RT Journal Article SR Electronic T1 Personalized modeling of the human gut microbiome reveals distinct bile acid deconjugation and biotransformation potential in healthy and IBD individuals JF bioRxiv FD Cold Spring Harbor Laboratory SP 229138 DO 10.1101/229138 A1 Almut Heinken A1 Dmitry A. Ravcheev A1 Federico Baldini A1 Laurent Heirendt A1 Ronan M.T. Fleming A1 Ines Thiele YR 2017 UL http://biorxiv.org/content/early/2017/12/04/229138.abstract AB The human gut microbiome performs important functions human health and disease. Intestinal microbes are capable of deconjugation and biotransformation of human primary bile acids to secondary bile acids. Alterations of the bile acid pool as a result of microbial dysbiosis have been linked to multifactorial diseases, such as inflammatory bowel disease (IBD).Constraint-based modeling is a powerful approach for the mechanistic, systems-level analysis of metabolic interactions in microbial communities. Recently, we constructed a resource of 773 curated genome-scale reconstructions of human gut microbes, AGORA. Here, we performed a comparative genomic analysis of bile acid deconjugation and biotransformation pathways in 693 human gut microbial genomes to expand these AGORA reconstructions accordingly (available at http://vmh.life).To elucidate the metabolic potential of individual microbiomes, publicly available metagenomic data from a cohort of healthy Western individuals, as well as two cohorts of IBD patients and healthy controls, were mapped onto the reference set of AGORA genomes. We constructed for each individual a large-scale personalized microbial community model that take strain-level abundances into account. Using flux balance analysis, we found that distinct potential to deconjugate and tranform primary bile acids between the gut microbiomes of healthy individuals. Moreover, the microbiomes of pediatric IBD patients were significantly depleted in their bile acid production potential compared with controls. The contributions of each strain to overall bile acid production potential across individuals were found to be distinct between IBD patients and controls. IBD microbiomes were depleted in contributions of Bacteroidetes strains but enriched in contributions of Proteobacteria. Finally, bottlenecks limiting secondary bile acid production potential were identified in each microbiome model. For ursodeoxycholate, the abundance of strains producing the precursor rather than of strains directly producing this secondary bile acid was synthesis-limiting in certain microbiomes.In summary, we integrated for the first-time metagenomics data with large-scale personalized metabolic modeling of microbial communities. We provided mechanistic insight into the link between dysbiosis and metabolic potential in IBD microbiomes. This large-scale modeling approach provides a novel way of analyzing metagenomics data to accelerate our understanding of the metabolic interactions between human host and gut microbiomes in health and diseases states.