Abstract
The farnesoid X receptor (FXR), expressed in the liver and in the small intestine, is a key regulator of glucose and lipid metabolism. Its pharmacological modulation is explored as a potential treatment for obesity-related metabolic impairments. To develop effective pharmacological interventions, it is crucial to differentiate the individual contributions of intestinal and hepatic FXR to lipid metabolism. This study aimed to evaluate the impact of intestinal FXR ablation on gut microbiome composition and metabolic potential in high-fat diet (HFD)-fed mice. Additionally, we determined the genotype-specific effects of xanthohumol, a hop-derived ligand of FXR, known to mitigate metabolic dysfunction in HFD-fed mice. Intestinal FXR knockout prevented diet-induced obesity, a phenotype that correlated with a decrease in the predicted functional capacity of the gut microbiome. Intestinal FXR deficiency resulted in increased abundances of bacteria producing secondary bile acids, such as Oscillospira, and a decrease in beneficial bacteria, such as Akkermansia, both of which were mitigated by xanthohumol. Our findings provide insights to understand the contribution of intestinal FXR and gut microbiome to metabolic regulation under HFD conditions. We underscore the ability of xanthohumol to restore homeostasis, highlighting its potential to improve gut health.
Introduction
The farnesoid X receptor (FXR, NR1H4) belongs to the nuclear hormone receptors superfamily of proteins functioning as ligand-activated transcription factors to regulate the expression of target genes involved in various physiological processes. [1] FXR and its physiological ligands, bile acids (BAs) play a central role in intestinal absorption of nutrients such as fat-soluble vitamins and lipids, [2,3] the transcriptional regulation of cholesterol metabolism, [4,5] hepatic gluconeogenesis, glycogen synthesis and insulin sensitivity. [5,6] Being a key determinant of glucose and lipid fate in the body, regulating FXR signaling is considered an appealing approach for the treatment of metabolic disorders.
Genetic mouse models of FXR full-body and tissue specific knockout have uncovered the complexity of FXR in the regulation of metabolic dysfunction. [7,8] While liver-specific FXR knockout promotes hepatic lipid accumulation in mice fed a cholesterol-enriched diet [9] or western diet, [10] intestine-specific FXR-deficient (FXRInt-/-) mice do not exhibit this effect. In fact, FXRInt-/- mice have been shown to display resistance to HFD-induced obesity and insulin resistance. [11] These findings were reinforced by tissue-dependent pharmacological modulation of FXR yielding specific effects on metabolism. [12–14] Selective antagonism of intestinal FXR has been demonstrated to improve metabolic phenotypes in obese mice. [13,15] These reports suggest that tissue-specific manipulations of FXR signaling should be exploited to combat obesity-related metabolic disorders. [16,17] However, our current understanding of the functional contribution of intestinal FXR to lipid metabolism remains limited. Some studies indicate that intestinal FXR antagonism reduces the expression of genes involved in ceramide synthesis in the small intestine, an effect mediated by the gut microbiome. [11,13,15] Although there is a substantial body of evidence supporting the involvement of the gut microbiome in lipid accumulation and the onset of obesity, [18] the interplay between FXR signaling, BAs and the gut microbiota is insufficiently explored.
To address this gap, we assessed BA composition and gut microbiome changes in HFD-fed FXRInt-/- and WT mice to uncover alterations in the gut microbiome that could influence phenotypic responses to HFD. Additionally, we evaluate the effects of xanthohumol (XN), a prenylated chalcone isolated from hops (Humulus lupulus) that improves dysfunctional glucose and lipid metabolism in HFD-fed animals. [19,20] XN modulates BA and gut microbiome composition in WT and hepatic FXR knockout mice. [21,22] Our results show that the resistance of FXRInt-/- mice to HFD-induced obesity is linked to disrupted epithelial integrity, alterations in gut microbiome composition and changes in its predicted function. Importantly, all of these effects were prevented by XN supplementation.
Materials and methods
Animal studies
All animal experiments were performed in accordance with institutional and National Health and Medical Research Council guidelines. The experimental protocol was approved by the Institutional Animal Care and Use Committee at Oregon State University and the studies were carried out in accordance with the approved protocol (IACUC 2019-0001). Nine-week-old wild-type (WT) male and female C57BL/6J mice were obtained from Jackson Laboratory (Bar Harbor, ME, USA). FXRInt-/- were generated by crossing FXRFL/FL mice with mice harboring the Cre recombinase under the control of the villin promoter (VillinCre) to produce the VillinCre:FXRFL/FL or FXRInt-/- mice. [7] All mice were bred on a C57BL/6J genetic background for over 12 generations. Mice were housed in groups of 2–3 in ventilated cages under a 12–12-hr light-dark cycle and fed a HFD (Dyets Inc., Bethlehem, PA, USA) containing 60%, 20% and 20% total calories from fat, carbohydrate, and protein, respectively. XN (purity > 99%) from Hopsteiner Inc. (New York, NY, USA) was mixed into the diet as previously described [20] to deliver a dose of 60 mg/kg body weight/day. The control diet contained an identical amount of the vehicle. 15 WT mice (8 females, 7 males) and 12 FXRInt-/- mice (7 females, 5 males) were fed a control HFD. Low-fat diet controls were not included because they do not develop a Metabolic Syndrome (MetS) phenotype characterized by dyslipidemia and hyperglycemia. 15 WT mice (8 females, 7 males) and 13 FXRInt-/- mice (7 females, 6 males) were supplemented with XN for 12 weeks. Food intake and body weights were recorded weekly.
At week 10, glucose tolerance was tested after 6 h fasting. Following intraperitoneal bolus injection of 1.5 g/kg of D-glucose, blood glucose levels were measured at 0, 15, 30, 60, and 120 min using the One Touch UltraMini glucometer (LifeScan Inc., Milpitas, CA, USA). At the end of 12 weeks of feeding, fed-state mice were euthanized by cervical dislocation, their blood collected, and their liver, ileum and cecum were dissected for further analyses. Feces of fed-state mice were collected over two-hour period on the day before organ collection.
Histology
Liver tissues from n = 3 male mice were fixed in 4% paraformaldehyde (PFA), embedded in OCT and 10 µm-thick sections were used for histology. Hematoxylin and Eosin (H&E) and Sudan black staining (lipids) were performed as previously described [23].
16S ribosomal RNA gene sequencing and PICRUSt
Whole genomic DNA was extracted from cecal contents (n = 9 per group) using the QIAamp PowerFecal Pro DNA kit (Qiagen, Hilden, Germany). The V4 hypervariable region of the 16S rRNA gene was amplified using universal bacterial primers previously described. [24] Library preparation was performed as previously described. [25] Amplicons were sequenced on an Illumina MiSeq instrument at the Center for Quantitative Life Sciences at Oregon State University. The resulting V4 16S rRNA forward and reverse sequence reads were quality-controlled and subjected to amplicon sequence variance (ASV) clustering, taxonomic annotation, and phylogenetic reconstruction using the DADA2 workflow. [26] Sequences were annotated using the Greengene database and subsequent analysis of alpha diversity and beta diversity were performed with QIIME 2019.7. [27] Data visualization and statistical tests were performed with QIIME2. Functional profiles of microbial communities were predicted using the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States or PICRUSt2. [28] Metagenome functions were infered using predicted MetaCyc pathways. [29] Spearman correlation analysis was performed in MetaboAnalyst version 6.0.
Fecal bile acids analysis
Fecal droppings (20 mg, n = 12–15 per group) were spiked with 0.25 ng cholic acid-d4 per mg of feces and extracted twice with 1 mL of 100% MeOH using a counter-top bullet blender for 60 min and centrifuged at 13,000 rpm for 10 min. The supernatants were pooled, dried under vacuum, reconstituted in 200 μL of 50% MeOH, and centrifuged again. The supernatant was collected and stored at −80°C until UPLC-MS analysis. Samples were analyzed by UPLC-MS/MS as previously described. [22] The heatmap was generated using MetaboAnalyst version 5.0.
Real-time PCR
RNA (n = 4–7 per group) was isolated from mouse ileum (RNeasy Mini Kit, Qiagen, Hilden, Germany), and gene expression analyses were conducted as previously described. [22] Gene expression was normalized to levels of Polymerase-II. Relative gene expression was calculated using the 2-ddCt method. All primers were purchased from IDT technologies (Coralville, IA, USA) and are listed in Table S2.
Statistical analyses
Differences in metabolic parameters, gene expression and bile acid concentrations among groups were evaluated by one-way ANOVA with post hoc Tukey’s test. Repeated-measures two-way ANOVA with post hoc Tukey’s test was used to test differences in body weight over the duration of the feeding and blood glucose concentrations over the duration of the test. Differences in bacterial abundances between two groups were computed with two-tailed Mann-Whitney U test. Differences in pathway abundances were calculated by one-way ANOVA following logarithmic transformation to normalize the data.
Results
XN mitigates dysfunctional energy metabolism in DIO-resistant FXRInt-/- mice
Nine-week-old WT and FXRInt-/- mice were subjected to a 12-week high-fat diet (HFD) challenge (Fig 1a). At day 0, FXRInt-/- mice had similar body weight to their wildtype (WT) counterparts (S1 Table) and gained less weight over the feeding period than HFD-fed WT mice (Fig 1b). The weight gain difference could not be attributed to variations in food intake (S1 Fig).
Fig 1. Effect of XN on body weight and glucose homeostasis in FXRInt-/- HFD-fed mice.
(a) Schematic representation of experimental timeline of HFD feeding to WT and FXRInt-/- mice. (b) Mean of body weight recorded during 12 weeks of HFD feeding. (c) Blood glucose and (d) area under the curve upon glucose challenge in HFD-fed WT and FXRInt-/- mice at week 10. Values are expressed as mean ± SEM (n = 12-15 mice per group). *p < 0.05 for effect of XN treatment; #p < 0.05 for genotype effect; n.s., nonsignificant; repeated-measures two-way ANOVA (b,c) or one-way ANOVA (d), both with post hoc Tukey’s test.
A glucose tolerance test (GTT) performed after 10 weeks revealed that HFD did not impair glucose metabolism in FXRInt-/- mice (Fig 1c) and the GTT area under the curve was significantly lower in FXRInt-/- mice than WT mice (p = 0.01, Fig 1d). Interestingly, XN-treated FXRInt-/- mice displayed higher blood glucose concentrations compared to untreated HFD-fed FXRInt-/- mice (Fig 1c and 1d). H&E staining shows fewer lipid vacuoles and Sudan Black staining for lipids shows less lipid accumulation in the liver of FXRInt-/- mice compared to WT mice (Fig 2). These data show that FXRInt-/- mice are resistant to HFD-induced changes including weight gain, hepatic lipid accumulation and insulin resistance. Our results suggest that treatment with XN restores FXRInt-/- mouse sensitivity to HFD.
Fig 2. Representative liver histology by H&E (a-d) and Sudan Black (e-h) of male WT and FXRInt-/- mice fed HFD ± XN.

Arrows indicate vacuoles, a characteristic structure of hepatic steatosis. Panels showing liver tissues from WT animals (left column) were reproduced from Paraiso et al. [22] with permission from Frontiers in Pharmacology.
Effects of XN on systemic inflammation, intestinal epithelium integrity/remodeling, and FXR signaling in HFD-fed WT and FXRInt-/- mice
To understand the mechanisms of diet-induced-obesity (DIO)-resistance in FXRInt-/- mice and reversal by XN, we investigated circulatory markers of inflammation, epithelial integrity and FXR signaling. There were no significant changes in circulatory pro-inflammatory cytokines such as monocyte chemoattractant protein-1 (MCP1) and interleukin-6 (IL6) in FXRInt-/- mice compared to WT mice (S1 Table).
The repression of intestinal FXR signaling has been associated with pathological exacerbation of intestinal inflammation in mice [30,31] and inflammatory bowel diseases in patients, [32] hence we assessed the integrity of the intestinal epithelium in HFD-fed FXRInt-/- mice. Considering the interplay between FXR signaling, BAs, and inflammation, we investigated genes involved in intestinal BA handling (Fig 3a). In WT mice, the decrease in small heterodimer partner-1 (Shp1) and the increase in apical sodium-BA transporter (Asbt) gene expression were consistent with intestinal FXR antagonism by XN (Fig 3b). In untreated FXRInt-/- mice, expression of genes encoding ASBT, and the ileal BA-binding protein (I-BABP) involved in the transport of BAs from the intestinal lumen to the portal circulation were increased. However, there were no changes in the organic solute transporter Ost-α gene expression (S2 Fig), which may result in the accumulation of BAs within the enterocytes of FXRInt-/- mice. XN treatment had no effect on FXR downstream targets in FXRInt-/- mice, i.e., SHP1, ASBT, and I-BABP (Fig 3b and 3c), indicating FXR-dependent signaling in WT mice.
Fig 3. Effects of intestinal FXR knockout and XN treatment on FXR signaling, markers of inflammation and mucosa remodeling.
(a) Schematic representation of FXR signaling in enterocytes. (b) Relative expression of FXR target genes in the ileum of HFD-fed WT and FXRInt-/- mice. (c) Relative expression of genetic markers of inflammation (Tnfα) and mucosal remodeling (metalloproteinases Mmp3/9, and mucin (Muc-2) in the ileum of HFD-fed WT and FXRInt-/- mice. Values are expressed as mean ± SEM (n = 4-7 per group). *p < 0.05 for effect of XN treatment, #p < 0.05 for genotype effect, one-way ANOVA with post hoc Tukey’s test.
We investigated whether genotype and treatment correlated with expression in genes driving epithelial and matrix remodeling in the intestine. Genes encoding for tumor necrosis factor-α (TNFα), matrix metalloproteinases MMP3 and MMP9 involved in inflammatory processes were decreased in XN-treated WT mice, but not in FXRInt-/- mice (Fig 3c). On the other hand, gene encoding for mucin-2 (MUC2), the main component of mucus trended towards an increase in XN-treated WT mice (p = 0.06). We observed no changes in expression of genes encoding tight junction membrane proteins (S2 Fig). Together, these data indicate that, while FXR signaling may contribute to the effect of XN on intestinal homeostasis in WT mice, other mechanisms are involved in FXRInt-/- mice.
XN regulates fecal BA homeostasis in FXRInt-/- mice
Considering the role of FXR signaling in the regulation of BAs, we sought to identify correlations between the phenotype and changes in BAs composition. Using a targeted quantitative UPLC-MS/MS approach, we screened for 34 fecal BAs and quantified 29 unconjugated, taurine conjugated, and glycine conjugated BAs (Fig 4a). Total fecal BA concentrations were slightly increased in HFD-fed FXRInt-/- mice (Fig 4b), an increase driven by secondary unconjugated BAs (p = 0.01). Secondary BAs such as DCA (Fig 4c) and derivatives, NDCA (Fig 3d), HDCA (Fig 4e) were increased in FXRInt-/- mice. Secondary BAs 12-KCDCA (Fig 3f) and 7-KCDCA (Fig 4g), products from the microbial transformation of CDCA in the GI tract were also increased in mutant mice. XN supplementation in FXRInt-/- mice resulted in a consistent decrease in these secondary BAs (Fig 4b–4g). LCA concentrations were not affected in FXRInt-/- mice and were decreased by XN treatment regardless of the genotype (Fig 4h). In summary, the metabolic profiling data revealed that accumulation of BAs in FXRInt-/- mice was orchestrated by secondary unconjugated BAs and prevented by XN treatment.
Fig 4. Effect of XN on fecal BAs in HFD-fed WT and FXRInt-/- mice.
Heatmaps of individual BAs (a), total BAs and their classes (b), DCA (c), NDCA (d), HDCA (e), 12-KCDCA (f), 7-KCDCA (g), and LCA (h) in the feces of HFD-fed WT and FXRInt-/- mice. Values are expressed as mean ± SEM (n = 12-15 per group). *p < 0.05 for effect of XN treatment, #p < 0.05 for genotype effect, one-way ANOVA with post hoc Tukey’s test. Abbreviations: allocholic acid (ACA), chenodeoxycholic acid (CDCA), coprocholic acid (CCA), cholic acid (CA), deoxycholic acid (DCA), glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), glycodeoxycholic acid (GDCA), glycolithocholic acid (GLCA), glycoursodeoxycholic acid (GUDCA), hyodeoxycholic acid (HDCA), ketochenodeoxycholic acid (KCDCA), ketodeoxycholic acid (KDCA), lithocholic acid (LCA), muricholic acid (MCA), nordeoxycholic acid (NDCA), taurochenodeoxycholic acid (TCDCA), taurocholic acid (TCA), tauromuricholic acid (T-MCA), taurodeoxycholic acid (TDCA), taurolithocholic acid (TLCA), tauroursodeoxycholic acid (TUDCA), ursocholic acid (UCA).
XN modulates gut microbiome composition in WT and FXRInt-/- mice
To ascertain whether changes in microbiome composition factored in the observed metabolic differences between WT and FXRInt-/- mice, we performed 16S rRNA sequencing on the cecal contents of these mice. Sequencing depth averaged 55,000 reads across samples and saturation was reached at 20,000 reads as observed in the rarefaction curve (S3 Fig). Alpha diversity measured by Chao1 richness index was higher in FXRInt-/- mice compared to WT and in female compared to male mice (Fig 5a). However, sex differences were not noted in beta-diversity taxonomy analyses (Fig 5b), and Bray-Curtis distance PCoA plots (S3 Fig). XN decreased the abundance of Firmicutes and increased Bacteroidetes, Verrucomicrobia and Proteobacteria phyla, regardless of the genotype (Fig 5c). While most differences at the phylum level were XN-induced, Verrucomicrobia and Proteobacteria abundances were affected in FXRInt-/- mice. Principal component analysis (PCA) at the family level showed distinct separation between genotypes (Fig 5d), diets (Fig 5e) and confirmed the lack of differences between sexes (Fig 5f). Particularly noteworthy was the elevated abundance of Ruminococcaceae in FXRInt-/- mice (S4 Fig), as species from this family are known metabolizers of primary BAs into secondary BAs. [33]
Fig 5. Effect of XN on gut microbiota composition in HFD-fed WT and FXRInt-/- mice.
(a) Alpha diversity Chao1 index, (b) Taxonomy at the phylum level in HFD-fed WT and FXRInt-/- mice. (c) Relative abundances of Firmicutes, Bacteroidetes, Verrucomicrobia and Proteobacteria phyla in HFD-fed WT and FXRInt-/- mice. Principal component analysis (PCA) of taxonomy at the family level comparing (d) genotype, (e) diet and (f) sex. Values are expressed as mean ± SEM (n = 9 per group). *p < 0.05, **p < 0.01, ***p < 0.001 for XN effect, #p < 0.05, ##p < 0.01 for genotype effect, two-tailed Mann-Whitney U test.
To identify associations between genotype, gut microbiome composition and metabolic profiles, we assessed the correlation between annotated genera and fecal BAs. XN treatment led to pronounced and diversified effect on genera from various phyla including Actinobacteria (Adlercreutzia), Bacteroidetes (Parabacteroides), Deferribacteres (Mucispirillum), Firmicutes (Oscillospira, Ruminococcus, Blautia, Dehalobacterium), Proteobacteria (Desulfovibrio, Sutterella) and Verrucomicrobia (Akkermansia) (Fig 6a–6h). Genera from the Firmicutes (Oscillospira, Dorea, Streptococcus, Coprococcus) and Bacteroidetes phyla (AF12, Odoribacter, Alistipes) were increased in FXRInt-/- mice, indicating a potential correlation with secondary BA synthesis.
Fig 6. Abundances of bacterial genera and their correlation to fecal BAs in HFD-fed WT and FXRInt-/- mice.
Relative abundances of (a) Adlercreutzia, (b) Parabacteroides, (c) Mucispirillum, (d) Coprococcus, (e) Oscillospira, (f) Ruminococcus, (g) Desulfovibrio, (h) Akkermansia genera in HFD-fed WT and FXRInt-/- mice. (i) Heatmap of correlation between fecal BA classes and gut microbial composition. Color coding reflects the R values of the spearman correlation. Relative abundances are expressed as mean ± SEM (n = 9 per group). *p < 0.05, **p < 0.01, ***p < 0.001 for XN effect, #p < 0.05, ##p < 0.01, ###p < 0.001 for genotype effect, two-tailed Mann-Whitney U test. In the correlation heatmap, black asterisks indicate positive correlations and white asterisks indicate negative correlations *p < 0.05, **p < 0.01, ***p < 0.001. Annotations: [Ruminococcus] (Lachnospiraceae family); Ruminococcus (Ruminococcaceae family).
The correlation analysis between bacterial genera and BAs showed that 14 out of 32 annotated genera were associated with secondary unconjugated BAs concentrations (Fig 6i). Secondary unconjugated BAs are of particular interest because their physiochemical and biological properties have linked them to inflammatory and carcinogenic processes. [34,35] We identified two main bacterial clusters with potential metabolic implications. The first cluster consists of genera positively correlated with secondary unconjugated BAs, including Adlercreutzia, AF12, Odoribacter, Mucispirillum, Streptococcus, Dehalobacterium, Oscillospira, Ruminococcus and Bilophila. These genera were found to be increased in FXRInt-/- mice and/or decreased by XN, suggesting a negative impact on gut integrity or metabolic phenotype. The second cluster comprises genera negatively correlated with secondary unconjugated BAs, including Parabacteroides, Coprobacillus, Sutterella, Desulfovibrio and Akkermansia. These genera were found to be decreased in FXRInt-/- mice and/or increased by XN treatment suggesting a beneficial effect on the metabolic phenotype. Altogether, our results reveal associations between secondary unconjugated BAs and gut bacteria that may determine the metabolic outcomes of HFD feeding in WT and FXRInt-/- mice.
XN reverses functional changes in the gut microbiota of FXRInt-/- mice
To understand the implications of the changes in gut microbiome composition induced by genotype and diet, we projected the metabolic potential of gut microbiota. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) was performed on the 16S rRNA gene abundance data to predict differentially enriched KEGG orthologs (KOs) and pathways. [36]
We identified 366 metabolic pathways, 49 of which were differentially regulated by XN in WT mice, 36 significantly increased and 13 significantly decreased in XN-treated mice (Fig 7a). The top differentially enriched KO pathways were biosynthesis pathways such as cofactor, carrier and vitamin biosynthesis pathways and amino acid biosynthesis pathways. Pathways depleted by XN included carbohydrate biosynthesis and nucleotide degradation pathways. Conversely, bacterial metabolic capacity was consistently decreased in FXRInt-/- mice; out of 31 differentially expressed pathways, 30 were decreased in FXRInt-/- mice compared to WT mice. XN supplementation increased the metabolic potential of FXRInt-/- mice, with 95 increased pathways out of 96 differentially expressed pathways. KO pathways differentially enriched by XN in FXRInt-/- mice notably included pathways involved in the generation of precursor metabolites and the processes facilitating energy release from these metabolites. These data suggest that lack of FXR in the intestine may lead to dysbiosis and decreased functional capabilities of the gut microbiome. XN regulated gut microbiome composition and improved gut metabolic potential in WT and FXRInt-/- mice.
Fig 7. Functional changes in the gut microbiota of HFD-fed WT and FXRInt-/- mice.

Heatmap of microbial metabolic pathways differentially modified in WT and FXRInt-/- mice (MetaCyc annotations). Significance was assessed by one-way ANOVA of log-normalized abundances, p < 0.05 were considered to indicate statistical significance.
Discussion
Independent studies have established the role of FXR signaling and the gut microbiota in the onset of obesity, thus, warranting the investigation of the interplay between intestinal FXR and the gut microbiome composition. Here, we combine 16S rRNA gene sequencing of the gut microbiome and targeted metabolomics to examine changes in the gut microbiota and BAs following intestinal FXR knockout, HFD and XN treatment.
Our findings revealed that resistance to HFD-induced obesity and glucose intolerance in FXRInt-/- mice were associated to gut epithelial function and changes in the gut microbiome composition. These results align with previous reports highlighting the role of FXR in preventing dysbiosis and preserving the integrity of the small intestine epithelial barrier. [37] The intestinal epithelium is protected by a thick layer of mucus that plays structural and functional roles. [38] The homeostasis of the mucus layer is influenced by transcriptional and epigenetic regulation of MUC2, which can be mediated by gut bacteria and microbial metabolites such as bile acids. [39–41] Secondary and unconjugated BAs, in particular, being more hydrophobic than their primary and conjugated forms, are more likely to cause toxicity [42] and impact gut barrier function and integrity. In FXRInt-/- mice, alterations in gut microbiome composition, coupled with the accumulation of secondary unconjugated BAs, could impact the structural integrity of the intestinal epithelium. These combined changes likely impede the absorption of nutrients, contributing to the observed resistance to HFD-induced obesity in FXRInt-/- mice.
XN improved the structural integrity of the intestinal epithelium, modulated microbial imbalance, and regulated glucose metabolism in DIO-resistant FXRInt-/- mice. In WT mice, XN treatment inhibited intestinal FXR signaling and downregulated the expression of pro-inflammatory genes such as Tnfα, Mmp3 and Mmp9. These markers can be regulated by FXR [43,44] and MMP9 was shown to directly influence intestinal mucosal barrier by inhibiting MUC2 expression in mice. [45] Our data suggests that inhibition of intestinal FXR by XN downregulates inflammation, thereby preserving epithelial integrity in HFD-fed WT mice. In FXRInt-/- mice, however, XN-related improvements were independent of intestinal FXR signaling and could be attributed to XN-mediated changes in the gut microbiota.
The gut microbiome affects host metabolism and modulates inflammation contributing to the susceptibility to metabolic syndrome (MetS) and obesity. [46] Additionally, gut bacteria play an active role in the conversion of primary BAs into secondary BAs via 7α-dehydroxylation. The BA-inducible (bai) gene cluster encoding for 7α-dehydroxylating enzymes is predominantly associated with Ruminococcaceae isolates. [33,47] Our data revealed that the increase in the Ruminococcaceae in FXRInt-/- mice was driven by the Oscillospira genus, which also showed a positive correlation with fecal secondary unconjugated BAs. Clinical studies have associated Oscillospira to lower BMI [48,49] and elevated secondary fecal BAs in gallstone patients. [50] By breaking down gut mucin glycans and prompting the host to replenish the degraded glycans, Oscillospira are believed to increase energy expenditure by the host, hence the correlation with leanness. [51] XN specifically reduced the abundance of Oscillospira in FXRInt-/- mice, supporting that, in addition to regulating secondary BAs, Oscillospira may play a critical role in the phenotype of resistance to DIO.
Conversely, genera such as Akkermansia and Parabacteroides were reduced in FXRInt-/- mice and exhibited a negative correlation with secondary unconjugated BAs. Species from Akkermansia and Parabacteroides genera have been associated with obesity-related metabolic improvements, enhanced intestinal integrity and decreased inflammation. [52,53] Consistent with previous studies in Swiss Webster mice, [54] our results demonstrated that XN supplementation leads to an enrichment of Akkermansia and Paracteroides in WT and FXRInt-/- mice. These findings provide evidence that XN increases the abundance of health-promoting bacteria, regardless of genetic background and metabolic status, suggesting its use as a bioactive compound in the prevention of metabolic diseases.
The gut microbiome is a key determinant of host metabolic markers [55,56] and complex metabolic phenotypes have been shown to be transmitted through fecal microbiota transplants. [57,58] However, research indicates that the gut metabolic potential has a stronger impact on the metabolome than microbial species themselves. [59] Moreover, when the physiological condition of the host is altered, bacteria can adapt their functional pathways without major modifications in the microbiota composition. [60] Therefore, assessing functional capabilities of the microbiome could more accurately predict phenotypic outcomes. Our findings indicate a widespread decrease in gut metabolic potential in the absence of intestinal FXR, further supporting that FXRInt-/- mice may be resistant to HFD-induced obesity due to reduced energy harvest from the diet by gut bacteria. In XN-treated mice, the upregulation of biosynthetic pathways may promote the synthesis of metabolites necessary for microbial survival such as biotin and heme [61,62] and the formation of precursor metabolites necessary for energy production. XN treatment predominantly improved microbial metabolic function in FXRInt-/- mice, whereas XN-induced changes in gut bacteria were comparable in WT and FXRInt-/- mice. This observation underscores that alterations in gut microbiome composition alone do not reliably mirror metabolic outcomes.
Some limitations of the study include the following. This is mostly a correlational study in the absence of mechanistic experiments. The use of 16S rRNA gene amplicon sequencing instead of the more comprehensive whole metagenomic shotgun sequencing to assess the metabolic activity of the microbiome. However, data from this study is in accordance with emerging reports that have linked FXR and the gut microbiome to health and disease, highlighting the role of both in the XN-mediated amelioration of metabolic dysfunction.
Supporting information
Means ± SEM represent per-cage averages of 2–3 animals per cage.
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(XLSX)
Acknowledgments
The authors thank Grace L. Guo (Rutgers University) for providing the FXRInt-/- mice and Cristobal Miranda (Linus Pauling Institute, Oregon State University) for providing technical guidance. The bile acid measurements were conducted in the Oregon State University Mass Spectrometry Center with assistance from Jeffrey Morré.
Data Availability
Sequencing data have been deposited in NCBI Sequence Read Archive (SRA) under the BioProject number PRJNA1146151.
Funding Statement
This work was supported by the National Institutes of Health under Grants S10RR022589 and R01AT009168-04S1, the Linus Pauling Institute, the OSU College of Pharmacy, and the OSU Foundation Buhler-Wang Research Fund.
References
- 1.Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: opening the X-files. Science. 2001;294(5548):1866–70. doi: 10.1126/science.294.5548.1866 [DOI] [PubMed] [Google Scholar]
- 2.Fan M, Wang X, Xu G, Yan Q, Huang W. Bile acid signaling and liver regeneration. Biochim Biophys Acta. 2015;1849(2):196–200. doi: 10.1016/j.bbagrm.2014.05.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ali AH, Carey EJ, Lindor KD. Recent advances in the development of farnesoid X receptor agonists. Ann Transl Med. 2015;3(1):5. doi: 10.3978/j.issn.2305-5839.2014.12.06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chiang JYL. Bile acid regulation of gene expression: roles of nuclear hormone receptors. Endocr Rev. 2002;23(4):443–63. doi: 10.1210/er.2000-0035 [DOI] [PubMed] [Google Scholar]
- 5.Trauner M, Claudel T, Fickert P, Moustafa T, Wagner M. Bile acids as regulators of hepatic lipid and glucose metabolism. Dig Dis. 2010;28(1):220–4. doi: 10.1159/000282091 [DOI] [PubMed] [Google Scholar]
- 6.Ma K, Saha PK, Chan L, Moore DD. Farnesoid X receptor is essential for normal glucose homeostasis. J Clin Invest. 2006;116(4):1102–9. doi: 10.1172/JCI25604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kim I, Ahn S-H, Inagaki T, Choi M, Ito S, Guo GL, et al. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res. 2007;48(12):2664–72. doi: 10.1194/jlr.M700330-JLR200 [DOI] [PubMed] [Google Scholar]
- 8.Parséus A, Sommer N, Sommer F, Caesar R, Molinaro A, Ståhlman M, et al. Microbiota-induced obesity requires farnesoid X receptor. Gut. 2017;66(3):429–37. doi: 10.1136/gutjnl-2015-310283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Schmitt J, Kong B, Stieger B, Tschopp O, Schultze SM, Rau M, et al. Protective effects of farnesoid X receptor (FXR) on hepatic lipid accumulation are mediated by hepatic FXR and independent of intestinal FGF15 signal. Liver Int. 2015;35(4):1133–44. doi: 10.1111/liv.12456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Clifford BL, Sedgeman LR, Williams KJ, Morand P, Cheng A, Jarrett KE, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metabolism. 2021;33(8):1671-1684.e4. doi: 10.1016/j.cmet.2021.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li F, Jiang C, Krausz KW, Li Y, Albert I, Hao H, et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun. 2013;4(1):2384. doi: 10.1038/ncomms3384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kong B, Wang L, Chiang JYL, Zhang Y, Klaassen CD, Guo GL. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology. 2012;56(3):1034–43. doi: 10.1002/hep.25740 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat Commun. 2015;6:10166. doi: 10.1038/ncomms10166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fang S, Suh JM, Reilly SM, Yu E, Osborn O, Lackey D, et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med. 2015;21(2):159–65. doi: 10.1038/nm.3760 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jiang C, Xie C, Li F, Zhang L, Nichols RG, Krausz KW, et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest. 2015;125(1):386–402. doi: 10.1172/JCI76738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Shyng S-L. Targeting the Gut Microbiota-FXR Signaling Axis for Glycemic Control: Does a Dietary Supplement Work Magic? Diabetes. 2017;66(3):571–3. doi: 10.2337/dbi16-0066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sun L, Cai J, Gonzalez FJ. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat Rev Gastroenterol Hepatol. 2021;18(5):335–47. doi: 10.1038/s41575-020-00404-2 [DOI] [PubMed] [Google Scholar]
- 18.Carmody RN, Bisanz JE. Roles of the gut microbiome in weight management. Nat Rev Microbiol. 2023;21(8):535–50. doi: 10.1038/s41579-023-00888-0 [DOI] [PubMed] [Google Scholar]
- 19.Miranda CL, Elias VD, Hay JJ, Choi J, Reed RL, Stevens JF. Xanthohumol improves dysfunctional glucose and lipid metabolism in diet-induced obese C57BL/6J mice. Arch Biochem Biophys. 2016;599:22–30. doi: 10.1016/j.abb.2016.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Miranda CL, Johnson LA, de Montgolfier O, Elias VD, Ullrich LS, Hay JJ, et al. Non-estrogenic xanthohumol derivatives mitigate insulin resistance and cognitive impairment in high-fat diet-induced obese mice. Sci Rep. 2018;8(1):613. doi: 10.1038/s41598-017-18992-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhang Y, Bobe G, Revel JS, Rodrigues RR, Sharpton TJ, Fantacone ML, et al. Improvements in metabolic syndrome by xanthohumol derivatives are linked to altered gut microbiota and bile acid metabolism. Mol Nutr Food Res. 2020;64(1):e1900789. doi: 10.1002/mnfr.201900789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Paraiso IL, Tran TQ, Magana AA, Kundu P, Choi J, Maier CS, et al. Xanthohumol ameliorates diet-induced liver dysfunction via farnesoid X receptor-dependent and independent signaling. Front Pharmacol. 2021;12:643857. doi: 10.3389/fphar.2021.643857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Chang C-N, Singh AJ, Gross MK, Kioussi C. Requirement of Pitx2 for skeletal muscle homeostasis. Dev Biol. 2019;445(1):90–102. doi: 10.1016/j.ydbio.2018.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A. 2011;108 Suppl 1(Suppl 1):4516–22. doi: 10.1073/pnas.1000080107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gaulke CA, Barton CL, Proffitt S, Tanguay RL, Sharpton TJ. Triclosan exposure is associated with rapid restructuring of the microbiome in adult zebrafish. PLoS One. 2016;11(5):e0154632. doi: 10.1371/journal.pone.0154632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–3. doi: 10.1038/nmeth.3869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37(8):852–7. doi: 10.1038/s41587-019-0209-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Douglas GM, Maffei VJ, Zaneveld J, Yurgel SN, Brown JR, Taylor CM. PICRUSt2: An improved and customizable approach for metagenome inference. BioRxiv. 2020. doi: 10.1101/672295 [DOI] [Google Scholar]
- 29.Caspi R, Billington R, Fulcher CA, Keseler IM, Kothari A, Krummenacker M, et al. The MetaCyc database of metabolic pathways and enzymes. Nucleic Acids Res. 2018;46(D1):D633–9. doi: 10.1093/nar/gkx935 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhou X, Cao L, Jiang C, Xie Y, Cheng X, Krausz KW, et al. PPARα-UGT axis activation represses intestinal FXR-FGF15 feedback signalling and exacerbates experimental colitis. Nat Commun. 2014;5:4573. doi: 10.1038/ncomms5573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fu T, Li Y, Oh TG, Cayabyab F, He N, Tang Q, et al. FXR mediates ILC-intrinsic responses to intestinal inflammation. Proc Natl Acad Sci U S A. 2022;119(51):e2213041119. doi: 10.1073/pnas.2213041119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Nijmeijer RM, Gadaleta RM, van Mil SWC, van Bodegraven AA, Crusius JBA, Dijkstra G, et al. Farnesoid X receptor (FXR) activation and FXR genetic variation in inflammatory bowel disease. PLoS One. 2011;6(8):e23745. doi: 10.1371/journal.pone.0023745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Vital M, Rud T, Rath S, Pieper DH, Schlüter D. Diversity of bacteria exhibiting bile acid-inducible 7α-dehydroxylation genes in the human gut. Comput Struct Biotechnol J. 2019;17:1016–9. doi: 10.1016/j.csbj.2019.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ajouz H, Mukherji D, Shamseddine A. Secondary bile acids: an underrecognized cause of colon cancer. World J Surg Oncol. 2014;12:164. doi: 10.1186/1477-7819-12-164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.McGarr SE, Ridlon JM, Hylemon PB. Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. J Clin Gastroenterol. 2005;39(2):98–109. [PubMed] [Google Scholar]
- 36.Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. 2013;31(9):814–21. doi: 10.1038/nbt.2676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Inagaki T, Moschetta A, Lee Y-K, Peng L, Zhao G, Downes M, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A. 2006;103(10):3920–5. doi: 10.1073/pnas.0509592103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Song C, Chai Z, Chen S, Zhang H, Zhang X, Zhou Y. Intestinal mucus components and secretion mechanisms: what we do and do not know. Exp Mol Med. 2023;55(4):681–91. doi: 10.1038/s12276-023-00960-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ghosh S, Whitley CS, Haribabu B, Jala VR. Regulation of intestinal barrier function by microbial metabolites. Cell Mol Gastroenterol Hepatol. 2021;11(5):1463–82. doi: 10.1016/j.jcmgh.2021.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut. 2020;69(12):2232–43. doi: 10.1136/gutjnl-2020-322260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Martin NA, Mount Patrick SK, Estrada TE, Frisk HA, Rogan DT, Dvorak B, et al. Active transport of bile acids decreases mucin 2 in neonatal ileum: implications for development of necrotizing enterocolitis. PLoS One. 2011;6(12):e27191. doi: 10.1371/journal.pone.0027191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gadaleta RM, Cariello M, Crudele L, Moschetta A. Bile salt hydrolase-competent probiotics in the management of IBD: unlocking the “Bile Acid Code”. Nutrients. 2022;14(15):3212. doi: 10.3390/nu14153212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Liu N, Meng Z, Lou G, Zhou W, Wang X, Zhang Y, et al. Hepatocarcinogenesis in FXR-/- mice mimics human HCC progression that operates through HNF1α regulation of FXR expression. Mol Endocrinol. 2012;26(5):775–85. doi: 10.1210/me.2011-1383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Guan B, Li H, Yang Z, Hoque A, Xu X. Inhibition of farnesoid X receptor controls esophageal cancer cell growth in vitro and in nude mouse xenografts. Cancer. 2013;119(7):1321–9. doi: 10.1002/cncr.27910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Garg P, Ravi A, Patel NR, Roman J, Gewirtz AT, Merlin D, et al. Matrix metalloproteinase-9 regulates MUC-2 expression through its effect on goblet cell differentiation. Gastroenterology. 2007;132(5):1877–89. doi: 10.1053/j.gastro.2007.02.048 [DOI] [PubMed] [Google Scholar]
- 46.Brown EM, Clardy J, Xavier RJ. Gut microbiome lipid metabolism and its impact on host physiology. Cell Host Microbe. 2023;31(2):173–86. doi: 10.1016/j.chom.2023.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rath S, Rud T, Karch A, Pieper DH, Vital M. Pathogenic functions of host microbiota. Microbiome. 2018;6(1). doi: 10.1186/s40168-018-0542-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tims S, Derom C, Jonkers DM, Vlietinck R, Saris WH, Kleerebezem M, et al. Microbiota conservation and BMI signatures in adult monozygotic twins. ISME J. 2013;7(4):707–17. doi: 10.1038/ismej.2012.146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen Y-R, Zheng H-M, Zhang G-X, Chen F-L, Chen L-D, Yang Z-C. High Oscillospira abundance indicates constipation and low BMI in the Guangdong Gut Microbiome Project. Sci Rep. 2020;10(1):9364. doi: 10.1038/s41598-020-66369-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Keren N, Konikoff FM, Paitan Y, Gabay G, Reshef L, Naftali T, et al. Interactions between the intestinal microbiota and bile acids in gallstones patients. Environ Microbiol Rep. 2015;7(6):874–80. doi: 10.1111/1758-2229.12319 [DOI] [PubMed] [Google Scholar]
- 51.Gophna U, Konikoff T, Nielsen HB. Oscillospira and related bacteria - From metagenomic species to metabolic features. Environ Microbiol. 2017;19(3):835–41. doi: 10.1111/1462-2920.13658 [DOI] [PubMed] [Google Scholar]
- 52.Shin J, Noh J-R, Chang D-H, Kim Y-H, Kim MH, Lee ES, et al. Elucidation of Akkermansia muciniphila Probiotic Traits Driven by Mucin Depletion. Front Microbiol. 2019;10:1137. doi: 10.3389/fmicb.2019.01137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wu T-R, Lin C-S, Chang C-J, Lin T-L, Martel J, Ko Y-F, et al. Gut commensal Parabacteroides goldsteinii plays a predominant role in the anti-obesity effects of polysaccharides isolated from Hirsutella sinensis. Gut. 2019;68(2):248–62. doi: 10.1136/gutjnl-2017-315458 [DOI] [PubMed] [Google Scholar]
- 54.Logan IE, Shulzhenko N, Sharpton TJ, Bobe G, Liu K, Nuss S, et al. Xanthohumol requires the intestinal microbiota to improve glucose metabolism in diet-induced obese mice. Mol Nutr Food Res. 2021;65(21):e2100389. doi: 10.1002/mnfr.202100389 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500(7464):541–6. doi: 10.1038/nature12506 [DOI] [PubMed] [Google Scholar]
- 56.Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The controversial role of human gut lachnospiraceae. Microorganisms. 2020;8(4):573. doi: 10.3390/microorganisms8040573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214. doi: 10.1126/science.1241214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lai Z-L, Tseng C-H, Ho HJ, Cheung CKY, Lin J-Y, Chen Y-J, et al. Fecal microbiota transplantation confers beneficial metabolic effects of diet and exercise on diet-induced obese mice. Sci Rep. 2018;8(1):15625. doi: 10.1038/s41598-018-33893-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Visconti A, Le Roy CI, Rosa F, Rossi N, Martin TC, Mohney RP, et al. Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019;10(1):4505. doi: 10.1038/s41467-019-12476-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Millares L, Pérez-Brocal V, Ferrari R, Gallego M, Pomares X, García-Núñez M, et al. Functional metagenomics of the bronchial microbiome in COPD. PLoS ONE. 2015;10(12):e0144448. doi: 10.1371/journal.pone.0144448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Webb ME, Marquet A, Mendel RR, Rébeillé F, Smith AG. Elucidating biosynthetic pathways for vitamins and cofactors. Nat Prod Rep. 2007;24(5):988–1008. doi: 10.1039/b703105j [DOI] [PubMed] [Google Scholar]
- 62.Romine MF, Rodionov DA, Maezato Y, Osterman AL, Nelson WC. Underlying mechanisms for syntrophic metabolism of essential enzyme cofactors in microbial communities. ISME J. 2017;11(6):1434–46. doi: 10.1038/ismej.2017.2 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Means ± SEM represent per-cage averages of 2–3 animals per cage.
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(DOCX)
(XLSX)
Data Availability Statement
Sequencing data have been deposited in NCBI Sequence Read Archive (SRA) under the BioProject number PRJNA1146151.





