Gut microbiome structure and metabolic activity in inflammatory bowel disease

Eric A Franzosa, Alexandra Sirota-Madi, Julian Avila-Pacheco, Nadine Fornelos, Henry J Haiser, Stefan Reinker, Tommi Vatanen, A Brantley Hall, Himel Mallick, Lauren J McIver, Jenny S Sauk, Robin G Wilson, Betsy W Stevens, Justin M Scott, Kerry Pierce, Amy A Deik, Kevin Bullock, Floris Imhann, Jeffrey A Porter, Alexandra Zhernakova, Jingyuan Fu, Rinse K Weersma, Cisca Wijmenga, Clary B Clish, Hera Vlamakis, Curtis Huttenhower, Ramnik J Xavier, Eric A Franzosa, Alexandra Sirota-Madi, Julian Avila-Pacheco, Nadine Fornelos, Henry J Haiser, Stefan Reinker, Tommi Vatanen, A Brantley Hall, Himel Mallick, Lauren J McIver, Jenny S Sauk, Robin G Wilson, Betsy W Stevens, Justin M Scott, Kerry Pierce, Amy A Deik, Kevin Bullock, Floris Imhann, Jeffrey A Porter, Alexandra Zhernakova, Jingyuan Fu, Rinse K Weersma, Cisca Wijmenga, Clary B Clish, Hera Vlamakis, Curtis Huttenhower, Ramnik J Xavier

Abstract

The inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are multifactorial chronic conditions of the gastrointestinal tract. While IBD has been associated with dramatic changes in the gut microbiota, changes in the gut metabolome-the molecular interface between host and microbiota-are less well understood. To address this gap, we performed untargeted metabolomic and shotgun metagenomic profiling of cross-sectional stool samples from discovery (n = 155) and validation (n = 65) cohorts of CD, UC and non-IBD control patients. Metabolomic and metagenomic profiles were broadly correlated with faecal calprotectin levels (a measure of gut inflammation). Across >8,000 measured metabolite features, we identified chemicals and chemical classes that were differentially abundant in IBD, including enrichments for sphingolipids and bile acids, and depletions for triacylglycerols and tetrapyrroles. While > 50% of differentially abundant metabolite features were uncharacterized, many could be assigned putative roles through metabolomic 'guilt by association' (covariation with known metabolites). Differentially abundant species and functions from the metagenomic profiles reflected adaptation to oxidative stress in the IBD gut, and were individually consistent with previous findings. Integrating these data, however, we identified 122 robust associations between differentially abundant species and well-characterized differentially abundant metabolites, indicating possible mechanistic relationships that are perturbed in IBD. Finally, we found that metabolome- and metagenome-based classifiers of IBD status were highly accurate and, like the vast majority of individual trends, generalized well to the independent validation cohort. Our findings thus provide an improved understanding of perturbations of the microbiome-metabolome interface in IBD, including identification of many potential diagnostic and therapeutic targets.

Conflict of interest statement

Competing interests

Figures

Figure 1.. IBD is associated with broad…
Figure 1.. IBD is associated with broad changes in subjects’ gut multi’omic profiles.
(A) We collected and profiled stool metagenomic and metabolomic data from two IBD cohorts: a 155-member discovery cohort (PRISM) and a 65-member validation cohort (NLIBD/LLDeep). (B) Principal coordinates analysis (PCoA) of PRISM cohort subjects based on gut metabolomic profiles (Bray-Curtis distance). (C) The same subjects ordinated on Bray-Curtis distances between gut metagenomic species profiles. (D, E) Subject fecal calprotectin (FC) levels (μg/g) plotted against the first PCoA axes from panels B and C, respectively. Note that FC measurements were not available for all subjects.
Figure 2.. Metabolic enrichments in IBD versus…
Figure 2.. Metabolic enrichments in IBD versus control phenotypes.
We applied Wilcoxon rank-sum tests to metabolites’ individual differential abundance trends (t-statistics from the linear models) to identify classes of molecules that were broadly enriched in IBD. Focusing on classes of molecules with at least 10 putative members (see the “n=“ column), (A) eight were significantly (FDR q<0.05) positively enriched in CD, meaning that their members tended to be more abundant in CD, and 17 classes were significantly negatively enriched, meaning that their members tended to be more abundant in controls (nominal p-values were two-tailed). (B) A subset of these trends were similarly significant in comparisons between UC and controls, with the remainder (gray) tending to trend in the same direction as CD vs. control comparisons. The dotted line indicates the significance threshold for an individual metabolic feature [abs(t)>2.61]. Panels C through H highlight examples of individually differentially abundant standards measured across 68 CD, 53 UC, and 34 non-IBD control subjects. Metabolites highlighted in panels C, D, E, and F are representatives of broader classes analyzed in A and B. Abundances are in units of parts per million (PPM) after separately sum-normalizing within each LC-MS method; values are square-root scaled for visualization. Boxplot “boxes” indicate the first, second, and third quartiles of the data. Boxplot “whiskers” indicate the inner fences of the data, with points outside the inner fences plotted as outliers.
Figure 3.. Clusters of chemically related, IBD-perturbed…
Figure 3.. Clusters of chemically related, IBD-perturbed metabolites revealed by abundance covariation.
We clustered differentially abundant (DA) metabolites after regressing out the effects of diagnosis, subject age, and medication use (Methods). A small number of (large) clusters explained many of the DA metabolites. (A) The second-largest cluster contained 39 metabolite features, all of them significantly elevated among CD patients (and one in UC patients as well). This cluster was enriched for putative bile acids and derivatives. Multiple variants of the standards cholate (light green triangles) and chenodeoxycholate (dark green triangles) occur in this cluster. (B) The largest cluster contained 62 metabolite features, all of them significantly elevated in non-IBD controls. This cluster was enriched for putative tetrapyrroles and derivatives. The 155 samples (columns) are ordered the same way in both panels according to Bray-Curtis similarity (and phenotype) of overall metabolic profile (as established in Supplementary Fig. 1). Note the control-like versus CD-like substructure among UC subjects.
Figure 4.. Potentially mechanistic associations between IBD-linked…
Figure 4.. Potentially mechanistic associations between IBD-linked microbes and metabolites.
(A) Covariation between microbes and small molecules DA in IBD, specifically those linking FDR-significant, confirmed-in-controls metagenomic species and metabolites matched against standards (Spearman correlation with two-tailed nominal p-values). When multiple metabolomic features matched the same standard, the feature with the highest mean absolute correlation was selected for plotting. Starred (*) metabolites indicate a match to a standard with isomeric forms that could not be differentiated. The standard L-1,2,3,4-Tetrahydro-beta-carboline-3-carboxylic acid is listed as “cyclomethyltryptophan.” (B), (C), and (D) highlight examples of individual correlations across 68 CD, 53 UC, and 34 non-IBD control subjects (see text). Metabolites and species in these examples are colored in panel A. Values plotted are raw measurements (not residuals) normalized to parts per million (PPM) units and then log10-transformed. Values <1 PPM (including 0s) were set to 1 PPM for plotting; corresponding points are shown without fill and jittered (all other points have solid fill).
Figure 5.. IBD-associated changes in microbial function…
Figure 5.. IBD-associated changes in microbial function and their metabolic associations.
(A) - (E) highlight examples of metagenomically contributed enzymes that were differentially abundant in IBD, annotated by their taxonomic contributors (A - C are enriched in IBD; D and E are depleted). In each case, the enzyme was contributed by a mixture of species across the cohort, and not dominated by a single species. Each set of stacked bars represents one of the 155 PRISM metagenomes (arrayed on horizontal axes). Community enzyme abundance (log10-transformed parts per million) is represented by the top of each stack of bars; contributions from major species are linearly scaled within the total bar height. Samples are first sorted according to the dominant contributor to a function and then grouped by phenotype (sample ordering differs between panels). (F) and (G) illustrate correlations between community-total enzyme abundance and IBD-associated metabolites across 68 CD, 53 UC, and 34 non-IBD control subjects. Values plotted are raw measurements (not residuals) normalized to parts per million (PPM) units and then log10-transformed. Values <1 PPM (including 0s) were set to 1 PPM for plotting; corresponding points are shown without fill and jittered (all other points have solid fill). The given r values indicate Spearman correlation.
Figure 6.. Predicting IBD status and subtype…
Figure 6.. Predicting IBD status and subtype from gut microbiome multi’omic features.
We trained random forest classifiers on metabolites, microbial species, and their combination to identify IBD patients and IBD subtypes. Training/testing was carried out within the PRISM cohort using five-fold cross-validation, in addition to models trained on the full PRISM cohort and then tested (validated) on the independent Netherlands cohorts. (A) ROC curves depict trade-offs between classifiers’ true positive rates (TPRs) and false positive rates (FPRs) as classification stringency varies. The area under the curve (AUC) statistic is a summary measure of classifier performance: AUC values close to 1 indicate that a high TPR was achieved with low FPR (ideal performance), while AUC values close to 0.5 indicate random performance. (B) “Confusion matrix” evaluations of IBD subtype classifiers within the Boston PRISM cohort. The number in row i and column j indicates how many samples were labeled as subtype i but assigned to subtype j. A perfect subtype classifier (100% accuracy) would have 0 counts for all non-diagonal entries (i.e. no misclassified samples). Matrix cells are shaded within-row in proportion to their value (red for CD, orange for UC, and blue for non-IBD control). (C) Confusion matrix evaluations of IBD subtype classifiers trained on the Boston PRISM cohort and tested on the independent Netherlands cohorts. Accuracy values in B and C indicate the fraction of correctly classified instances; error values reflect the standard error of a proportion.

References

    1. Wlodarska M, Kostic AD & Xavier RJ An integrative view of microbiome-host interactions in inflammatory bowel diseases. Cell Host Microbe 17, 577–591 (2015).
    1. Imhann F et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut (2016).
    1. Huttenhower C, Kostic AD & Xavier RJ Inflammatory bowel disease as a model for translating the microbiome. Immunity 40, 843–854 (2014).
    1. Morgan XC et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13, R79 (2012).
    1. Gevers D et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 15, 382–392 (2014).
    1. Haberman Y et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J. Clin. Invest. 124, 3617–3633 (2014).
    1. Lane ER, Zisman TL & Suskind DL The microbiota in inflammatory bowel disease: current and therapeutic insights. J. Inflamm. Res. 10, 63–73 (2017).
    1. Blander JM, Longman RS, Iliev ID, Sonnenberg GF & Artis D Regulation of inflammation by microbiota interactions with the host. Nat. Immunol. 18, 851–860 (2017).
    1. Dorrestein PC, Mazmanian SK & Knight R Finding the missing links among metabolites, microbes, and the host. Immunity 40, 824–832 (2014).
    1. McHardy IH et al. Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. Microbiome 1, 17 (2013).
    1. Wu GD Diet, the gut microbiome and the metabolome in IBD. Nestle Nutr. Inst. Workshop Ser. 79, 73–82 (2014).
    1. Kim S, Kim J-H, Park BO & Kwak YS Perspectives on the therapeutic potential of short-chain fatty acid receptors. BMB Rep. 47, 173–178 (2014).
    1. Smith PM et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).
    1. Fernando MR, Saxena A, Reyes J-L & McKay DM Butyrate enhances antibacterial effects while suppressing other features of alternative activation in IL-4-induced macrophages. Am. J. Physiol. Gastrointest. Liver Physiol. 310, G822–831 (2016).
    1. Marchesi JR et al. Rapid and noninvasive metabonomic characterization of inflammatory bowel disease. J. Proteome Res. 6, 546–551 (2007).
    1. Wikoff WR et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U. S. A. 106, 3698–3703 (2009).
    1. Williams BB et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 16, 495–503 (2014).
    1. Zelante T et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).
    1. Lamas B et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 22, 598–605 (2016).
    1. Le Gall G et al. Metabolomics of fecal extracts detects altered metabolic activity of gut microbiota in ulcerative colitis and irritable bowel syndrome. J. Proteome Res. 10, 4208–4218 (2011).
    1. Bjerrum JT et al. Metabonomics of human fecal extracts characterize ulcerative colitis, Crohn’s disease and healthy individuals. Metabolomics 11, 122–133 (2015).
    1. De Preter V et al. Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD. Gut 64, 447–458 (2015).
    1. Jansson J et al. Metabolomics reveals metabolic biomarkers of Crohn’s disease. PLoS One 4, e6386 (2009).
    1. Kolho K-L, Pessia A, Jaakkola T, de Vos WM & Velagapudi V Faecal and Serum Metabolomics in Paediatric Inflammatory Bowel Disease. J. Crohns. Colitis 11, 321–334 (2017).
    1. Jacobs JP et al. A Disease-Associated Microbial and Metabolomics State in Relatives of Pediatric Inflammatory Bowel Disease Patients. Cell Mol Gastroenterol Hepatol 2, 750–766 (2016).
    1. Melnik AV et al. Coupling Targeted and Untargeted Mass Spectrometry for Metabolome-Microbiome-Wide Association Studies of Human Fecal Samples. Anal. Chem. 89, 7549–7559 (2017).
    1. Sokol H & Seksik P The intestinal microbiota in inflammatory bowel diseases: time to connect with the host. Curr. Opin. Gastroenterol. 26, 327–331 (2010).
    1. Joossens M et al. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 60, 631–637 (2011).
    1. Sokol H et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm. Bowel Dis. 15, 1183–1189 (2009).
    1. Wishart DS et al. HMDB: the Human Metabolome Database. Nucleic Acids Res. 35, D521–526 (2007).
    1. Mosli MH et al. C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 110, 802–819; quiz 820 (2015).
    1. Benjamini Y & Hochberg Y Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol, 289–300 (1995).
    1. Duboc H et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut 62, 531–539 (2013).
    1. Abdel Hadi L, Di Vito C & Riboni L Fostering Inflammatory Bowel Disease: Sphingolipid Strategies to Join Forces. Mediators Inflamm. 2016, 3827684 (2016).
    1. An D et al. Sphingolipids from a symbiotic microbe regulate homeostasis of host intestinal natural killer T cells. Cell 156, 123–133 (2014).
    1. Braun A et al. Alterations of phospholipid concentration and species composition of the intestinal mucus barrier in ulcerative colitis: a clue to pathogenesis. Inflamm. Bowel Dis. 15, 1705–1720 (2009).
    1. Qi Y et al. PPARα-dependent exacerbation of experimental colitis by the hypolipidemic drug fenofibrate. Am. J. Physiol. Gastrointest. Liver Physiol. 307, G564–573 (2014).
    1. Fischbeck A et al. Sphingomyelin induces cathepsin D-mediated apoptosis in intestinal epithelial cells and increases inflammation in DSS colitis. Gut 60, 55–65 (2011).
    1. Heimerl S et al. Alterations in intestinal fatty acid metabolism in inflammatory bowel disease. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1762, 341–350 (2006).
    1. Hove H & Mortensen PB Influence of intestinal inflammation (IBD) and small and large bowel length on fecal short-chain fatty acids and lactate. Dig. Dis. Sci. 40, 1372–1380 (1995).
    1. Stuart JM, Segal E, Koller D & Kim SK A gene-coexpression network for global discovery of conserved genetic modules. Science 302, 249–255 (2003).
    1. Wolfe CJ, Kohane IS & Butte AJ Systematic survey reveals general applicability of” guilt-by-association” within gene coexpression networks. BMC Bioinformatics 6, 227 (2005).
    1. Frank DN et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. U. S. A. 104, 13780–13785 (2007).
    1. Lewis JD et al. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn’s Disease. Cell Host Microbe 18, 489–500 (2015).
    1. Desbois AP & Smith VJ Antibacterial free fatty acids: activities, mechanisms of action and biotechnological potential. Appl. Microbiol. Biotechnol. 85, 1629–1642 (2010).
    1. German JB & Dillard CJ Saturated fats: a perspective from lactation and milk composition. Lipids 45, 915–923 (2010).
    1. Galland L Magnesium and inflammatory bowel disease. Magnesium 7, 78–83 (1988).
    1. Lih-Brody L et al. Increased oxidative stress and decreased antioxidant defenses in mucosa of inflammatory bowel disease. Dig. Dis. Sci. 41, 2078–2086 (1996).
    1. Yang JY et al. Molecular networking as a dereplication strategy. J Nat Prod 76, 1686–1699 (2013).
    1. Jaskowski TD, Litwin CM & Hill HR Analysis of serum antibodies in patients suspected of having inflammatory bowel disease. Clin. Vaccine Immunol. 13, 655–660 (2006).
    1. Tigchelaar EF et al. Cohort profile: LifeLines DEEP, a prospective, general population cohort study in the northern Netherlands: study design and baseline characteristics. BMJ Open 5, e006772 (2015).
    1. Bolger AM, Lohse M & Usadel B Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).
    1. Langmead B & Salzberg SL Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
    1. Segata N et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods 9, 811–814 (2012).
    1. Franzosa E et al. Functionally profiling metagenomes and metatranscriptomes at species-level resolution. Nature Methods (In Proof).
    1. Suzek BE et al. UniRef clusters: a comprehensive and scalable alternative for improving sequence similarity searches. Bioinformatics 31, 926–932 (2015).
    1. Buchfink B, Xie C & Huson DH Fast and sensitive protein alignment using DIAMOND. Nat. Methods 12, 59–60 (2015).
    1. Apweiler R et al. UniProt: the Universal Protein knowledgebase. Nucleic Acids Res. 32, D115–119 (2004).
    1. Benjamini Y & Hochberg Y Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).

Source: PubMed

3
Subscribe