Intest Res.  2023 Jan;21(1):148-160. 10.5217/ir.2021.00168.

Compositional changes in fecal microbiota associated with clinical phenotypes and prognosis in Korean patients with inflammatory bowel disease

Affiliations
  • 1Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
  • 2Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
  • 3Department of Internal Medicine, Graduate Program in System Health Science and Engineering, Ewha Womans University College of Medicine, Seoul, Korea
  • 4Department of Preventive Medicine, Graduate Program in System Health Science and Engineering, Ewha Womans University College of Medicine, Seoul, Korea
  • 5South Texas Center of Emerging Infectious Diseases (STCEID) and Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
  • 6Division of Gastroenterology, Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea

Abstract

Background/Aims
The fecal microbiota of Korean patients with inflammatory bowel disease (IBD) was investigated with respect to disease phenotypes and taxonomic biomarkers for diagnosis and prognosis of IBD.
Methods
Fecal samples from 70 ulcerative colitis (UC) patients, 39 Crohn’s disease (CD) patients, and 100 healthy control individuals (HC) were collected. The fecal samples were amplified via polymerase chain reaction and sequenced using Illumina MiSeq. The relationships between fecal bacteria and clinical phenotypes were analyzed using the EzBioCloud database and 16S microbiome pipeline.
Results
The alpha-diversity of fecal bacteria was significantly lower in UC and CD (P<0.05) compared to that in HC. Bacterial community compositions in UC and CD were significantly different from that of HC according to Bray-Curtis dissimilarities, and there was also a difference between community composition in UC and CD (P=0.01). In UC, alpha-diversity was further decreased when the disease was more severe and the extent of disease was greater, and community composition significantly differed depending on the extent of the disease. We identified 9 biomarkers of severity and 6 biomarkers of the extent of UC. We also identified 5 biomarkers of active disease and 3 biomarkers of ileocolonic involvement in CD. Lachnospiraceae and Ruminococcus gnavus were biomarkers for better prognosis in CD.
Conclusions
The fecal microbiota profiles of IBD patients were different from those of HC, and several bacterial taxa may be used as biomarkers to determine disease phenotypes and prognosis. These data may also help discover new therapeutic targets for IBD.

Keyword

Inflammatory bowel disease; Microbiota; Biomarkers; Phenotype; Prognosis

Figure

  • Fig. 1. Diversity of fecal microbiota in a HC, UC patients, and CD patients. (A) Chao 1 index, (B) Shannon index, and (C) beta diversity based on Bray-Curtis dissimilarities. Both richness and diversity were significantly lower in inflammatory bowel disease patients compared with HC, and Shannon diversity index was significantly lower in CD compared with UC. Post hoc analyses using pair-wise comparisons showed that 3 groups were significantly different from each other (HC vs. UC, P=0.001; HC vs. CD, P=0.001; UC vs. CD, P=0.001). HC, healthy control individuals; UC, ulcerative colitis; CD, Crohn’s disease.

  • Fig. 2. Diversity of fecal microbiota in ulcerative colitis (UC) patients according to disease severity (A) and disease extent (B). Alpha diversity decreased as the disease severity and extent deteriorated.

  • Fig. 3. Composition of fecal microbiota in HC, UC patients, and CD patients at the phylum (A), genus (B), and species (C) level. Compared to HC and UC patients, CD patients had significantly higher abundances of the phylum Proteobacteria, the genus Escherichia, and the species Escherichia coli. HC, healthy control individuals; UC, ulcerative colitis; CD, Crohn’s disease.

  • Fig. 4. Taxa list according to linear discriminate analysis values determined from comparisons between HC and UC patients (A), and between HC and CD patients (B). Effect size estimation analysis identified 9 bacterial taxa that were significantly more abundant in UC patients than HC (A), and 14 bacterial taxa that were significantly more abundant in CD patients than HC. HC, healthy control individuals; UC, ulcerative colitis; CD, Crohn’s disease.

  • Fig. 5. Taxa list according to linear discriminate analysis values determined from comparisons according to disease severity and extent in ulcerative colitis (UC) patients. Comparisons between moderate to severe and mild UC (A), between moderate to severe UC and remission (B), and between left sided or extensive UC and proctitis (C). Some bacterial taxa (red color) were identified as potential biomarkers for moderate to severe UC compared to mild UC (A), and remission status (B). Several bacterial taxa (red color) were found to be associated with left sided or extensive UC (C).

  • Fig. 6. Taxa list according to linear discriminate analysis values determined from comparisons according to disease severity and extent in Crohn’s disease (CD) patients. Comparisons between remission and active CD (A) and between ileocolonic and small bowel CD (B). Some bacterial taxa (red color) were identified as possible biomarkers for active disease status, compared to remission status (A). Several bacterial taxa (red color) were identified as possible biomarkers for ileocolonic involvement, compared to small bowel involvement only (B).

  • Fig. 7. Relative abundance of particular taxa in Crohn’s disease (CD) patients based on prognosis. (A) Lachnospiraceae and (B) Ruminococcus gnavus. Better prognosis is defined as patients who did not consume biologic agents including anti-tumor necrosis factor (TNF)-α agents or surgical treatment after fecal sampling and worse prognosis is defined as patients who either were administered biologic agents including anti-TNF-α agents or required surgical treatment after fecal sampling (n=9). Lachnospiraceae and R. gnavus were significantly more abundant in the better prognosis group compared to the worse prognosis group.


Reference

1. Danese S, Fiocchi C. Ulcerative colitis. N Engl J Med. 2011; 365:1713–1725.
Article
2. Ungaro R, Mehandru S, Allen PB, Peyrin-Biroulet L, Colombel JF. Ulcerative colitis. Lancet. 2017; 389:1756–1770.
Article
3. Mizoguchi E, Low D, Ezaki Y, Okada T. Recent updates on the basic mechanisms and pathogenesis of inflammatory bowel diseases in experimental animal models. Intest Res. 2020; 18:151–167.
Article
4. Ooi CJ, Hilmi I, Banerjee R, et al. Best practices on immunomodulators and biologic agents for ulcerative colitis and Crohn’s disease in Asia. Intest Res. 2019; 17:285–310.
Article
5. Watanabe K. Clinical management for small bowel of Crohn’s disease in the treat-to-target era: now is the time to optimize treatment based on the dominant lesion. Intest Res. 2020; 18:347–354.
Article
6. Toedter GP, Blank M, Lang Y, Chen D, Sandborn WJ, de Villiers WJ. Relationship of C-reactive protein with clinical response after therapy with ustekinumab in Crohn’s disease. Am J Gastroenterol. 2009; 104:2768–2773.
Article
7. Sands BE. Biomarkers of inflammation in inflammatory bowel disease. Gastroenterology. 2015; 149:1275–1285.
Article
8. Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology. 2009; 136:65–80.
Article
9. Pittayanon R, Lau JT, Leontiadis GI, et al. Differences in gut microbiota in patients with vs without inflammatory bowel diseases: a systematic review. Gastroenterology. 2020; 158:930–946.
Article
10. Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006; 55:205–211.
Article
11. Zuo T, Ng SC. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front Microbiol. 2018; 9:2247.
Article
12. Yilmaz B, Juillerat P, Øyås O, et al. Microbial network disturbances in relapsing refractory Crohn’s disease. Nat Med. 2019; 25:323–336.
Article
13. Franzosa EA, Sirota-Madi A, Avila-Pacheco J, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat Microbiol. 2019; 4:293–305.
Article
14. Neumann C, Blume J, Roy U, et al. c-Maf-dependent Treg cell control of intestinal TH17 cells and IgA establishes host-microbiota homeostasis. Nat Immunol. 2019; 20:471–481.
Article
15. Mahajan R, Midha V, Singh A, et al. Incidental benefits after fecal microbiota transplant for ulcerative colitis. Intest Res. 2020; 18:337–340.
Article
16. Sehgal K, Khanna S. Gut microbiome and checkpoint inhibitor colitis. Intest Res. 2021; 19:360–364.
Article
17. Bamba S, Inatomi O, Nishida A, et al. Relationship between the gut microbiota and bile acid composition in the ileal mucosa of Crohn’s disease. Intest Res. 2022; 20:370–380.
Article
18. Schroeder KW, Tremaine WJ, Ilstrup DM. Coated oral 5-aminosalicylic acid therapy for mildly to moderately active ulcerative colitis: a randomized study. N Engl J Med. 1987; 317:1625–1629.
Article
19. Best WR, Becktel JM, Singleton JW, Kern F Jr. Development of a Crohn’s disease activity index. National Cooperative Crohn’s Disease Study. Gastroenterology. 1976; 70:439–444.
20. Frank DN, Robertson CE, Hamm CM, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm Bowel Dis. 2011; 17:179–184.
Article
21. Papa E, Docktor M, Smillie C, et al. Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PLoS One. 2012; 7:e39242.
Article
22. Satokari R. Contentious host-microbiota relationship in inflammatory bowel disease: can foes become friends again? Scand J Gastroenterol. 2015; 50:34–42.
Article
23. Vester-Andersen MK, Mirsepasi-Lauridsen HC, Prosberg MV, et al. Increased abundance of proteobacteria in aggressive Crohn’s disease seven years after diagnosis. Sci Rep. 2019; 9:13473.
Article
24. Alam MT, Amos G, Murphy A, Murch S, Wellington E, Arasaradnam RP. Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels. Gut Pathog. 2020; 12:1.
Article
25. Gevers D, Kugathasan S, Denson LA, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014; 15:382–392.
Article
26. Darfeuille-Michaud A, Boudeau J, Bulois P, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004; 127:412–421.
Article
27. Lupp C, Robertson ML, Wickham ME, et al. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe. 2007; 2:119–129.
Article
28. Kolho KL, Korpela K, Jaakkola T, et al. Fecal microbiota in pediatric inflammatory bowel disease and its relation to inflammation. Am J Gastroenterol. 2015; 110:921–930.
Article
29. Pilarczyk-Zurek M, Chmielarczyk A, Gosiewski T, et al. Possible role of Escherichia coli in propagation and perpetuation of chronic inflammation in ulcerative colitis. BMC Gastroenterol. 2013; 13:61.
Article
30. Mylonaki M, Rayment NB, Rampton DS, Hudspith BN, Brostoff J. Molecular characterization of rectal mucosa-associated bacterial flora in inflammatory bowel disease. Inflamm Bowel Dis. 2005; 11:481–487.
Article
31. Zhang SL, Wang SN, Miao CY. Influence of microbiota on intestinal immune system in ulcerative colitis and its intervention. Front Immunol. 2017; 8:1674.
Article
32. Olbjørn C, Cvancarova Småstuen M, Thiis-Evensen E, et al. Fecal microbiota profiles in treatment-naïve pediatric inflammatory bowel disease: associations with disease phenotype, treatment, and outcome. Clin Exp Gastroenterol. 2019; 12:37–49.
33. Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scand J Gastroenterol. 2016; 51:1407–1415.
Article
34. O’Mahony L, Feeney M, O’Halloran S, et al. Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther. 2001; 15:1219–1225.
Article
35. Peran L, Camuesco D, Comalada M, et al. Preventative effects of a probiotic, Lactobacillus salivarius ssp. salivarius, in the TNBS model of rat colitis. World J Gastroenterol. 2005; 11:5185–5192.
36. Feighery LM, Smith P, O’Mahony L, Fallon PG, Brayden DJ. Effects of Lactobacillus salivarius 433118 on intestinal inflammation, immunity status and in vitro colon function in two mouse models of inflammatory bowel disease. Dig Dis Sci. 2008; 53:2495–2506.
Article
37. Bossuyt P, Verhaegen J, Van Assche G, Rutgeerts P, Vermeire S. Increasing incidence of Clostridium difficile-associated diarrhea in inflammatory bowel disease. J Crohns Colitis. 2009; 3:4–7.
Article
38. Singh H, Nugent Z, Yu BN, Lix LM, Targownik LE, Bernstein CN. Higher incidence of Clostridium difficile infection among individuals with inflammatory bowel disease. Gastroenterology. 2017; 153:430–438.
Article
39. Rodemann JF, Dubberke ER, Reske KA, Seo DH, Stone CD. Incidence of Clostridium difficile infection in inflammatory bowel disease. Clin Gastroenterol Hepatol. 2007; 5:339–344.
Article
40. Ananthakrishnan AN, Binion DG. Impact of Clostridium difficile on inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2010; 4:589–600.
41. Maharshak N, Barzilay I, Zinger H, Hod K, Dotan I. Clostridium difficile infection in hospitalized patients with inflammatory bowel disease: prevalence, risk factors, and prognosis. Medicine (Baltimore). 2018; 97:e9772.
42. Ben-Horin S, Margalit M, Bossuyt P, et al. Combination immunomodulator and antibiotic treatment in patients with inflammatory bowel disease and Clostridium difficile infection. Clin Gastroenterol Hepatol. 2009; 7:981–987.
Article
43. Peeters M, Joossens S, Vermeire S, Vlietinck R, Bossuyt X, Rutgeerts P. Diagnostic value of anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease. Am J Gastroenterol. 2001; 96:730–734.
Article
44. Andersen K, Vogt C, Blondin D, et al. Multi-detector CT-colonography in inflammatory bowel disease: prospective analysis of CT-findings to high-resolution video colonoscopy. Eur J Radiol. 2006; 58:140–146.
Article
45. Scanlan PD, Shanahan F, O’Mahony C, Marchesi JR. Culture-independent analyses of temporal variation of the dominant fecal microbiota and targeted bacterial subgroups in Crohn’s disease. J Clin Microbiol. 2006; 44:3980–3988.
Article
46. Willing BP, Dicksved J, Halfvarson J, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010; 139:1844–1854.
Article
47. Seksik P, Rigottier-Gois L, Gramet G, et al. Alterations of the dominant faecal bacterial groups in patients with Crohn’s disease of the colon. Gut. 2003; 52:237–242.
Article
48. Wang W, Chen L, Zhou R, et al. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol. 2014; 52:398–406.
Article
49. Song YL, Liu CX, McTeague M, Summanen P, Finegold SM. Clostridium bartlettii sp. nov., isolated from human faeces. Anaerobe. 2004; 10:179–184.
Article
50. Rosario D, Benfeitas R, Bidkhori G, et al. Understanding the representative gut microbiota dysbiosis in metformin-treated type 2 diabetes patients using genome-scale metabolic modeling. Front Physiol. 2018; 9:775.
Article
51. Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013; 504:451–455.
Article
52. Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013; 504:446–450.
Article
53. Choi SI, Son JH, Kim N, et al. Changes in cecal microbiota and short-chain fatty acid during lifespan of the rat. J Neurogastroenterol Motil. 2021; 27:134–146.
Article
54. Beaud D, Tailliez P, Anba-Mondoloni J. Genetic characterization of the beta- glucuronidase enzyme from a human intestinal bacterium, Ruminococcus gnavus. Microbiology (Reading). 2005; 151(Pt 7):2323–2330.
Article
55. Joossens M, Huys G, Cnockaert M, et al. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut. 2011; 60:631–637.
Article
56. Tap J, Cools-Portier S, Pavan S, et al. Effects of the long-term storage of human fecal microbiota samples collected in RNAlater. Sci Rep. 2019; 9:601.
Article
Full Text Links
  • IR
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