Successful Dietary Therapy in Paediatric Crohn's Disease is Associated with Shifts in Bacterial Dysbiosis and Inflammatory Metabotype Towards Healthy Controls

Charlotte M Verburgt, Katherine A Dunn, Mohammed Ghiboub, James D Lewis, Eytan Wine, Rotem Sigall Boneh, Konstantinos Gerasimidis, Raanan Shamir, Susanne Penny, Devanand M Pinto, Alejandro Cohen, Paul Bjorndahl, Vaios Svolos, Joseph P Bielawski, Marc A Benninga, Wouter J de Jonge, Johan E Van Limbergen, Charlotte M Verburgt, Katherine A Dunn, Mohammed Ghiboub, James D Lewis, Eytan Wine, Rotem Sigall Boneh, Konstantinos Gerasimidis, Raanan Shamir, Susanne Penny, Devanand M Pinto, Alejandro Cohen, Paul Bjorndahl, Vaios Svolos, Joseph P Bielawski, Marc A Benninga, Wouter J de Jonge, Johan E Van Limbergen

Abstract

Background and aims: Nutritional therapy with the Crohn's Disease Exclusion Diet + Partial Enteral Nutrition [CDED+PEN] or Exclusive Enteral Nutrition [EEN] induces remission and reduces inflammation in mild-to-moderate paediatric Crohn's disease [CD]. We aimed to assess if reaching remission with nutritional therapy is mediated by correcting compositional or functional dysbiosis.

Methods: We assessed metagenome sequences, short chain fatty acids [SCFA] and bile acids [BA] in 54 paediatric CD patients reaching remission after nutritional therapy [with CDED + PEN or EEN] [NCT01728870], compared to 26 paediatric healthy controls.

Results: Successful dietary therapy decreased the relative abundance of Proteobacteria and increased Firmicutes towards healthy controls. CD patients possessed a mixture of two metabotypes [M1 and M2], whereas all healthy controls had metabotype M1. M1 was characterised by high Bacteroidetes and Firmicutes, low Proteobacteria, and higher SCFA synthesis pathways, and M2 was associated with high Proteobacteria and genes involved in SCFA degradation. M1 contribution increased during diet: 48%, 63%, up to 74% [Weeks 0, 6, 12, respectively.]. By Week 12, genera from Proteobacteria reached relative abundance levels of healthy controls with the exception of E. coli. Despite an increase in SCFA synthesis pathways, remission was not associated with increased SCFAs. Primary BA decreased with EEN but not with CDED+PEN, and secondary BA did not change during diet.

Conclusion: Successful dietary therapy induced correction of both compositional and functional dysbiosis. However, 12 weeks of diet was not enough to achieve complete correction of dysbiosis. Our data suggests that composition and metabotype are important and change quickly during the early clinical response to dietary intervention. Correction of dysbiosis may therefore be an important future treatment goal for CD.

Keywords: Crohn’s disease; Diet; inflammatory bowel disease; microbiome; treatment.

© The Author(s) 2022. Published by Oxford University Press on behalf of European Crohn’s and Colitis Organisation.

Figures

Figure 1.
Figure 1.
Beta-diversity plots of Bray–Curtis distances showing a gradual correction at Weeks 0, 6, 12 remission and Week 12 non-remission for patients achieving remission in Week 6 with EEN [n = 22, 20, 14, 6, respectively] and CDED + PEN [n = 30, 28, 25, 1, respectively] compared with healthy paediatric controls [n = 26] [Permanova p = 0.001]. EEN, exclusive enteral nutrition; CDED + PEN, Crohn’s disease exclusion diet + partial enteral nutrition.
Figure 2.
Figure 2.
LEfSe cladogram of taxa showing significant [p <0.05] differences in abundance of baseline treatment-naïve [Week 0] CD children [CDED + PEN and EEN groups, n = 52] compared with healthy controls [HC] [n = 26]. Analyses include only samples from patients reaching remission at Week 6. Each dot represents identified taxa in this data. Taxa highlighted in red are increased and highlighted in green are decreased in treatment-naïve [week 0] CD samples compared with HC. Detailed marked figure can be found in Supplementary Figure 1. CD, Crohn’s disease; EEN, exclusive enteral nutrition; CDED + PEN, Crohn’s disease exclusion diet + partial enteral nutrition; LEfSe, linear discriminant analysis of effect size.
Figure 3.
Figure 3.
LEfSe cladogram of taxa showing significant [p

Figure 4.

Changes in relative abundance [incl.…

Figure 4.

Changes in relative abundance [incl. standard deviation] of selected Proteobacteria across time points…

Figure 4.
Changes in relative abundance [incl. standard deviation] of selected Proteobacteria across time points for children on CDED + PEN and EEN compared to healthy controls. EEN, exclusive enteral nutrition; CDED + PEN, Crohn’s disease exclusion diet + partial enteral nutrition.

Figure 5.

Metabotype 1 community structure. Cytoscape…

Figure 5.

Metabotype 1 community structure. Cytoscape visualisation of the community structure identified from BioMiCo…

Figure 5.
Metabotype 1 community structure. Cytoscape visualisation of the community structure identified from BioMiCo analysis of Metabotype 1 samples using taxa identified in 16S rRNA gene data. Nodes represent predominate taxa identified in the analysis, with size showing the summed posterior probability of the contribution of those taxa across assemblages. Edges indicate taxa that co-occurred in an assemblage.

Figure 6.

Metabotype 2 community structure. Cytoscape…

Figure 6.

Metabotype 2 community structure. Cytoscape visualisation of the community structure identified from BioMiCo…

Figure 6.
Metabotype 2 community structure. Cytoscape visualisation of the community structure identified from BioMiCo analysis of metabotype 2 samples using taxa identified in 16S rRNA gene data. Nodes represent predominate taxa identified in the analysis, with size showing the summed posterior probability of the contribution of those taxa across assemblages. Edges indicate taxa that co-occurred in an assemblage.
Figure 4.
Figure 4.
Changes in relative abundance [incl. standard deviation] of selected Proteobacteria across time points for children on CDED + PEN and EEN compared to healthy controls. EEN, exclusive enteral nutrition; CDED + PEN, Crohn’s disease exclusion diet + partial enteral nutrition.
Figure 5.
Figure 5.
Metabotype 1 community structure. Cytoscape visualisation of the community structure identified from BioMiCo analysis of Metabotype 1 samples using taxa identified in 16S rRNA gene data. Nodes represent predominate taxa identified in the analysis, with size showing the summed posterior probability of the contribution of those taxa across assemblages. Edges indicate taxa that co-occurred in an assemblage.
Figure 6.
Figure 6.
Metabotype 2 community structure. Cytoscape visualisation of the community structure identified from BioMiCo analysis of metabotype 2 samples using taxa identified in 16S rRNA gene data. Nodes represent predominate taxa identified in the analysis, with size showing the summed posterior probability of the contribution of those taxa across assemblages. Edges indicate taxa that co-occurred in an assemblage.

References

    1. Coward S, Clement F, Benchimol EI, et al. . Past and future burden of inflammatory bowel diseases based on modeling of population-based data. Gastroenterology 2019;156:1345–53.e1344.
    1. Levine A, Sigall Boneh R, Wine E.. Evolving role of diet in the pathogenesis and treatment of inflammatory bowel diseases. Gut 2018;67:1726–38.
    1. Lloyd-Price J, Arze C, Ananthakrishnan AN, et al. . Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019;569:655–62.
    1. Gevers D, Kugathasan S, Denson LA, et al. . The treatment-naive microbiome in new-onset Crohn s disease. Cell Host Microbe 2014;15:382–92.
    1. Duvallet C, Gibbons SM, Gurry T, et al. . Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun 2017;8:1784.
    1. Douglas GM, Hansen R, Jones CMA, et al. . Multi-omics differentially classify disease state and treatment outcome in pediatric Crohn’s disease. Microbiome 2018;6:13.
    1. Vieira-Silva S, Sabino J, Valles-Colomer M, et al. . Quantitative microbiome profiling disentangles inflammation- and bile duct obstruction-associated microbiota alterations across PSC/IBD diagnoses. Nat Microbiol 2019. doi:10.1038/s41564-019-0483-9.
    1. Ding NS, McDonald JAK, Perdones-Montero A, et al. . Metabonomics and the gut microbiome associated with primary response to anti-TNF therapy in Crohn’s disease. J Crohns Colitis 2020. doi: 10.1093/ecco-jcc/jjaa039.
    1. Kennedy NA, Heap GA, Green HD, et al. . Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn’s disease: a prospective, multicentre, cohort study. Lancet Gastroenterol Hepatol 2019;4:341–53.
    1. Levine A, Wine E, Assa A, et al. . Crohn’s disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology 2019;157:440–50.e448.
    1. Turnbaugh PJ. Diet should be a tool for researchers, not a treatment. Nature 2020;577:S23.
    1. Sanna S, van Zuydam NR, Mahajan A, et al. . Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 2019;51:600–5.
    1. Quince C, Ijaz UZ, Loman N, et al. . Extensive modulation of the fecal metagenome in children with Crohn’s disease during exclusive enteral nutrition. Am J Gastroenterol 2015;110:1718–29; quiz 1730.
    1. Connors J, Basseri S, Grant A, et al. . Exclusive enteral nutrition therapy in paediatric Crohn’s disease results in long-term avoidance of corticosteroids: results of a propensity-score matched cohort analysis. J Crohns Colitis 2017;11:1063–70.
    1. Cohen-Dolev N, Sladek M, Hussey S, et al. . Differences in outcomes over time with exclusive enteral nutrition compared with steroids in children with mild to moderate Crohn’s disease: results from the GROWTH CD study. J Crohns Colitis 2018;12:306–12.
    1. Grover Z, Lewindon P.. Two-year outcomes after exclusive enteral nutrition induction are superior to corticosteroids in pediatric Crohn’s disease treated early with thiopurines. Dig Dis Sci 2015;60:3069–74.
    1. Levine A, Turner D, Pfeffer Gik T, et al. . Comparison of outcomes parameters for induction of remission in new onset pediatric Crohn’s disease: evaluation of the porto IBD group ‘growth relapse and outcomes with therapy’ [GROWTH CD] study. Inflamm Bowel Dis 2014;20:278–85.
    1. Ruemmele FM, Veres G, Kolho KL, et al. . Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn’s disease. J Crohns Colitis 2014;8:1179–207.
    1. Van Limbergen J, Haskett J, Griffiths AM, et al. . Toward enteral nutrition for the treatment of pediatric Crohn disease in Canada: a workshop to identify barriers and enablers. Can J Gastroenterol. Hepatol 2015;29:351–6.
    1. Lawley M, Wu JW, Navas-Lopez VM, et al. . Global variation in use of enteral nutrition for pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2018;67:e22–9.
    1. Svolos V, Hansen R, Nichols B, et al. . Treatment of active Crohn’s disease with an ordinary food-based diet that replicates exclusive enteral nutrition. Gastroenterology 2019;156:1354–67.e1356.
    1. Logan M, Clark CM, Ijaz UZ, et al. . The reduction of faecal calprotectin during exclusive enteral nutrition is lost rapidly after food re-introduction. Aliment Pharmacol Ther 2019. doi: 10.1111/apt.15425.
    1. Gerasimidis K, Bryden K, Chen X, et al. . The impact of food additives, artificial sweeteners and domestic hygiene products on the human gut microbiome and its fibre fermentation capacity. Eur J Nutr 2019. doi:10.1007/s00394-019-02161-8.
    1. Sigall Boneh R, Van Limbergen J, Wine E, et al. . Dietary therapies induce rapid response and remission in active paediatric Crohn’s disease. Clin Gastroenterol Hepatol 2021;19(4):752–59. doi:10.1016/j.cgh.2020.04.006.
    1. Chassaing B, Compher C, Bonhomme B, et al. . Randomized controlled-feeding study of dietary emulsifier carboxymethylcellulose reveals detrimental impacts on the gut microbiota and metabolome. Gastroenterology 2022;162:743–56.
    1. Maslowski KM, Mackay CR.. Diet, gut microbiota and immune responses. Nat Immunol 2011;12:5–9.
    1. Houron C, Ciocan D, Trainel N, et al. . Gut microbiota reshaped by pectin treatment improves liver steatosis in obese mice. Nutrients 2021;13. doi:10.3390/nu13113725.
    1. Shinohara K, Ohashi Y, Kawasumi K, Terada A, Fujisawa T.. Effect of apple intake on fecal microbiota and metabolites in humans. Anaerobe 2010;16:510–5.
    1. Turner D, Griffiths AM, Walters TD, et al. . Mathematical weighting of the pediatric Crohn’s disease activity index [PCDAI] and comparison with its other short versions. Inflamm Bowel Dis 2012;18:55–62.
    1. Lewis JD, Chen EZ, Baldassano RN, et al. . Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn’s disease. Cell Host Microbe 2015;18:489–500.
    1. Wu GD, Chen J, Hoffmann C, et al. . Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–8.
    1. Bolger AM, Lohse M, Usadel B.. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20.
    1. Langmead B, Salzberg SL.. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357–9.
    1. Franzosa EA, McIver LJ, Rahnavard G, et al. . Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018;15:962–8.
    1. Tange O. GNU Parallel 2018. Geneva: CERN, Zenodo, 2018, doi: 10.5281/zenodo.1146014.
    1. Kanehisa M, Goto S.. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000;28:27–30.
    1. Shafiei M, Dunn KA, Chipman H, Gu H, Bielawski JP.. BiomeNet: a Bayesian model for inference of metabolic divergence among microbial communities. PLoS Comput Biol 2014;10:e1003918.
    1. Segata N, Waldron L, Ballarini A, et al. . Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods 2012;9:811–4.
    1. Segata N, Izard J, Waldron L, et al. . Metagenomic biomarker discovery and explanation. Genome Biol 2011;12:R60.
    1. Morton JT, Marotz C, Washburne A, et al. . Establishing microbial composition measurement standards with reference frames. Nat Commun 2019;10:2719.
    1. Fernandes AD, Reid J, Macklaim JM, McMurrough TA, et al. . Unifying the analysis of high-throughput sequencing datasets: characterizing RNA-seq, 16S rRNA gene sequencing and selective growth experiments by compositional data analysis. Microbiome 2014;5:15.
    1. Fedarko MW, Martino C, Morton JT, et al. . Visualizing ‘omic feature rankings and log-ratios using Qurro. NAR Genom Bioinform 2020;2:lqaa023.
    1. Verburgt CM, Dunn KA, Van Limbergen JE.. Dietary therapy reduces pro-inflammatory microbiome features in pediatric Crohn’s disease. J Crohns Colitis 2021. doi:10.1093/ecco-jcc/jjab197.
    1. Abulizi N, Quin C, Brown K, et al. . Gut mucosal proteins and bacteriome are shaped by the saturation index of dietary lipids. Nutrients 2019;11. doi:10.3390/nu11020418.
    1. Sigall Boneh R, Van Limbergen J, Assa A, et al. . DOP42 Dietary therapies induce rapid response and remission in active paediatric Crohn’s disease. J Crohns Colitis 2019;13:S050.
    1. Horng YT, Wang CJ, Chung WT, et al. . Phosphoenolpyruvate phosphotransferase system components positively regulate Klebsiella biofilm formation. J Microbiol Immunol Infect 2018;51:174–83.
    1. Lin D, Fan J, Wang J, et al. . The fructose-specific phosphotransferase system of Klebsiella pneumoniae is regulated by global regulator CRP and linked to virulence and growth. Infect Immun 2018;86. doi:10.1128/iai.00340-18.
    1. Lim S, Seo HS, Jeong J, Yoon H.. Understanding the multifaceted roles of the phosphoenolpyruvate: phosphotransferase system in regulation of Salmonella virulence using a mutant defective in ptsI and crr expression. Microbiol Res 2019;223–225:63–71.
    1. Deutscher J, Aké FMD, Derkaoui M, et al. . The bacterial phosphoenolpyruvate:carbohydrate phosphotransferase system: regulation by protein phosphorylation and phosphorylation-dependent protein-protein interactions. Microbiol Mol Biol Rev 2014;78, 231–56.
    1. Jenkins LS, Nunn WD.. Regulation of the ato operon by the atoC gene in Escherichia coli. J Bacteriol 1987;169:2096–102.
    1. Llewellyn SR, Britton GJ, Contijoch EJ, et al. . Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice. Gastroenterology 2018;154:1037–46.e1032.
    1. Singh V, Yeoh BS, Walker RE, et al. . Microbiota fermentation-NLRP3 axis shapes the impact of dietary fibres on intestinal inflammation. Gut 2019. doi:10.1136/gutjnl-2018-316250.
    1. Makki K, Deehan EC, Walter J, Backhed F.. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe 2018;23:705–15.
    1. Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F.. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016;165:1332–45.
    1. Ijaz UZ, Quince C, Hanske L, et al. . The distinct features of microbial ‘dysbiosis’ of Crohn’s disease do not occur to the same extent in their unaffected, genetically-linked kindred. PLoS One 2017;12:e0172605.
    1. Aden K, Rehman A, Waschina S, et al. . Metabolic functions of gut microbes associate with efficacy of tumor necrosis factor antagonists in patients with inflammatory bowel diseases. Gastroenterology 2019;157:1279–92.e1211.
    1. Effenberger M, Reider S, Waschina S, et al. . Microbial butyrate synthesis indicates therapeutic efficacy of azathioprine in IBD patients. J Crohns Colitis 2020. doi:10.1093/ecco-jcc/jjaa152.
    1. Chen ML, Takeda K, Sundrud MS.. Emerging roles of bile acids in mucosal immunity and inflammation. Mucosal Immunol 2019;12:851–61.
    1. Wahlström A, Sayin SI, Marschall HU, Bäckhed F.. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 2016;24:41–50.
    1. Connors J, Dunn KA, Allott J, et al. . The relationship between fecal bile acids and microbiome community structure in pediatric Crohn’s disease. Isme J 2020;14:702–13.
    1. Duboc H, Rajca S, Rainteau D, et al. . Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut 2013;62:531–9.
    1. Sinha SR, Haileselassie Y, Nguyen LP, et al. . Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 2020;27:659–70.e655.
    1. Lee D, Baldassano RN, Otley AR, et al. . Comparative effectiveness of nutritional and biological therapy in North American children with active Crohn’s disease. Inflamm Bowel Dis 2015;21:1786–93.

Source: PubMed

3
Tilaa