Baat −/− KO mice have heightened pathology compared to wild-type mice under DSS treatment
As an approach to investigate the roles of BA conjugation in gut inflammation, we employed a recently developed Baat−/− KO mouse model that has highly reduced conjugated BAs, particularly reduced TCA (full characterization of Baat−/− KO model available in [26, 38]). The animal experiment was carried out according to the procedure shown in Fig. 1a. Mice in the DSS groups were treated with 2.5% DSS for 7 days and were then humanely euthanized on day 7. Daily measurement of body weight showed that Baat−/− KO mice lost more weight compared with WT animals during the course of the experiment (Fig. 1b). The DAI score (Fig. 1c) in the KO-DSS group was significantly higher after 7 days compared to the WT-DSS group (p < 0.05), indicating greater impact of DSS on Baat−/− KO mice. Additionally, the measurement of colon lengths demonstrated that Baat−/− KO group mice displayed significantly (p < 0.05) shorter colons compared to the WT-DSS group (Fig. 1d, e). There was no significant difference in histologic score, epithelial damage, nor mucosal depth in Baat−/− KO mice compared with WT mice under DSS treatment (Fig. 1f, g). Taken together, the results indicated that Baat−/− KO mice showed enhanced signs of DSS-induced colitis compared to the wild-type mice, without a notable difference in histopathology.
TCA supplementation rescues severe phenotypes of Baat−/− KO mice under DSS-induced colitis
Because our previous study found that the Baat−/− KO mice had 27-fold lower taurine-conjugated BAs than WT in their liver [26], we hypothesized that TCA plays a vital role in the heightened host response to DSS in these animals. To test this, we supplemented Baat−/− KO mice with chow containing 0.3% TCA with and without DSS treatment (Fig. 2a). After 6 days of treatment with 2.5% DSS, mice in the DSS-TCA treatment group lost less weight compared to those only exposed to DSS (Fig. 2b). Moreover, TCA supplementation resulted in a more rapid recovery of body weight during the 7-day recovery period post-DSS treatment than for mice only exposed to DSS (p < 0.05, Fig. 2b). Additionally, TCA supplementation had a positive effect on the length of the mouse colon (Fig. 2c, d, p < 0.05) and reduced the impact of DSS treatment on the spleen index (spleen weight: body weight ratio) (Fig. 2e, p < 0.05). The DAI score on day 13 in the DSS-TCA group was also significantly lower compared to the DSS group (Fig. 2f, p < 0.05). While the overall histologic scores were not significantly different (Fig. 2g, h), the number of goblet cells counted by AB staining was higher in DSS-TCA group compared to the DSS group (p = 0.062), with visual detection of goblet cells and mucus staining returning in mice receiving TCA supplementation (Fig. 2i, j).
TCA treatment increased the expression of mucosal integrity and BA receptors markers in Baat−/− KO mice on DSS
In light of the histological analysis indicating improved goblet cell function and mucus production we explored the gene expression changes related to cell barrier integrity and mucus physiology in the colons in DSS mice supplemented with TCA by RT-qPCR. The gene expression of MUC-2 was increased in the DSS-TCA group compared to the DSS group (Fig. 2k, p < 0.05), which is consistent with the AB-PAS goblet cell counts. Additionally, we found that the expression of intercellular tight junction genes, including Claudin-1, Occludin-1, and ZO-1 was higher in the TCA-supplemented groups compared to the non-supplemented groups, though only ZO-1 reached significance at an alpha-level below 0.05 (Fig. 2i and Figure S3a, b, Claudin-1 p = 0.09).
To gain further insight into the mechanism by which TCA affects intestinal inflammation, we also measured mRNA expression levels of pro-inflammatory factors and bile acid-related receptors (Figure S3c, d). Tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) gene expressions were higher in DSS treated animals as expected, however, the DSS-TCA and DSS groups were not significantly different from each other (p > 0.05). Expression of BA receptors FXR and TGR5 were significantly increased in the TCA treatment groups compared to untreated groups (p < 0.05) and were significantly higher in the TCA group than in the TCA-DSS group (Fig. 2m, n, p < 0.05).
Microbiome changes from TCA treatment of Baat−/− KO mice on DSS
To further investigate the effects of TCA supplementation on Baat−/− KO mice under DSS-induced colitis, the cecum and feces were collected for 16S rRNA gene amplicon sequencing. Microbiome community changes among the treatment groups were similar in the cecum and fecal data (Figure S1), we therefore focused more detailed analysis on the cecum profiles. TCA treatment slightly decreased the microbiome alpha-diversity and species richness compared to untreated animals (p > 0.05) and DSS treatment decreased these measures further (p < 0.05, Fig. 3a). TCA supplementation during DSS treatment prevented the DSS-induced loss in microbiome diversity, making its measure more similar to the TCA only treated group (Fig. 3a, p < 0.05). The PCoA analysis results showed that the experimental groups had significantly different microbiome profiles from each other with the DSS and non-DSS groups being distinctly different. Interestingly, TCA supplementation during DSS treatment shifted the microbiome to become more similar to the non-DSS animals, indicating some recovery of the microbiome structure (Fig. 3b, as indicated by PERMANOVA testing p < 0.05). Analysis of taxa changes at the phylum level between DSS and DSS-TCA treatments showed a reduction in the relative abundance of Firmicutes_A and Bacteroidota, and an increase in the relative abundance of Proteobacteria in the DSS treatment (Figure S2a). At the genus level, the relative abundance of Akkermansia was increased in the DSS-TCA treatment compared to DSS treatment in both the cecum and feces, but this did not reach statistical significance (p = 0.28, Figure S2b). TCA supplementation significantly diminished the DSS-induced loss in the relative abundance of Lachnospiraceae family (p < 0.05, Fig. 3c), a group known to metabolize BAs and produce secondary BAs such as deoxycholate (DCA).
DSS and TCA treatment alter conjugated and secondary BA profiles in Baat−/− KO mice
We used untargeted metabolomics to profile changes in the metabolome under TCA supplementation in the DSS Baat−/− KO mouse experiment. Although TCA supplementation had no significant effect on overall metabolite structure of the liver or serum (Fig. 3d, e, as indicated by PERMANOVA analysis p > 0.05), it had a pronounced impact on cecum and fecal metabolomes (Fig. 3e, f, as indicated by PERMANOVA analysis p < 0.05). Interestingly, the beta diversity results of the metabolomes showed that the TCA supplemented groups were more similar to each other regardless of DSS treatment, though the effect of DSS was also strong (Fig. 3f, g).
As expected, supplementation of the control and DSS treated animal’s diet with TCA significantly increased its abundance in liver, serum, and cecum (p < 0.05), but not in the fecal samples (p > 0.05, Fig. 3h). Similar to reports in human IBD literature [8], the induction of gut inflammation by DSS alone increased TCA abundance in feces (Fig. 3h, p > 0.05). The other primary conjugated BA glycocholic acid (GCA), only detected in cecum and feces, was also higher in samples from the DSS treatment group compared to the combined DSS-TCA treatment or the non-DSS treatments (Fig. 3h, p < 0.05). Accordingly, the abundance of CA was markedly increased by TCA treatment in the liver, serum, cecum, and feces of both control and DSS treated animals (Fig. 3h, p < 0.05). TCA treatment also increased the abundance of secondary BAs produced by the microbiome including DCA in serum, cecum, and feces (Fig. 3h) and the oxidized secondary BAs (oxoCAs) in cecum and feces (Fig. 3h, p < 0.05). This indicated that TCA was being hydrolyzed by the intestinal microbiome in the colon to generate CA, DCA and oxoCAs, though other aspects of BA metabolism and signaling may contribute to increased CA. This finding contrasted that from the human IBD literature, where there is abundant TCA in those with IBD but secondary BAs, such as DCA, are not increased [8].
Due to the growing diversity of BA conjugation biochemistry from the host and its microbiome [18, 26], we investigated differences in the abundances of some of the novel conjugated BAs in response to DSS and TCA treatments. The abundance of the host produced CA-methylcysteamine (CA-MCY), CA-methylcysteamine sulfoxide (CA-MCYO) and CA-methylcysteamine sulfodioxide (CA-MCYO2) was generally increased in the inflamed gut of mice on DSS, with statistical significance found in the fecal samples (HC group compared to DSS group p < 0.05, Fig. 3h). TCA supplementation subsequently reduced the levels of these unique cysteamine-conjugated BAs particularly CA-MCYO and CA-MCYO2 in the feces (p < 0.05, Fig. 3h). MCBAs, including CysCA, GluCA and LysCA, were also significantly higher in the feces of mice on DSS treatment (p < 0.05, Fig. 3h), another trend matching that observed in people with IBD [7, 20]. TCA decreased MCBAs under DSS-induced colitis, though this did not reach statistical significance between the DSS and DSS-TCA groups.
Highly variable hydrolytic activity of gut bacteria on conjugated BAs in vitro and in vivo
Collectively, our multi-omics profiling of Baat−/− KO mice pointed to the importance of diverse conjugated BAs in promoting health in the inflamed mammalian gut; however, there was also evidence for extensive hydrolysis of the supplemented TCA and production of various secondary BAs by the microbiome, which could compromise the beneficial effects. Therefore, to better understand the relationships between altered conjugated and secondary BA profiles and microbiome structure in a model of GI inflammation, we explored the BSH hydrolysis activity among diverse gut bacteria on diverse conjugated BAs. A total of 17 gut bacterial strains belonging to 9 different genera (strain list in supplementary Table S2) and a fresh community of gut microbes from mouse fecal samples were evaluated for their ability to hydrolyze TCA, GCA and varied MCBAs in vitro. Genome sequence searching determined that 14 of these strains contained an annotated bsh gene and 3 (Clostridium hylemonae, Clostridium scindens and Peptostreptococcus anaerobius) did not. The strains and fecal community were separately cultured for 48 h with each of the 10 conjugated BAs at 1 mM concentration then analyzed for hydrolysis by UPLC-MS/MS. We observed an overall low level of conjugate hydrolysis, which expectedly, included those strains without a known bsh gene (Fig. 4a). However, C. scindens was able to hydrolyze diverse conjugated BAs (particularly those with small amino acid conjugates GCA, SerCA and AlaCA), even though it does not have a known bsh enzyme in its genome. This is similar to its MCBA production activity as reported in [18]. Among bacteria containing the bsh gene, we observed variable levels of hydrolysis that depended on the amino acid conjugate. Enterococcus faecalis, Lactiplantibacillus plantarum, Clostridium perfringens and Enterocloster bolteae showed robust hydrolysis of the conjugates, but with some limition in their ability to hydrolyze GluCA and ThrCA. Bacteroides fragilis exhibited a strong propensity for the host-produced TCA, GCA, with some ability to hydrolyze the MCBAs SerCA and AlaCA. In addition, in vitro culture of a mouse fecal community also showed strong hydrolysis of TCA, SerCA, TyrCA, and AlaCA, but GluCA, ThrCA, and LeuCA were more recalcitrant (Figure S4). Ruminococcus gnavus (newly Mediterraneibacter gnavus) and Enterocloster clostridioformis, which were enriched in CD patients (see below), exhibited a weak ability to hydrolyze conjugated BAs. Overall, these in vitro experiments showed that that TCA, GCA, SerCA and AlaCA were more prone to hydrolysis whereas GluCA, and ThrCA were more resistant. These varied hydrolytic capacities of different gut bacteria may explain both observations in our murine experiments, that linked an altered gut microbiome and secondary BA production in the inflamed murine gut, and evidence in the literature, showing altered conjugated BA profiles from the host and its microbiome in patients with IBD [7].
Given that GluCA was more resistant to hydrolysis than TCA by bacteria in vitro, as well as its markedly enriched in CD patients [7], we further compared the differences in the effects of TCA and GluCA supplementation on BA hydrolysis in DSS induced colitis model. Wild type C57BL/6J mice in DSS-TCA and DSS-GluCA groups mice were fed peanut butter pellets infused with 50 mg/kg of TCA or GluCA every day for 17 days. On day 10, mice in the DSS groups were treated with 2.5% DSS for 7 days and then all animals were humanely euthanized on day 17 (Fig. 4b). In this C57BL/6J wild type background, there was no significant difference among the DSS, DSS-TCA and DSS-GluCA groups in terms of disease signs including weight loss, colon length or spleen index (Figure S5a-c, p > 0.05). Metabolomics analysis showed that TCA significantly increased the abundance of CA and other secondary BAs (oxo-BAs), but GluCA treatment did not (Fig. 4c-d, p < 0.05). DCA showed a similar trend, but this did not reach statistical significance. This suggests that TCA was more readily hydrolyzed by the gut microbiome to produce secondary BAs than GluCA treatment, mirroring the effects shown from bacterial cultures in vitro.
BSHs characterization in IBD
Our in vivo and in vitro data show that conjugate BAs, particularly TCA, are important for reducing pathology in the inflamed murine gut, but the abundance of both conjugate BAs and secondary BAs are dictated by the activity of BSH enzymes from the gut microbiome, which have highly varied activity depending on the gene sequence and BA conjugate encountered. Therefore, we mined the bsh gene sequences and abundance from 774 samples of 6 publicly available metagenomics datasets (Table S3) for comparison among disease types. Subjects with either Crohn’s or ulcerative colitis had lower Shannon diversity indices and observed sequences of BSHs than healthy individuals (Fig. 5a, b, p < 0.05). The alpha-diversity of BSHs was significantly lower in CD and UC patients than in healthy controls (Figure. 5a, b), but there was some variation depending on the cohort analyzed (Figure S6a, b). Beta-diversity of BSH profiles was also different between IBD patients and healthy controls (as indicated by PERMANOVA analysis of the collective cohorts, p < 0.05), with some variation within cohorts (Figure S7). Furthermore, the healthy controls exhibited the highest cumulative relative abundance of BSHs, while the CD patients exhibited the lowest (Fig. 5d; Figure S8). Our results suggest reduced BSHs diversity and abundance in IBD patients, especially those with CD, and the collective sequence space of BSH genes in the microbiome are unique among IBD subtypes.
A BSH phylogenetic tree was built using the amino sequences of the 244 identified BSHs (Figure S9) analyzed for enrichment in particular bacterial BSH sequences. The BSHs were assigned to 76 genera from 4 phyla, including Actinomycetota, Bacillota, Euryarchaeota, and Pseudomonadota (Fig. 5e). The majority of BSHs belonged to the Lachnospiraceae, Oscillospiraceae, and Clostridiaceae families (Fig. 5f), and the Blautia, Clostridium, Ruminococcus, Gemmiger, Roseburia, and Eubacterium genera (Fig. 5g). The abundance changes of BSHs in patients with CD (n = 207) and UC (n = 186) compared to healthy controls (n = 381) were tested via Wilcox rank-sum test followed by BH correction. The results showed that there were 218 and 159 BSHs significantly (p < 0.05) changed in CD and UC patients, respectively, compared to healthy controls. Among these, the proportion of BSHs with increased relative abundance in patients with CD and UC was 14.68% (32/218) and 29.56% (47/159), respectively. This reflected that the relative abundance of BSHs was mostly decreased in the patients with IBD. The BSHs from Lachnospiraceae were significantly decreased in patients with CD compared to healthy controls (Fig. 5h, p < 0.05). Additionally, the BSHs from unclassified Clostridium, unclassified Bacillota and Boudabousia were significantly decreased in the CD patients compared to the healthy controls across the 6 IBD cohorts (p < 0.05). Notably, the relative abundance of the BSH gene of an unknown Ruminococcus spp. was significantly reduced in CD patients (Fig. 5i), however, Ruminococcus gnavus BSH (found in all cohorts except for HMP and PRJNA42990) was significantly increased (Fig. 5j, p < 0.05). The gene from the Lachnospiraceae member Enterocloster clostridioformis was significantly also increased in LLdeep, PRISM and PRJEB1220 cohorts compared to the compared to healthy controls (Fig. 5k, p < 0.05). Collectively, this analysis of data from subjects with IBD shows the varied BSH sequence space between diseased and healthy subjects maybe responsible for the altered conjugated BA profiles which have been reported in the IBD literature [8, 23, 24].