Intest Res.  2022 Jul;20(3):370-380. 10.5217/ir.2021.00054.

Relationship between the gut microbiota and bile acid composition in the ileal mucosa of Crohn’s disease

Affiliations
  • 1Division of Gastroenterology, Shiga University of Medical Science, Otsu, Japan
  • 2Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
  • 3Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
  • 4Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
  • 5Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA

Abstract

Background/Aims
Crosstalk between the gut microbiota and bile acid plays an important role in the pathogenesis of gastrointestinal disorders. We investigated the relationship between microbial structure and bile acid metabolism in the ileal mucosa of Crohn’s disease (CD).
Methods
Twelve non-CD controls and 38 CD patients in clinical remission were enrolled. Samples were collected from the distal ileum under balloon-assisted enteroscopy. Bile acid composition was analyzed by liquid chromatography-mass spectrometry. The gut microbiota was analyzed by 16S rRNA gene sequencing.
Results
The Shannon evenness index was significantly lower in endoscopically active lesions than in non-CD controls. β-Diversity, evaluated by the UniFrac metric, revealed a significant difference between the active lesions and non-CD controls (P=0.039). The relative abundance of Escherichia was significantly higher and that of Faecalibacterium and Roseburia was significantly lower in CD samples than in non-CD controls. The increased abundance of Escherichia was more prominent in active lesions than in inactive lesions. The proportion of conjugated bile acids was significantly higher in CD patients than in non-CD controls, but there was no difference in the proportion of primary or secondary bile acids. The genera Escherichia and Lactobacillus were positively correlated with the proportion of conjugated bile acids. On the other hand, Roseburia, Intestinibacter, and Faecalibacterium were negatively correlated with the proportion of conjugated bile acids.
Conclusions
Mucosa-associated dysbiosis and the alteration of bile acid composition were identified in the ileum of CD patients. These may play a role in the pathophysiology of ileal lesions in CD patients.

Keyword

Single-balloon enteroscopy; Crohn disease; Mucosa associated microbiota

Figure

  • Fig. 1. Comparative analysis of the gut microbial communities in non-CD controls (n=9) and inactive (n=18) and active lesions (n=9) of CD patients. (A) Observed species. (B) Chao1 index. (C) Shannon index. (D) β-Diversity estimated using the UniFrac metric and visualized using NMDS ordination. There was a significant difference between active lesions and non-CD samples (P=0.039, permutational multivariate analysis of variance). aP<0.05, Mann-Whitney U test. CD, Crohn’s disease; NMDS, non-metric multidimensional scaling; NS, not significant.

  • Fig. 2. Comparative analysis of the taxonomic composition of the microbial community at the phylum level in non-CD controls (n=9) and inactive (n=18) and active lesions (n=9) of CD patients. (A) Firmicutes. (B) Bacteroidetes. (C) Proteobacteria. (D) Fusobacteria. (E) Actinobacteria. aP<0.05, Mann-Whitney U test. CD, Crohn’s disease; NS, not significant.

  • Fig. 3. Comparative analysis of the taxonomic composition of the microbial community at the genus level using linear discriminant analysis effect size. (A) Comparison between non-CD (n=9) and CD samples (n=27). (B) Comparison between inactive (n=18) and active lesions (n=9) of CD patients. CD, Crohn’s disease.

  • Fig. 4. Bile acid composition in the ileum of non-CD controls (n=10) and inactive (n=25) and active CD patients (n=8). (A) Bile acid composition was visualized using NMDS ordination. There was a significant difference between active CD and non-CD controls (P=0.033, permutational multivariate analysis of variance). (B) Total bile acids. (C) Proportion of primary bile acids. (D) Proportion of secondary bile acids. (E) Proportion of conjugated bile acids. (F) Proportion of unconjugated bile acids. (G) Ratio of glycine-conjugated to taurine-conjugated bile acids. aP<0.05, Mann-Whitney U test. CD, Crohn’s disease; NMDS, non-metric multidimensional scaling; NS, not significant.


Cited by  1 articles

Compositional changes in fecal microbiota associated with clinical phenotypes and prognosis in Korean patients with inflammatory bowel disease
Seung Yong Shin, Young Kim, Won-Seok Kim, Jung Min Moon, Kang-Moon Lee, Sung-Ae Jung, Hyesook Park, Eun Young Huh, Byung Chang Kim, Soo Chan Lee, Chang Hwan Choi
Intest Res. 2023;21(1):148-160.    doi: 10.5217/ir.2021.00168.


Reference

1. Sheehan D, Moran C, Shanahan F. The microbiota in inflammatory bowel disease. J Gastroenterol. 2015; 50:495–507.
Article
2. Goldsmith JR, Sartor RB. The role of diet on intestinal microbiota metabolism: downstream impacts on host immune function and health, and therapeutic implications. J Gastroenterol. 2014; 49:785–798.
Article
3. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018; 11:1–10.
Article
4. Mak WY, Zhao M, Ng SC, Burisch J. The epidemiology of inflammatory bowel disease: east meets west. J Gastroenterol Hepatol. 2020; 35:380–389.
Article
5. Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2015; 12:205–217.
Article
6. Kaplan GG, Ng SC. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology. 2017; 152:313–321.
Article
7. Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology. 2008; 134:577–594.
Article
8. Takahashi K, Nishida A, Fujimoto T, et al. Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in Crohn’s disease. Digestion. 2016; 93:59–65.
Article
9. Nagalingam NA, Lynch SV. Role of the microbiota in inflammatory bowel diseases. Inflamm Bowel Dis. 2012; 18:968–984.
Article
10. 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
11. Nishino K, Nishida A, Inoue R, et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J Gastroenterol. 2018; 53:95–106.
Article
12. Litvak Y, Byndloss MX, Tsolis RM, Bäumler AJ. Dysbiotic Proteobacteria expansion: a microbial signature of epithelial dysfunction. Curr Opin Microbiol. 2017; 39:1–6.
Article
13. Lloyd-Price J, Arze C, Ananthakrishnan AN, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019; 569:655–662.
Article
14. Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol. 2018; 15:111–128.
Article
15. Sinha SR, Haileselassie Y, Nguyen LP, et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe. 2020; 27:659–670.
Article
16. Begley M, Gahan CG, Hill C. The interaction between bacteria and bile. FEMS Microbiol Rev. 2005; 29:625–651.
Article
17. 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
18. Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020; 17:223–237.
Article
19. Duboc H, Rajca S, Rainteau D, et al. Connecting dysbiosis, bileacid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut. 2013; 62:531–539.
Article
20. 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.
21. Hasegawa M, Inohara N. Regulation of the gut microbiota by the mucosal immune system in mice. Int Immunol. 2014; 26:481–487.
Article
22. Schloss PD, Westcott SL, Ryabin T, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009; 75:7537–7541.
Article
23. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009; 326:1694–1697.
Article
24. Murakami M, Iwamoto J, Honda A, et al. Detection of gut dysbiosis due to reduced Clostridium subcluster XIVa using the fecal or serum bile acid profile. Inflamm Bowel Dis. 2018; 24:1035–1044.
Article
25. Shoda J, Mahara R, Osuga T, et al. Similarity of unusual bile acids in human umbilical cord blood and amniotic fluid from newborns and in sera and urine from adult patients with cholestatic liver diseases. J Lipid Res. 1988; 29:847–858.
Article
26. Nagayama M, Yano T, Atarashi K, et al. TH1 cell-inducing Escherichia coli strain identified from the small intestinal mucosa of patients with Crohn’s disease. Gut Microbes. 2020; 12:1788898.
27. Johansson ME, Hansson GC. Immunological aspects of intestinal mucus and mucins. Nat Rev Immunol. 2016; 16:639–649.
Article
28. Litvak Y, Byndloss MX, Bäumler AJ. Colonocyte metabolism shapes the gut microbiota. Science. 2018; 362:eaat9076.
Article
29. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007; 369:1641–1657.
Article
30. Ridlon JM, Harris SC, Bhowmik S, Kang DJ, Hylemon PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes. 2016; 7:22–39.
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