quality of life and well-being research. Dordrecht Springer;Netherlands: 2014. p. 1354–7.
Article
65. Sofo L, Caprino P, Schena CA, Sacchetti F, Potenza AE, Ciociola A. New perspectives in the prediction of postoperative complications for high-risk ulcerative colitis patients: machine learning preliminary approach. Eur Rev Med Pharmacol Sci. 2020; 24:12781–7.
66. Udristoiu AL, Stefanescu D, Gruionu G, et al. Deep learning algorithm for the confirmation of mucosal healing in Crohn’s disease, based on confocal laser endomicroscopy images. J Gastrointestin Liver Dis. 2021; 30:59–65.
Article
67. Flynn S, Eisenstein S. Inflammatory bowel disease presentation and diagnosis. Surg Clin North Am. 2019; 99:1051–62.
Article
68. Gumaste V, Sachar DB, Greenstein AJ. Benign and malignant colorectal strictures in ulcerative colitis. Gut. 1992; 33:938–41.
Article
69. Mitomi H, Okayasu I, Bronner MP, et al. Comparative histologic assessment of proctocolectomy specimens from Japanese and American patients with ulcerative colitis with or without dysplasia. Int J Surg Pathol. 2005; 13:259–65.
Article
70. Yamagata M, Mikami T, Tsuruta T, et al. Submucosal fibrosis and basic-fibroblast growth factor-positive neutrophils correlate with colonic stenosis in cases of ulcerative colitis. Digestion. 2011; 84:12–21.
Article
71. Bettenworth D, Bokemeyer A, Baker M, et al. Assessment of Crohn’s disease-associated small bowel strictures and fibrosis on cross-sectional imaging: a systematic review. Gut. 2019; 68:1115–26.
Article
72. Agrawal M, Spencer EA, Colombel JF, Ungaro RC. Approach to the management of recently diagnosed inflammatory bowel disease patients: a user’s guide for adult and pediatric gastroenterologists. Gastroenterology. 2021; 161:47–65.
Article
73. Li C, Yang CW, Park JH, et al. Pravastatin treatment attenuates interstitial inflammation and fibrosis in a rat model of chronic cyclosporine-induced nephropathy. Am J Physiol Renal Physiol. 2004; 286:F46–57.
Article
74. Abe Y, Murano M, Murano N, et al. Simvastatin attenuates intestinal fibrosis independent of the anti-inflammatory effect by promoting fibroblast/myofibroblast apoptosis in the regeneration/healing process from TNBS-induced colitis. Dig Dis Sci. 2012; 57:335–44.
Article
75. Gordon JR, Zhang X, Li F, Nayyar A, Town J, Zhao X. Amelioration of pathology by ELR-CXC chemokine antagonism in a swine model of airway endotoxin exposure. J Agromedicine. 2009; 14:235–41.
Article
76. Stillie R, Farooq SM, Gordon JR, Stadnyk AW. The functional significance behind expressing two IL-8 receptor types on PMN. J Leukoc Biol. 2009; 86:529–43.
Article
77. Speca S, Rousseaux C, Dubuquoy C, et al. Novel PPARgamma modulator GED-0507-34 levo ameliorates inflammation-driven intestinal fibrosis. Inflamm Bowel Dis. 2016; 22:279–92.
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr