J Neurogastroenterol Motil 2022; 28(4): 549-561  https://doi.org/10.5056/jnm22129
Bile Acid and Gut Microbiota in Irritable Bowel Syndrome
Yang Won Min,1,2 Ali Rezaie,1,3 and Mark Pimentel1,3*
1Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA; 2Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; and 3Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai, Los Angeles, CA, USA
Correspondence to: *Mark Pimentel, MD, FRCPC
Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, 700 N San Vicente, Suite G271, West Hollywood, CA 90069, USA
Tel: +1-310-423-0617, E-mail: mark.pimentel@cshs.org
Received: August 1, 2022; Accepted: August 31, 2022; Published online: October 30, 2022
© The Korean Society of Neurogastroenterology and Motility. All rights reserved.

cc This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Gut microbiota and their metabolites like bile acid (BA) have been investigated as causes of irritable bowel syndrome (IBS) symptoms. Primary BAs are synthesized and conjugated in the liver and released into the duodenum. BA biotransformation by gut microbiota begins in the intestine and results in production of a broad range of secondary BAs. Deconjugation is considered the gateway reaction for further modification and is mediated by bile salt hydrolase, which is widely expressed by the gut microbiota. However, gut bacteria that convert primary BAs to secondary BAs belong to a limited number of species, mainly Clostridiales. Like gut microbiota modify BA profile, BAs can shape gut microbiota via direct and indirect actions. BAs have prosecretory effects and regulates gut motility. BAs can also affect gut sensitivity. Because of the vital role of the gut microbiota and BAs in gut function, their bidirectional relationship may contribute to the pathophysiology of IBS. Individuals with IBS have been reported to have altered microbial profiles and modified BA profiles. A significant increase in fecal primary BA and a corresponding decrease in secondary BA have been observed in IBS with predominant diarrhea. In addition, primary BA was positively correlated with IBS symptoms. In IBS with predominant diarrhea, bacteria with reduced abundance mainly belonged to the genera in Ruminococcaceae and exhibited a negative correlation with primary BAs. Integrating the analysis of the gut microbiota and BAs could better understanding of IBS pathophysiology. The gap in this field needs to be further filled in the future.
Keywords: Bile acids and salts; Feces; Irritable bowel syndrome; Intestines; Microbiota
Introduction

Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disorder characterized by recurrent abdominal pain related to defecation and/or changes in the frequency or form of stool.1 According to the predominant stool form of the patients, IBS is classified as IBS with predominant diarrhea (IBS-D), IBS with predominant constipation (IBS-C), IBS with mixed bowel habits, and IBS unclassified.2 The mechanism of symptom generation is multifactorial, including altered motility of the gut, visceral hypersensitivity, central dysfunction, low-grade inflammation, increased intestinal permeability, disorders of the brain-gut axis, and altered gut microbiota.3-5 Over the last decade, microbiota and their metabolites have been paid attention to as the cause of IBS symptoms.5-8

Alteration of the gut microbiota has been reported in patients with IBS.9 The impact of the microbiome on disease etiology could occur via the actions of microbiota-derived metabolites, including bile acids (BAs). BAs are amphipathic molecules produced in the liver, which solubilize lipids into micelles for digestion and absorption.10 Approximately 95% of secreted BAs are reabsorbed in the terminal ileum, and the remaining BAs reach the colon, where they are metabolized by gut microbiota, forming a plethora of microbially modified secondary BAs.11 According to the characteristics of BA profiles, BAs can exert their variable effects on gut function, including fluid secretion, mucosal permeability, and bowel motility.12-14 BAs can also modify gut microbiota.15 Given the vital role of gut microbiota and BAs in regulating gut function, their bidirectional relationship may contribute to the pathophysiology of IBS. Indeed, individuals with IBS have been reported to have altered microbial profiles and modified BA profiles.16

In this review, we describe BA synthesis and enterohepatic circulation (EHC), transformations of BAs, BA signaling mechanisms, and influences of BA on gut microbiota and functions, and summarize the clinical trials investigating alterations of gut microbiota and BA profiles in patients with IBS.

Bile Acid Synthesis and Enterohepatic Circulation

BAs are hydroxylated, amphipathic molecules synthesized in the peroxisomes of the liver from cholesterol through 2 major pathways.17 The newly synthesized BAs are termed primary BAs, including chenodeoxycholic acid (CDCA) and cholic acid (CA), to distinguish them from the products of microbial transformation, termed secondary BAs. Of the 2 major pathways, the classical pathway is more important in adult humans and produces both primary BAs favoring CA biosynthesis.18,19 The alternative pathway results in CDCA biosynthesis and involves less than 10% of BA synthesis.19,20 The enzyme cholesterol 7α-hydroxylase (CYP7A1) is the rate-limiting step in the classical pathway.21,22 7α-hydroxy-4-cholesten-3-one (C4) is a downstream product of CYP7A1, reflecting the enzymatic activity of hepatic CYP7A1. Thus, measuring serum C4 is a simple test for analyzing hepatic BA synthesis, although it requires a standardized specimen collection time because of diurnal variability.8,21 The primary BAs (CDCA and CA) are conjugated to the hydrophilic amino acids, either glycine or taurine (GCDCA/TCDCA and GCA/TCA) in the liver. Humans preferably use glycine for conjugation.23 The conjugation of BAs permits complete ionization of BAs, which increases their solubility and decreases their passive diffusion across the intestinal epithelial barrier, leading to high intraluminal concentrations that facilitate micellar solubilization of dietary lipids.24,25 These primary BAs are secreted into the gallbladder, where they are stored until the consumption of food. Ingestion of food triggers the release of cholecystokinin by enteroendocrine cells, which causes gallbladder contraction and the release of BAs into the duodenum.26,27 There, BAs facilitate the digestion and absorption of dietary lipids, fatty acids, cholesterol, fat-soluble vitamins, and other hydrophobic components of the diet via their surfactant properties, which emulsify fats into micelles.10 Approximately 95% of secreted BAs are reabsorbed in the terminal ileum and transported back into the liver via the EHC.28 The ileal apical Na+-dependent bile salt transporter (ASBT), which has a greater affinity for conjugated than non-conjugated BAs, actively reuptakes conjugated BAs.8,11 After the uptake of BAs by ASBT, ileal lipid-binding proteins bind to intracellular BAs, shuttling them to the heterodimeric protein, organic solute transporter alpha-beta, which efficiently exports them to the portal circulation.29,30 Some passive diffusion across the gut epithelium can also occur for both conjugated and non-conjugated BAs.31 The remaining 5% of BAs that reach the colon are either reabsorbed via passive diffusion or lost in the feces (Figure).11

Figure 1. Bile acids (BAs) synthesis, enterohepatic circulation, and factors affecting BA profiles. BA synthesis could be affected by conditions (eg, decreased in cirrhosis). Small intestinal bacterial overgrowth (SIBO) elevates level of unconjugated BAs in the small intestine. Dysbiosis, BA malabsorption, and rapid gut transit could be associated with increased fecal primary BAs. BSH, bile salt hydrolase.

In ileal enterocytes, BAs activate the nuclear receptor farnesoid X receptor (FXR), with CDCA being the most potent agonist (Table 1).20,32 FXR then induces the expression of fibroblast growth factor 19 (FGF19; rodent ortholog is FGF15). FGF19 is secreted from enterocytes into the portal circulation and activates the cell surface receptor, a complex of the β-klotho protein and FGF receptor 4 in hepatocytes, resulting in the downregulation of CYP7A1 and thereby reducing BA synthesis.8,33-35 As serum FGF19 decreases and is inversely related to serum C4 during BA malabsorption, it could be used for screening tests for malabsorption.8,36-38

Table 1 . Receptors Involved in the Signaling of Bile Acids

ReceptorSitesBA agonistFunctions
FXRNuclear receptor, widespread throughout the body, abundant in the liver, intestine, and kidneysCDCA > DCA > LCA > CARegulation of BA synthesis, absorption, and transport
Maintenance of metabolic homeostasis
Modulation of immune system
TGR5Membrane receptor, widespread throughout the body including the intestine, liver, biliary tract, and gallbladderLCA > DCA > CDCA > CARegulation of the intestinal motility and secretion
Maintenance of metabolic homeostasis
Maintenance of intestinal immune homeostasis
PXRNuclear receptor, abundant in the liver and intestineLCA, only weakly to CDCA, DCA, CA, and conjugated BAsDetoxication of xenobiotics and LCA
Maintenance of intestinal immune homeostasis
Modulation of BA homeostasis
VDRNuclear receptor, widespread throughout the body, abundant in the intestineLCADetoxication of LCA
Modulation of BA synthesis
Maintenance of bone and calcium homeostasis

BA, bile acid; FXR, farnesoid X receptor; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; LCA, lithocholic acid; CA, cholic acid; TGR5, Takeda G protein-coupled receptor 5; PXR, pregnane X receptor; VDR, vitamin D receptor.


Microbial Transformations of Bile Acids

Small quantities of primary BAs that escape EHC reach the colon and undergo extensive microbial biotransformations, including deconjugation, 7α-dehydroxylation, oxidation/epimerization, and sulfation by the gut microbiota, to produce a broad range of secondary BAs.39 In fact, BA biotransformations begin in the small intestine and continues in the colon. Deconjugation of BAs changes their physiochemical properties, making them more lipophilic and susceptible to microbial biotransformation; thus, this is considered the gateway reaction for further modification.40-44 Cleavage of amino acid side chains on conjugated BAs is mediated by bile salt hydrolase (BSH) enzymes that are widely expressed by the gut microbiota.39-43,45

The diversity of intestinal gram-positive bacteria, including Clostridium, Lactobacillus, Bifidobacterium, Enterococcus, and Listeria, contributes to amino acid hydrolysis.45-50 Some gram-negative bacteria such as Bacteroides, Stenotrophomonas, and Brucella are also capable of amino acid hydrolysis.51-53 Using metagenomic analysis, Jones et al44 identified functional BSH among all major bacterial divisions and archaeal species in the gut. Most metagenomic BSH-active clones belonged to the phyla Firmicutes, Bacteroidetes, and Actinobacteria. In addition, Methanobrevibacter smithii and Methanosphera stadmanae encode proteins with high identity to bacterial BSH enzymes. This widespread distribution indicates that BSH is enriched in the human gut community. However, BSHs display different catalytic efficiencies and substrate specificities.54 The organization and regulation of genes encoding BSH differ between species and genera, and conjugations are important in substrate specificity.41 In a taxonomic analysis of BSHs among 11 different populations from 6 continents, 591 BSHs were identified over 117 genera from 12 phyla.54 Among the bacteria positive for BSH activity, more than half of the bacteria belonged to Firmicutes. Notably, significant variations in BSH distribution patterns were also observed based on the geographic region but not sex, age, or body mass index. In addition, BSHs within genera showed a broad range of sequence dissimilarities, owing to the paralogs of BSHs in many strains. Thus, the genus-level patterns of BSH abundance did not reflect the functional variations, necessitating the reclassification of BSHs. BSH activity and subsequent BA modification could significantly affect host physiology, including the regulation of cholesterol metabolism, energy, and inflammation homeostasis.55-57 When treating recurrent Clostridioides difficile infection with fecal microbiota transplant, the restoration of gut BSH functionality contributes to the efficacy of transplant.58 Because of the important roles of BSHs, gaps in the understanding of these enzymes require further research.

Once deconjugated, free primary BAs are metabolized by the resident microbiota into free secondary BAs, such as deoxycholic acid (DCA), lithocholic acid (LCA), and ursodeoxycholic acid (UDCA) via 7α-dehydroxylation and oxidation/epimerization.44,59-61 CDCA is transformed into DCA and CA into LCA by 7α-dehydroxylation. Because DCA and LCA predominate in human feces, 7α-dehydroxylation is the most quantitatively important microbial transformation.41 Human intestinal bacteria capable of 7α-dehydroxylation belong to the genus Clostridium.62-64 Multiple bai genes encoding proteins required for 7α-dehydroxylation have been characterized from Clostridium scindens.41

Epimerization of the 3-, 7-, and 12-hydroxy groups of BAs is carried out by hydroxysteroid dehydrogenase (HSDH) expressed by intestinal bacteria, which diversifies the chemistry of secondary BAs. Epimerization is a reversible change in stereochemistry from the α to β configuration (or vice versa) with the generation of a stable oxo-BA intermediate. While α-hydroxy BAs are amphipathic, both faces of BA are hydrophilic in β orientation.41 Epimerization requires 2 distinct steps: oxidation of the hydroxyl group by a position-specific HSDH, followed by the reduction of the hydroxyl group by another position-specific HSDH.39,41 CA can be epimerized to form ursoCA, 12-epiCA, or isoCA, and CDCA can be epimerized to form either UDCA (7β-hydroxy) or isoCDCA. Epimerization of UDCA to CDCA can also be carried out by 7β-HSDH.41 3-oxoLCA and isoLCA produced by 3α-HSDH are known to suppress differentiation of T helper cells expressing IL-17A and may contribute to gut immune homeostasis.65 Although their enzyme characteristics vary, several intestinal microbes have been observed to produce HSDHs, including a small number of species of Clostridium, Rumminococcus, Bacteroides, and Escherichia coli.66-77

Secondary BAs may undergo further modification including sulphation, imparting changes in their solubility, metabolism, excretion, and toxicity.78,79 Specifically, sulfated BAs are more rapidly excreted in the urine, and sulfated LCA is less efficiently reabsorbed in the intestine than non-sulfated.79 Sulfation of BAs may be associated with constipation.80 In a subset of children with functional constipation, dominant fecal BA was the 3-sulfate of CDCA. Such sulfation may abolish the secretory activity of CDCA and contribute to constipation. In an animal experiment, sulfation prevented secretion caused by di-α-hydroxy BAs (DCA and CDCA) in the colon.81 Notably, increased fecal sulfated BAs were also observed in patients with IBS-D compared with that in IBS-C.36 Further research that includes measurements of sulfotransferase and sulfatase activity is necessary.

Bile Acids in Small Intestinal Bacterial Overgrowth

Small intestinal bacterial overgrowth (SIBO) is one manifestation of gut microbiome dysbiosis and is highly prevalent in IBS.82 Many bacteria in the small intestine have the capacity to metabolize BAs. Shindo et al83 isolated bacterial species from the jejunal fluid obtained from patients with progressive systemic sclerosis and positive 14CO2 breath test. The isolated Bacteroides vulgatus, Eubacterium lentum, Enterococcus, and Lactobacillus bifidus (except for E. coli and Aerobacter aerogenes) were capable of hydrolyzing conjugated BAs in ox gall. Similarly, higher unconjugated BAs from the upper gut aspirate have been observed in malabsorption syndrome patients with SIBO than those without SIBO.84 However, the amount of unconjugated BAs did not correlated with colony counts of isolated bacteria. This observation suggests the different BSH activity among the isolated bacteria. As unconjugated BAs are absorbed from the small intestine into the portal blood, elevated unconjugated serum BA levels have also been found in patients with SIBO.85 Because BA profiles in the small intestine are less affected by gut transit and ileal absorption, investigating the associations between the BA profile and gut microbiome using small bowel aspiration samples of individuals with SIBO could offer us opportunities to further fill the gap of knowledge (Figure).

Bile Acid Signaling Mechanisms

Because of the variety of levels and types of BAs in the intestine, biliary tract, and liver, BAs have emerged as important regulators of epithelial physiology and pathophysiology.11,86 Since the discovery of BA receptors, there have been advances in understanding how BAs exert their effects.

G protein-coupled BA receptor 1, also called Takeda G protein-coupled receptor 5 (TGR5), is responsive to BAs as a cell surface receptor.87,88 TGR5 is a member of the G protein-coupled receptor family, which stimulates cAMP synthesis and activates protein kinase-A, leading to the expression of target genes.87 TGR5 is widely expressed throughout the body, including in the intestine, liver, biliary tract, and gallbladder.56,87,89-91 The functions of TGR5 are thought to be broader than just being a metabolic regulator of energy homeostasis, BA homeostasis, and glucose metabolism.92 Particularly notable is that the activation of TGR5 on the intestinal motor neurons by BAs regulates intestinal motility.93 Conjugated and unconjugated BAs bind to TGR5, with the secondary BAs LCA and DCA being most potent, followed by CDCA and CA.87

Physiological concentrations of free and conjugated BAs activate the nuclear receptor FXR as ligands.94,95 The structure–activity relationship of BAs in activating FXR shows the order of potency of CDCA > DCA > LCA > CA.96 CDCA is an extremely effective activator of FXR, whereas CA is inactive. CA and conjugated BAs are hydrophilic compounds that do not readily cross cell membranes; instead, they are passively diffused or facilitated by BA transport proteins.96,97 FXR is widely expressed throughout the body and highly expressed in the liver, intestine, and kidneys.32,98 As aforementioned, the primary function of FXR activation by BAs is the feedback inhibition of BA synthesis through the downregulation of CYP7A1.8,33-35 In addition, FXR is important in metabolic homeostasis.99,100 Pregnane X receptor (PXR) is another nuclear receptor that can be activated by BAs and is highly expressed in the liver and intestine.101 The role of PXR in the detoxication of xenobiotics and LCA is well known.102,103 PXR also contributes to maintaining intestinal immune homeostasis104,105 PXR downregulates BSH-active bacteria in the intestine and modulates BA homeostasis.101 PXR is activated by LCA but only weakly responds to CDCA, DCA, CA, and conjugated BAs.106 Vitamin D receptors (VDR) are another type of BA-sensitive nuclear receptor, widely expressed throughout the body, with an abundance in the intestine.107 In addition to the classic endogenous ligand, 1,25-dihydroxy vitamin D3, LCA can activate VDR.108 It contributes to the metabolism of BAs as well as calcium homeostasis and bone maintenance. Activation of VDR by LCA or vitamin D induced the expression of CYP3A and the multidrug resistance-associated protein-3 (MRP3).108,109 Hydroxylation of LCA by CYP3A reduces the toxicity of LCA, which is hepatotoxic and a potential enteric carcinogen in the liver and intestine, and MRP3 effluxes LCA into the blood to protect colon cells from LCA toxicity.

Influence of Bile Acids on Gut Microbiota

Like bacterial enzymes chemically modify BA profile, BAs can modify the gut microbiota. BAs are potent antimicrobials and play an important role in the innate immune defense within the intestine.15 As BAs act as detergents in the gut, they allow for the disruption of bacterial membranes, leading to the leakage of proton, potassium ion, and other cellular components and eventually cell death.110 The antimicrobial potency of DCA is greater than that of CA due to its hydrophobicity and detergent properties on bacterial membranes.111 When tested against Staphylococcus aureus, unconjugated BAs exhibit more potent antibacterial action than conjugated BAs.15 Because deconjugation by BSH makes the BAs more lipophilic,112 unconjugated BAs are likely to disrupt membranes and cause intracellular damage.110,113 On the other hand, conjugated BAs can have a more indirect action on the gut microbiota. Activation of FXR induces genes involved in enteroprotection and inhibits bacterial overgrowth and mucosal injury, resulting in the protection of the small intestine from bacterial invasion.114

Gram-negative bacteria are thought to have a higher BA tolerance than Gram-positives.115 Salmonella, E. coli, and Campylobacter are very bile resistant and have been isolated from the gallbladder. Although gram-positive bacteria are more sensitive to the deleterious effects of bile than gram-negative bacteria, bile tolerance is a strain-specific trait, and tolerance of species cannot be generalized.116 For example, Listeria monocytogenes cholecystitis has been reported suggesting a very high level of bile resistance.117 However, decreased levels of BAs in the gut favor gram-negative bacteria allowing proinflammatory microbial taxa to expand, and increased BAs levels favor gram-positive bacteria of the Firmicutes, including bacteria that 7α-dehydroxylate primary BAs to toxic secondary BAs.115,118 In rats, CA feeding induced the significant expansion of DCA-producing bacteria, expanding phylum Firmicutes, class Clostridia, and genus Blautia.119

Influence of Bile Acids on Gut Functions

Generally, BAs induce colonic fluid secretion at high levels.13 However, there is a marked structural specificity for BA-induced secretion, and the α-dihydroxy BAs CDCA and DCA have prosecretory effects.120,121 The trihydroxy BA (CA) does not have prosecretory effects, and UDCA (7β-OH epimer of CDCA) has antisecretory effects.13,122 The conjugation status of BAs is also an important determining factor of their secretory effects.121 Because conjugated BAs are hydrophilic and need to be more lipophilic to cross the cell membrane, they do not have secretory effects. However, conjugated BAs can increase epithelial permeability at a relatively high concentration, which allows them to gain access to regions where they can exert their secretory effects.123 In contrast to prosecretory effects at pathophysiological concentrations, lower levels of DCA is known to downregulate colonic epithelial secretory function.124 These observations suggest that BAs play an important role in regulating colonic fluid levels.

In addition to the regulation of fluid transport, BAs can exert their effects on gut motility.12-14 In humans, rectal CDCA infusion induces propagating pressure waves arising in the proximal colon.14 In another human infusion study, CDCA was more strongly associated with a higher colonic motility index than TCA, which contrasts with the animal (rabbit) results obtained in the same study.125 In an in vitro study, DCA increased isolated human colon motility, whereas CDCA and CA did not.126 The mechanism of action of DCA on smooth muscle activity was revealed as a local neuronal phenomenon in the rabbit colon in vitro.127 However, inhibitory actions of BAs on colon motility have also been shown in animal studies. Luminal bile from the gallbladder and conjugated primary BA (TCA and TCDCA) inhibit contractions of the intestine (isolated rabbit terminal ileal segment and isolated guinea pig ileum smooth muscle strips).128,129 A mouse intestine study demonstrated that DCA inhibits intestinal motility by activating TGR5 on inhibitory motor neurons to release nitric oxide, whereas the effects of UDCA and TDCA were not significant.93 However, contractile inhibition of in vitro colon tissue in particular muscle strips does not indicate decreased gut motility, because peristalsis requires the both contraction and relaxation of gut muscles. In a mouse study, DCA reduced the contractility of colonic longitudinal muscles but could stimulate the ascending contraction and descending relaxation components of the peristaltic reflex of the flat sheet preparation of the proximal colon.130 Furthermore, oral administration of CDCA improved bowel function in patients with either IBS-C or chronic constipation.131,132

BAs can also affect gut sensitivity. Rectal infusion of DCA and CDCA at physiological concentrations reduces rectal sensory thresholds.14,133 The mechanism of visceral hypersensitivity induced by BAs has been investigated in animal studies.134,135 BAs stimulate the release of nerve growth factor from mucosal mast cells through the activation of FXR, resulting in the activation of transient receptor potential vanilloid 1.134 TGR5 agonists, including DCA, also activate subsets of colonic sensory neurons and evoke colonic afferent mechanical hypersensitivity via a transient receptor potential ankyrin A1-dependent mechanism.135

Altered Bile Acid Profile and Gut Microbiota in Patients With Irritable Bowel Syndrome

In several clinical studies, the BA profiles of patients with IBS and those of healthy controls (HC) differ (Table 2).16,36,136-141 The level of total fecal BA was higher in IBS-D patients than that in HC, according to 3 studies of Asian groups.136-138 On the other hand, Western studies demonstrated no differences in the level of total fecal BA between IBS-D groups and HC.16,36,139 Most studies had measured total BA excretion in a single stool, which might be acceptable but is not ideal.142 When stool samples were collected over 48 hours, the level of total fecal BA was higher in IBS-D group than in IBS-C group, but not in HC.139 In addition, total fecal BA correlated with stool weight. However, because total fecal BA was determined from total 3α hydroxy BAs, some subgroups of BAs could have been missed. Taken together, the levels of fecal total BAs in the IBS-D group showed an increasing tendency compared with the levels in IBS-C group or HC. Notably, a systematic review showed that 32% of patients with symptoms consistent with IBS-D had moderate BA malabsorption (75selenium homotaurocholic acid test 7 day retention < 5% of baseline value).143 Zhao et al137 investigated the connection between the gut microbiota in IBS-D group and BA excretion. Twenty-five percent of patients with IBS-D (71 of 290, BA+IBS-D) had an excess of total BA excretion in feces by the 90th percentile cutoff value, determined from the HC (n = 89). BA+IBS-D group exhibited increased C4 and decreased FGF19 levels in sera, as well as an increased severity of diarrheal symptoms compared with the corresponding values in the BAIBS-D group and HC. Different microbial profiles were found in BA+IBS-D compared to either HC or BAIBS-D. The relative abundances of the phyla Firmicutes, Actinobacteria, Fusobacteria, and Proteobacteria increased, and that of Bacteroidetes decreased in the BA+IBS-D group. At the genus level, the abundance of Clostridia bacteria, including Ruminococcus, Clostridium, Eubacterium, and Dorea, was increased in BA+IBS-D. The abundance of Bifidobacterium, Escherichia, and Bilophila was also increased. Correlation analysis revealed that the abundance of Clostridia genera and C. scindens species was positively correlated with the concentrations of total fecal BAs and serum C4 but negatively correlated with serum FGF19 levels, suggesting that Clostridia-rich microbiota influences BA synthesis and excretion in IBS-D. In addition, Clostridia-derived BAs attenuated intestinal FGF19/15 production.

Table 2 . Summary of Clinical Studies Investigating the Bile Acid Profiles and Fecal Microbiota in Irritable Bowel Syndrome

StudyParticipants (n)Fecal BA profileFecal microbiota
Wong et al (2012)139HC (26), IBS-C (26), and IBS-D (26)

Higher total BA in IBS-D than in IBS-C but not than in HC

Total BA correlated with stool weight and fat

Not investigated
Duboc et al (2012)16HC (18) and IBS-D (14)

Similar total BA in IBS-D and HC

Increased PBA (%) and decreased SBA (%) in IBS-D than in HC

Decreased the leptum and Bifidobacterium groups in IBS-D than in HC

Increased E. coli species in IBS-D than in HC

Shin et al (2013)140HC (30), IBS-C (30), and IBS-D or FD (31)

Increased total UBA in IBS-D than in IBS-C but not than in HC

Higher primary UBA (%) in IBS-D than in HC

Lower secretory CDCA and DCA (%) in IBS-C than in HC

Higher non-secretory secondary LCA (%) in IBS-C than in HC

Not investigated
Dior et al (2016)36HC (15), IBS-C (15), and IBS-D (16)

No differences of total BAs among the three groups

Increased PBAs and decreased SBAs in IBS-D compared to HC

Increased sulfated BAs and UDCA in IBS-D compared to HC

Increased CDCA, sulfated BAs, and UDCA in IBS-D compared to IBS-C

No differences in the total fecal bacteria counts in the 3 groups

Increased relative counts of E. coli in IBS-D compared to HC

Increased relative counts of Bacteroides and Bifidobacterium in IBS-C compared to IBS-D and HC

Zhao et al (2020)137HC (89) and IBS-D (290)

Increased total BA in IBS-D than in HC

Increased amounts of CA, CDCA, DCA, LCA, 7-KDCA, UDCA, and ωMCA and increased % of CA, CDCA, UDCA, and 7-KDCA and decreased % of LCA and 12-KLCA in IBS-D with high BA excretion compared with HC

Increased abundances of Clostridia bacteria, Bifidobacterium, Escherichia, and Bilophila and decreased abundances of Alistipes and Bacteroides in IBS-D with high fecal BA excretion

Positive correlation of the abundances of Clostridia genera and C. scindens species with total BAs and serum C4

Wei et al (2020)136HC (28) and IBS-D (55)

Increased total fecal BA in IBS-D than in HC

Increased PBAs and decreased SBAs in IBS-D than in HC

Decreased LCA in IBS-D than in HC

Increased CBAs, UBAs, and ratio of CBAs/UBAs in IBS-D

Increased Proteobacteria, Gammaproteobacteria, Enterobacteriales, and Enterobacteriaceae and decreased Clostridia, Clostridiales, and Ruminococcaceae in IBS-D

Decreased 9 genera including 5 from Ruminococcaceae in IBS-D

Negative correlation of PBAs and positive correlation of SBAs with 8 genera among decreased 9

Wei et al (2021)138HC (32) and IBS-D (52)

Increased total fecal BA in IBS-D than in HC

Increased PBAs and decreased SBAs in IBS-D than in HC

Not investigated
James et al (2021)141HC (97), IBS-D (52), and IBS-C (24)

Increased CA in IBS-D than in HC but not in IBS-C

Increased CDCA in IBS-D than in IBS-C and in HC

Decreased GCA in IBS-C than in IBS-D and in HC

Not investigated

BA, bile acid; HC, healthy control; IBS-C, constipation-predominant irritable bowel syndrome; IBS-D, diarrhea-predominant irritable bowel syndrome; PBA, primary bile acid; SBA, secondary bile acid; E. coli, Escherichia coli; FD, functional diarrhea; UBA, unconjugated bile acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; LCA, lithocholic acid; UDCA, ursodeoxycholic acid; CA, cholic acid; 7-KDCA, 7-ketodeoxycholic acid; ωMCA, ω-muricholic acid; 12-KLCA, 12-ketolithocholic acid; C. scindens, Clostridium scindens; C4, 7α-hydroxy-4-cholesten-3-one; CBA, conjugated bile acid; GCA, glyco-CA.



As the fecal BA pool is modulated by the gut microbiota and gut dysbiosis is implicated in the pathophysiology of IBS.6 Although conjugated BAs increased in IBS-D group, only a small subgroup of patients had a high conjugated BAs to unconjugated BAs ratio.136 Thus, impaired deconjugation of BAs may not widely exist in IBS-D.136 Duboc et al16 observed a significant increase in fecal primary BA and a corresponding decrease in secondary BA in IBS-D compared with that in HC, which has consistently been reported.36,136,138 Moreover, fecal primary BA percentage was positively correlated with the Bristol stool score and stool frequency, whereas secondary BA was negatively correlated with these parameters. Similarly, in other studies, primary BA was positively correlated with the Bristol stool score36 and defecation frequency,36,138 and secondary BA was negatively correlated with defecation frequency.138 Two α-dihydroxy BAs, CDCA and DCA, have prosecretory effects. Indeed, CDCA and DCA correlate with stool frequency and the score in IBS-D,140 and percentages of CDCA and DCA were lower in IBS-C than that in HC.140,141 However, decreased secondary DCA in IBS-D was also observed.16 From other studies providing the absolute amounts of individual BAs, differences in DCA were not significant, but CDCA was 20 times to 30 times higher in IBS-D than in HC.136,138 Thus, the change in CDCA amount seems to predominantly exert an effect on the stool score and frequency in IBS-D. The relationship between fecal BA excretion, fecal BA profile, and colon transit appears to be very complex as being a cause and/or consequence. First, clarification is necessary regarding whether the altered BA profile is due to a dysfunction of microbial biotransformation related to gut dysbiosis or due to rapid gut transit, decreasing the time for gut microbiota to metabolize BAs in patients with IBS-D.

In addition, primary BA showed a positive correlation with abdominal pain in IBS-D.36 Wei et al138 investigated the relationship between BAs and their receptors (TGR5 and VDR) in patients with IBS-D. They observed that the level of TGR5 immunoreactivity in rectosigmoid mucosal biopsies was higher in IBS-D than that in HC. Furthermore, the level of TGR5 was higher in patients with more severe or frequent abdominal pain and was positively associated with primary BAs and negatively associated with secondary BAs. Although no direct link between fecal BAs and abdominal pain has been demonstrated, BAs might contribute to the hypersensitivity of patients with IBS-D via increased TGR5 level in the colon. Increased TGR5 expression is thought to be a compensatory response to decreased levels of potent agonists, secondary BAs (LCA and DCA).

Importantly, an imbalance between primary and secondary BAs in IBS-D has been consistently reported. Because BAs are metabolized by the gut microbiota, dysbiosis could be a critical factor in the altered BA profiles observed in IBS-D patients. Duboc et al16 demonstrated the presence of gut dysbiosis and altered BA profiles in patients with IBS. They observed an increase in E. coli and a decrease in Bifidobacterium and Clostridium leptum in patients with IBS-D compared with that in HC. Although a decrease in the number of bacteria involved in BA transformation could lower the biotransformation of BAs, the results did not show a direct causal link. Thus, they performed an in vivo test to determine the ability of feces to deconjugate BAs in the following study.36 Deconjugation activity was decreased in IBS compared with that in HS and did not differ between IBS-D and IBS-C groups. The BA profiles in stool and blood were also similar between IBS-D and IBS-C. However, the IBS-D group showed an increase in E. coli compared with that in the HC group, and IBS-C showed an increase in Bifidobacterium and Bacteroides compared with that in the HC and IBS-D groups.

To determine whether the alteration of BA profiles is due to a dysfunction in biotransformation related to gut dysbiosis, establishing a direct link between the BA profile alteration and microbial variations is necessary. Wei et al136 assessed the correlation between fecal BAs (CA, CDCA, DCA, LCA, and UDCA) and the gut microbiome in Chinese patients with IBS-D. At the genus level, nine genera were significantly less abundant, including the genera in Ruminococcaceae (Anaerofilum, Anaerotruncus, Faecalibacterium, Gemmiger, and Oscillibacter), in Lachnospiraceae (Coprococcus), in Porphyromonadaceae (Odoribacter), in Rikenellaceae (Alistipes), and in Synergistaceae (Cloacibacillus); 8 genera were more abundant, including Escherichia/Shigella, Enterococcus, Streptococcus, Rothia, Klebsiella, Saccharibacteria genera incertae sedis, Fusobacterium, and Veillonella. The 8 genera that were reduced in IBS-D, except for Cloacibacillus, exhibited a negative correlation with primary BAs (CA and CDCA) and a positive correlation with secondary BAs (DCA, LCA, and UDCA). However, correlations cannot be equated to causal associations, and a longitudinal study is required to confirm these results.

Other Conditions That Affect the Luminal Bile Acid Profile

Conditions other than microbial biotransformation are linked to changes in luminal BA characteristics. The BA pool in patients with cirrhosis is depleted due to decreased synthesis.144 The decrease in fecal BAs promotes the depletion of Firmicutes and expansion of proinflammatory pathogenic bacteria of Proteobacteria.115 Depletion of 7α-dehydroxylation bacteria leads to an increased primary to secondary BA ratio in patients with cirrhosis. Because the gallbladder stores and releases primary BAs, cholecystectomy may affect BA homeostasis. Gallbladder removal may increase the formation and pool size of secondary BAs due to the increased exposure of primary BAs to bacterial biotransformation in the intestine. However, cholecystectomy does not lead to significant changes in the BA profile in the long term.145,146 Defects in the formation and transport of BA could affect the BA pool.147 Several inherited defects in enzymes, including CYP7A1, could be involved in BA synthesis. In multiple biosynthetic pathways, a single enzyme defect is usually not sufficient to block the production of all BAs. These rare genetic diseases are characterized by cholestasis, neurological disorders, and fat-soluble vitamin deficiency.148 Inherited transporter defects are also rare, and the spectrum ranges from benign conditions such as benign recurrent intrahepatic cholestasis to progressive familial intrahepatic cholestasis.149 Typically, the first presentation of progressive familial intrahepatic cholestasis is in early childhood, frequently followed by a severe course requiring liver transplantation before adulthood.150

Conclusion

Diverse BA profiles can regulate gut functions in terms of fluid absorption and secretion, motility, and sensitivity. Elucidating the underlying mechanisms of action help clarify their contributions to the pathophysiology of IBS, especially in IBS-D. Because of the reciprocal relationship between altered BA profiles and dysbiosis in IBS, integrating their analysis seems necessary and could provide insights into the pathophysiology and treatment of IBS. However, few relevant studies have been conducted, and they involved only a small number of subjects, mostly patients with IBS-D. Furthermore, the observational and cross-sectional study designs did not show causal associations among altered BA profiles, gut dysbiosis, and bowel symptoms. Therefore, associations between the BA profile and gut microbiome require further investigation using several conditions and samples, for example, small bowel aspiration samples of patients with SIBO. In addition, large longitudinal and interventional studies are warranted to verify previous observations.

Financial support

None.

Conflicts of interest

None.

Author contributions

Yang Won Min drafted the manuscript; Ali Rezaie performed critical revision of the manuscript; and Mark Pimentel contributed to editing and revision of the manuscript. All the authors approved the final version of the manuscript.

References
  1. Song KH, Jung HK, Kim HJ, et al. Clinical practice guidelines for irritable bowel syndrome in Korea, 2017 revised edition. J Neurogastroenterol Motil 2018;24:197-215.
    Pubmed KoreaMed CrossRef
  2. Mearín F, Lacy BE, Chang L, et al. Bowel disorders. Gastroenterology 2016;15:P1393-1407, e5.
    Pubmed CrossRef
  3. Ford AC, Lacy BE, Talley NJ. Irritable bowel syndrome. N Engl J Med 2017;376:2566-2578.
    Pubmed CrossRef
  4. Camilleri M, Ford AC. Irritable bowel syndrome: pathophysiology and current therapeutic approaches. Handb Exp Pharmacol 2017;239:75-113.
    Pubmed CrossRef
  5. Camilleri M. Peripheral mechanisms in irritable bowel syndrome. N Engl J Med 2013;368:578-579.
    Pubmed CrossRef
  6. Pimentel M, Lembo A. Microbiome and its role in irritable bowel syndrome. Dig Dis Sci 2020;65:829-839.
    Pubmed CrossRef
  7. Xiao L, Liu Q, Luo M, Xiong L. Gut microbiota-derived metabolites in irritable bowel syndrome. Front Cell Infect Microbiol 2021;11:729346.
    Pubmed KoreaMed CrossRef
  8. Camilleri M. bile acid diarrhea: prevalence, pathogenesis, and therapy. Gut Liver 2015;9:332-339.
    Pubmed KoreaMed CrossRef
  9. Singh P, Lembo A. Emerging role of the gut microbiome in irritable bowel syndrome. Gastroenterol Clin North Am 2021;50:523-545.
    Pubmed CrossRef
  10. Hofmann AF, Borgstroem B. The intraluminal phase of fat digestion in man: the lipid content of the micellar and oil phases of intestinal content obtained during fat digestion and absorption. J Clin Invest 1964;43:247-257.
    Pubmed KoreaMed CrossRef
  11. Hegyi P, Maléth J, Walters JR, Hofmann AF, Keely SJ. Guts and gall: bile acids in regulation of intestinal epithelial function in health and disease. Physiol Rev 2018;98:1983-2023.
    Pubmed CrossRef
  12. Chadwick VS, Gaginella TS, Carlson GL, Debongnie JC, Phillips SF, Hofmann AF. Effect of molecular structure on bile acid-induced alterations in absorptive function, permeability, and morphology in the perfused rabbit colon. J Lab Clin Med 1979;94:661-674.
    Pubmed
  13. Mekjian HS, Phillips SF, Hofmann AF. Colonic secretion of water and electrolytes induced by bile acids: perfusion studies in man. J Clin Invest 1971;50:1569-1577.
    Pubmed KoreaMed CrossRef
  14. Bampton PA, Dinning PG, Kennedy ML, Lubowski DZ, Cook IJ. The proximal colonic motor response to rectal mechanical and chemical stimulation. Am J Physiol Gastrointest Liver Physiol 2002;282:G443-G449.
    Pubmed CrossRef
  15. Sannasiddappa TH, Lund PA, Clarke SR. In vitro antibacterial activity of unconjugated and conjugated bile salts on Staphylococcus aureus. Front Microbiol 2017;8:1581.
    Pubmed KoreaMed CrossRef
  16. Duboc H, Rainteau D, Rajca S, et al. Increase in fecal primary bile acids and dysbiosis in patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil 2012;24:513-520, e246-e247.
    Pubmed CrossRef
  17. Ferdinandusse S, Houten SM. Peroxisomes and bile acid biosynthesis. Biochim Biophys Acta 2006;1763:1427-1440.
    Pubmed CrossRef
  18. Zhan K, Zheng H, Li J, et al. Gut microbiota-bile acid crosstalk in diarrhea-irritable bowel syndrome. Biomed Res Int 2020;2020:3828249.
    Pubmed KoreaMed CrossRef
  19. Liu T, Song X, Khan S, et al. The gut microbiota at the intersection of bile acids and intestinal carcinogenesis: an old story, yet mesmerizing. Int J Cancer 2020;146:1780-1790.
    Pubmed CrossRef
  20. Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol 2018;15:111-128.
    Pubmed KoreaMed CrossRef
  21. Gälman C, Arvidsson I, Angelin B, Rudling M. Monitoring hepatic cholesterol 7alpha-hydroxylase activity by assay of the stable bile acid intermediate 7alpha-hydroxy-4-cholesten-3-one in peripheral blood. J Lipid Res 2003;44:859-866.
    Pubmed CrossRef
  22. Camilleri M, Nadeau A, Tremaine WJ, et al. Measurement of serum 7alpha-hydroxy-4-cholesten-3-one (or 7alphaC4), a surrogate test for bile acid malabsorption in health, ileal disease and irritable bowel syndrome using liquid chromatography-tandem mass spectrometry. Neurogastroenterol Motil 2009;21:734-e43.
    Pubmed KoreaMed CrossRef
  23. Grüner N, Mattner J. Bile acids and microbiota: multifaceted and versatile regulators of the liver-gut axis. Int J Mol Sci 2021;22:1397.
    Pubmed KoreaMed CrossRef
  24. Bortolini O, Medici A, Poli S. Biotransformations on steroid nucleus of bile acids. Steroids 1997;62:564-577.
    Pubmed CrossRef
  25. Camilleri M. Bile acid detergency: permeability, inflammation and effects of sulfation. Am J Physiol Gastrointest Liver Physiol 2022;322:G480-G488.
    Pubmed KoreaMed CrossRef
  26. Hofmann AF. The continuing importance of bile acids in liver and intestinal disease. Arch Intern Med 1999;159:2647-2658.
    Pubmed CrossRef
  27. Di Ciaula A, Garruti G, Lunardi Baccetto R, et al. Bile Acid Physiology. Ann Hepatol 2017;16(suppl 1):S4-S14.
    Pubmed CrossRef
  28. Dowling RH. The enterohepatic circulation. Gastroenterology 1972;62:122-140.
    Pubmed CrossRef
  29. Dawson PA. Roles of ileal ASBT and OSTalpha-OSTbeta in regulating bile acid signaling. Dig Dis 2017;35:261-266.
    Pubmed KoreaMed CrossRef
  30. Dawson PA. Role of the intestinal bile acid transporters in bile acid and drug disposition:169-203.
    Pubmed KoreaMed CrossRef
  31. Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res 2015;56:1085-1099.
    Pubmed KoreaMed CrossRef
  32. Gadaleta RM, van Mil SW, Oldenburg B, Siersema PD, Klomp LW, van Erpecum KJ. Bile acids and their nuclear receptor FXR: relevance for hepatobiliary and gastrointestinal disease. Biochim Biophys Acta 2010;1801:683-692.
    Pubmed CrossRef
  33. Hofmann AF. The syndrome of ileal disease and the broken enterohepatic circulation: cholerheic enteropathy. Gastroenterology 1967;52:752-757.
    Pubmed CrossRef
  34. Duboc H, Rajca S, Rainteau D, et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut 2013;62:531-539.
    Pubmed CrossRef
  35. Sinha J, Chen F, Miloh T, Burns RC, Yu Z, Shneider BL. β-Klotho and FGF-15/19 inhibit the apical sodium-dependent bile acid transporter in enterocytes and cholangiocytes. Am J Physiol Gastrointest Liver Physiol 2008;295:G996-G1003.
    Pubmed KoreaMed CrossRef
  36. Dior M, Delagrèverie H, Duboc H, et al. Interplay between bile acid metabolism and microbiota in irritable bowel syndrome. Neurogastroenterol Motil 2016;28:1330-1340.
    Pubmed CrossRef
  37. Pattni SS, Brydon WG, Dew T, et al. Fibroblast growth factor 19 in patients with bile acid diarrhoea: a prospective comparison of FGF19 serum assay and SeHCAT retention. Aliment Pharmacol Ther 2013;38:967-976.
    Pubmed CrossRef
  38. Walters JR, Pattni SS. Managing bile acid diarrhoea. Therap Adv Gastroenterol 2010;3:349-357.
    Pubmed KoreaMed CrossRef
  39. Guzior DV, Quinn RA. Review: microbial transformations of human bile acids. Microbiome 2021;9:140.
    Pubmed KoreaMed CrossRef
  40. Begley M, Hill C, Gahan CG. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol 2006;72:1729-1738.
    Pubmed KoreaMed CrossRef
  41. Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006;47:241-259.
    Pubmed CrossRef
  42. Batta AK, Salen G, Arora R, Shefer S, Batta M, Person A. Side chain conjugation prevents bacterial 7-dehydroxylation of bile acids. J Biol Chem 1990;265:10925-10928.
    Pubmed CrossRef
  43. Van Eldere J, Celis P, De Pauw G, Lesaffre E, Eyssen H. Tauroconjugation of cholic acid stimulates 7 alpha-dehydroxylation by fecal bacteria. Appl Environ Microbiol 1996;62:656-661.
    Pubmed KoreaMed CrossRef
  44. Jones BV, Begley M, Hill C, Gahan CG, Marchesi JR. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc Natl Acad Sci USA 2008;105:13580-13585.
    Pubmed KoreaMed CrossRef
  45. Bourgin M, Kriaa A, Mkaouar H, et al. Bile salt hydrolases: at the crossroads of microbiota and human health. Microorganisms 2021;9:1122.
    Pubmed KoreaMed CrossRef
  46. Coleman JP, Hudson LL. Cloning and characterization of a conjugated bile acid hydrolase gene from Clostridium perfringens. Appl Environ Microbiol 1995;61:2514-2520.
    Pubmed KoreaMed CrossRef
  47. Corzo G, Gilliland SE. Bile salt hydrolase activity of three strains of Lactobacillus acidophilus. J Dairy Sci 1999;82:472-480.
    Pubmed CrossRef
  48. Kim GB, Yi SH, Lee BH. Purification and characterization of three different types of bile salt hydrolases from Bifidobacterium strains. J Dairy Sci 2004;87:258-266.
    Pubmed CrossRef
  49. Wijaya A, Hermann A, Abriouel H, et al. Cloning of the bile salt hydrolase (bsh) gene from Enterococcus faecium FAIR-E 345 and chromosomal location of bsh genes in food enterococci. J Food Prot 2004;67:2772-2778.
    Pubmed CrossRef
  50. Dussurget O, Cabanes D, Dehoux P, et al. Listeria monocytogenes bile salt hydrolase is a PrfA-regulated virulence factor involved in the intestinal and hepatic phases of listeriosis. Mol Microbiol 2002;45:1095-1106.
    Pubmed CrossRef
  51. Kawamoto K, Horibe I, Uchida K. Purification and characterization of a new hydrolase for conjugated bile acids, chenodeoxycholyltaurine hydrolase, from Bacteroides vulgatus. J Biochem 1989;106:1049-1053.
    Pubmed CrossRef
  52. Dean M, Cervellati C, Casanova E, et al. Characterization of cholylglycine hydrolase from a bile-adapted strain of Xanthomonas maltophilia and its application for quantitative hydrolysis of conjugated bile salts. Appl Environ Microbiol 2002;68:3126-3128.
    Pubmed KoreaMed CrossRef
  53. Delpino MV, Marchesini MI, Estein SM, et al. A bile salt hydrolase of Brucella abortus contributes to the establishment of a successful infection through the oral route in mice. Infect Immun 2007;75:299-305.
    Pubmed KoreaMed CrossRef
  54. Song Z, Cai Y, Lao X, et al. Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. Microbiome 2019;7:9.
    Pubmed KoreaMed CrossRef
  55. Houten SM, Watanabe M, Auwerx J. Endocrine functions of bile acids. EMBO J 2006;25:1419-1425.
    Pubmed KoreaMed CrossRef
  56. Watanabe M, Houten SM, Mataki C, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 2006;439:484-489.
    Pubmed CrossRef
  57. Martin FP, Dumas ME, Wang Y, et al. A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model. Mol Syst Biol 2007;3:112.
    Pubmed KoreaMed CrossRef
  58. Mullish BH, McDonald JAK, Pechlivanis A, et al. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection. Gut 2019;68:1791-1800.
    Pubmed KoreaMed CrossRef
  59. Ridlon JM, Harris SC, Bhowmik S, Kang DJ, Hylemon PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes 2016;7:22-39.
    Pubmed KoreaMed CrossRef
  60. Gérard P. Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens 2013;3:14-24.
    Pubmed KoreaMed CrossRef
  61. Hamilton JP, Xie G, Raufman JP, et al. Human cecal bile acids: concentration and spectrum. Am J Physiol Gastrointest Liver Physiol 2007;293:G256-263.
    Pubmed CrossRef
  62. Hirano S, Nakama R, Tamaki M, Masuda N, Oda H. Isolation and characterization of thirteen intestinal microorganisms capable of 7 alpha-dehydroxylating bile acids. Appl Environ Microbiol 1981;41:737-745.
    Pubmed KoreaMed CrossRef
  63. Kitahara M, Takamine F, Imamura T, Benno Y. Assignment of Eubacterium sp. VPI 12708 and related strains with high bile acid 7alpha-dehydroxylating activity to Clostridium scindens and proposal of Clostridium hylemonae sp. nov., isolated from human faeces. Int J Syst Evol Microbiol 2000;50(pt 3):971-978.
    Pubmed CrossRef
  64. Wells JE, Williams KB, Whitehead TR, Heuman DM, Hylemon PB. Development and application of a polymerase chain reaction assay for the detection and enumeration of bile acid 7alpha-dehydroxylating bacteria in human feces. Clin Chim Acta 2003;331:127-134.
    Pubmed CrossRef
  65. Paik D, Yao L, Zhang Y, et al. Human gut bacteria produce TH17-modulating bile acid metabolites. Nature 2022;603:907-912.
    Pubmed KoreaMed CrossRef
  66. Hylemon PB, Sherrod JA. Multiple forms of 7-alpha-hydroxysteroid dehydrogenase in selected strains of Bacteroides fragilis. J Bacteriol 1975;122:418-424.
    Pubmed KoreaMed CrossRef
  67. Macdonald IA, Meier EC, Mahony DE, Costain GA. 3alpha-, 7alpha- and 12alpha-hydroxysteroid dehydrogenase activities from Clostridium perfringens. Biochim Biophys Acta 1976;450:142-153.
    Pubmed CrossRef
  68. MacDonald IA, Jellett JF, Mahony DE, Holdeman LV. Bile salt 3 alpha- and 12 alpha-hydroxysteroid dehydrogenases from Eubacterium lentum and related organisms. Appl Environ Microbiol 1979;37:992-1000.
    Pubmed KoreaMed CrossRef
  69. Sutherland JD, Williams CN. Bile acid induction of 7 alpha- and 7 beta-hydroxysteroid dehydrogenases in Clostridium limosum. J Lipid Res 1985;26:344-350.
    Pubmed CrossRef
  70. Akao T, Akao T, Hattori M, Namba T, Kobashi K. Enzymes involved in the formation of 3 beta, 7 beta-dihydroxy-12-oxo-5 beta-cholanic acid from dehydrocholic acid by Ruminococcus sp. obtained from human intestine. Biochim Biophys Acta 1987;921:275-280.
    Pubmed CrossRef
  71. Edenharder R, Pfützner A, Hammann R. Characterization of NAD-dependent 3 alpha- and 3 beta-hydroxysteroid dehydrogenase and of NADP-dependent 7 beta-hydroxysteroid dehydrogenase from Peptostreptococcus productus. Biochim Biophys Acta 1989;1004:230-238.
    Pubmed CrossRef
  72. Edenharder R, Pfützner M, Hammann R. NADP-dependent 3 beta-, 7 alpha- and 7 beta-hydroxysteroid dehydrogenase activities from a lecithinase-lipase-negative Clostridium species 25.11.c. Biochim Biophys Acta 1989;1002:37-44.
    Pubmed CrossRef
  73. Gopal-Srivastava R, Mallonee DH, White WB, Hylemon PB. Multiple copies of a bile acid-inducible gene in Eubacterium sp. strain VPI 12708. J Bacteriol 1990;172:4420-4426.
    Pubmed KoreaMed CrossRef
  74. Coleman JP, Hudson LL, Adams MJ. Characterization and regulation of the NADP-linked 7 alpha-hydroxysteroid dehydrogenase gene from Clostridium sordellii. J Bacteriol 1994;176:4865-4874.
    Pubmed KoreaMed CrossRef
  75. Wells JE, Hylemon PB. Identification and characterization of a bile acid 7alpha-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7alpha-dehydroxylating strain isolated from human feces. Appl Environ Microbiol 2000;66:1107-1113.
    Pubmed KoreaMed CrossRef
  76. Mallonee DH, Lijewski MA, Hylemon PB. Expression in Escherichia coli and characterization of a bile acid-inducible 3 alpha-hydroxysteroid dehydrogenase from Eubacterium sp. strain VPI 12708. Curr Microbiol 1995;30:259-263.
    Pubmed CrossRef
  77. Yoshimoto T, Higashi H, Kanatani A, et al. Cloning and sequencing of the 7 alpha-hydroxysteroid dehydrogenase gene from Escherichia coli HB101 and characterization of the expressed enzyme. J Bacteriol 1991;173:2173-2179.
    Pubmed KoreaMed CrossRef
  78. Palmer RH. The formation of bile acid sulfates: a new pathway of bile acid metabolism in humans. Proc Natl Acad Sci USA 1967;58:1047-1050.
    Pubmed KoreaMed CrossRef
  79. Camilleri M. Bile acid detergency: permeability, inflammation, and effects of sulfation. Am J Physiol Gastrointest Liver Physiol 2022;322:G480-G488.
    Pubmed KoreaMed CrossRef
  80. Hofmann AF, Loening-Baucke V, Lavine JE, et al. Altered bile acid metabolism in childhood functional constipation: inactivation of secretory bile acids by sulfation in a subset of patients. J Pediatr Gastroenterol Nutr 2008;47:598-606.
    Pubmed CrossRef
  81. Breuer NF, Rampton DS, Tammar A, Murphy GM, Dowling RH. Effect of colonic perfusion with sulfated and nonsulfated bile acids on mucosal structure and function in the rat. Gastroenterology 1983;84(5 pt 1):969-977.
    Pubmed CrossRef
  82. Takakura W, Pimentel M. Small intestinal bacterial overgrowth and irritable bowel syndrome - an update. Front Psychiatry 2020;11:664.
    Pubmed KoreaMed CrossRef
  83. Shindo K, Machida M, Koide K, Fukumura M, Yamazaki R. Deconjugation ability of bacteria isolated from the jejunal fluid of patients with progressive systemic sclerosis and its gastric pH. Hepatogastroenterology 1998;45:1643-1650.
    Pubmed
  84. Bala L, Ghoshal UC, Ghoshal U, et al. Malabsorption syndrome with and without small intestinal bacterial overgrowth: a study on upper-gut aspirate using 1H NMR spectroscopy. Magn Reson Med 2006;56:738-744.
    Pubmed CrossRef
  85. Masclee A, Tangerman A, van Schaik A, van der Hoek EW, van Tongeren JH. Unconjugated serum bile acids as a marker of small intestinal bacterial overgrowth. Eur J Clin Invest 1989;19:384-389.
    Pubmed CrossRef
  86. Ní Dhonnabháín R, Xiao Q, O'Malley D. Aberrant gut-to-brain signaling in irritable bowel syndrome - the role of bile acids. Front Endocrinol (Lausanne) 2021;12:745190.
    Pubmed KoreaMed CrossRef
  87. Kawamata Y, Fujii R, Hosoya M, et al. A G protein-coupled receptor responsive to bile acids. J Biol Chem 2003;278:9435-9440.
    Pubmed CrossRef
  88. Maruyama T, Miyamoto Y, Nakamura T, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun 2002;298:714-719.
    Pubmed CrossRef
  89. Keitel V, Häussinger D. TGR5 in the biliary tree. Dig Dis 2011;29:45-47.
    Pubmed CrossRef
  90. Keitel V, Cupisti K, Ullmer C, Knoefel WT, Kubitz R, Häussinger D. The membrane-bound bile acid receptor TGR5 is localized in the epithelium of human gallbladders. Hepatology 2009;50:861-870.
    Pubmed CrossRef
  91. Maruyama T, Tanaka K, Suzuki J, et al. Targeted disruption of G protein-coupled bile acid receptor 1 (Gpbar1/M-Bar) in mice. J Endocrinol 2006;191:197-205.
    Pubmed CrossRef
  92. Guo C, Chen WD, Wang YD. TGR5, not only a metabolic regulator. Front Physiol 2016;7:646.
    Pubmed KoreaMed CrossRef
  93. Poole DP, Godfrey C, Cattaruzza F, et al. Expression and function of the bile acid receptor GpBAR1 (TGR5) in the murine enteric nervous system. Neurogastroenterol Motil 2010;22:814-825, e227-e228.
    Pubmed KoreaMed CrossRef
  94. Parks DJ, Blanchard SG, Bledsoe RK, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science 1999;284:1365-1368.
    Pubmed CrossRef
  95. Makishima M, Okamoto AY, Repa JJ, et al. Identification of a nuclear receptor for bile acids. Science 1999;284:1362-1365.
    Pubmed CrossRef
  96. Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell 1999;3:543-553.
    Pubmed CrossRef
  97. Craddock AL, Love MW, Daniel RW, et al. Expression and transport properties of the human ileal and renal sodium-dependent bile acid transporter. Am J Physiol 1998;274:G157-G169.
    Pubmed CrossRef
  98. Bishop-Bailey D, Walsh DT, Warner TD. Expression and activation of the farnesoid X receptor in the vasculature. Proc Natl Acad Sci USA 2004;101:3668-3673.
    Pubmed KoreaMed CrossRef
  99. Cariou B, van Harmelen K, Duran-Sandoval D, et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem 2006;281:11039-11049.
    Pubmed CrossRef
  100. Degirolamo C, Modica S, Vacca M, et al. Prevention of spontaneous hepatocarcinogenesis in farnesoid X receptor-null mice by intestinal-specific farnesoid X receptor reactivation. Hepatology 2015;61:161-170.
    Pubmed CrossRef
  101. Dempsey JL, Wang D, Siginir G, et al. Pharmacological activation of PXR and CAR downregulates distinct bile acid-metabolizing intestinal bacteria and alters bile acid homeostasis. Toxicol Sci 2019;168:40-60.
    Pubmed KoreaMed CrossRef
  102. Chai X, Zeng S, Xie W. Nuclear receptors PXR and CAR: implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab Toxicol 2013;9:253-266.
    Pubmed CrossRef
  103. de Aguiar Vallim TQ, Tarling EJ, Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab 2013;17:657-669.
    Pubmed KoreaMed CrossRef
  104. Terc J, Hansen A, Alston L, Hirota SA. Pregnane X receptor agonists enhance intestinal epithelial wound healing and repair of the intestinal barrier following the induction of experimental colitis. Eur J Pharm Sci 2014;55:12-19.
    Pubmed CrossRef
  105. Mencarelli A, Renga B, Palladino G, et al. Inhibition of NF-κB by a PXR-dependent pathway mediates counter-regulatory activities of rifaximin on innate immunity in intestinal epithelial cells. Eur J Pharmacol 2011;668:317-324.
    Pubmed CrossRef
  106. Schuetz EG, Strom S, Yasuda K, et al. Disrupted bile acid homeostasis reveals an unexpected interaction among nuclear hormone receptors, transporters, and cytochrome P450. J Biol Chem 2001;276:39411-39418.
    Pubmed CrossRef
  107. Wang Y, Zhu J, DeLuca HF. Where is the vitamin D receptor? Arch Biochem Biophys 2012;523:123-133.
    Pubmed CrossRef
  108. Makishima M, Lu TT, Xie W, et al. Vitamin D receptor as an intestinal bile acid sensor. Science 2002;296:1313-1316.
    Pubmed CrossRef
  109. McCarthy TC, Li X, Sinal CJ. Vitamin D receptor-dependent regulation of colon multidrug resistance-associated protein 3 gene expression by bile acids. J Biol Chem 2005;280:23232-23242.
    Pubmed CrossRef
  110. Kurdi P, Kawanishi K, Mizutani K, Yokota A. Mechanism of growth inhibition by free bile acids in lactobacilli and bifidobacteria. J Bacteriol 2006;188:1979-1986.
    Pubmed KoreaMed CrossRef
  111. Begley M, Gahan CG, Hill C. The interaction between bacteria and bile. FEMS Microbiol Rev 2005;29:625-651.
    Pubmed CrossRef
  112. Joyce SA, Shanahan F, Hill C, Gahan CG. Bacterial bile salt hydrolase in host metabolism: potential for influencing gastrointestinal microbe-host crosstalk. Gut Microbes 2014;5:669-674.
    Pubmed KoreaMed CrossRef
  113. Urdaneta V, Casadesús J. Interactions between bacteria and bile salts in the gastrointestinal and hepatobiliary tracts. Front Med (Lausanne) 2017;4:163.
    Pubmed KoreaMed CrossRef
  114. Inagaki T, Moschetta A, Lee YK, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci USA 2006;103:3920-3925.
    Pubmed KoreaMed CrossRef
  115. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol 2014;30:332-338.
    Pubmed KoreaMed CrossRef
  116. Jacobsen CN, Rosenfeldt Nielsen V, Hayford AE, et al. Screening of probiotic activities of forty-seven strains of Lactobacillus spp. by in vitro techniques and evaluation of the colonization ability of five selected strains in humans. Appl Environ Microbiol 1999;65:4949-4956.
    Pubmed KoreaMed CrossRef
  117. Allerberger F, Langer B, Hirsch O, Dierich MP, Seeliger HP. Listeria monocytogenes cholecystitis. Z Gastroenterol 1989;27:145-147.
    Pubmed CrossRef
  118. Kakiyama G, Pandak WM, Gillevet PM, et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 2013;58:949-955.
    Pubmed KoreaMed CrossRef
  119. Islam KB, Fukiya S, Hagio M, et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 2011;141:1773-1781.
    Pubmed CrossRef
  120. Gordon SJ, Kinsey MD, Magen JS, Joseph RE, Kowlessar OD. Structure of bile acids associated with secretion in the rat cecum. Gastroenterology 1979;77:38-44.
    Pubmed CrossRef
  121. Keely SJ, Scharl MM, Bertelsen LS, Hagey LR, Barrett KE, Hofmann AF. Bile acid-induced secretion in polarized monolayers of T84 colonic epithelial cells: structure-activity relationships. Am J Physiol Gastrointest Liver Physiol 2007;292:G290-G297.
    Pubmed CrossRef
  122. Kelly OB, Mroz MS, Ward JB, et al. Ursodeoxycholic acid attenuates colonic epithelial secretory function. J Physiol 2013;591:2307-2318.
    Pubmed KoreaMed CrossRef
  123. Dharmsathaphorn K, Huott PA, Vongkovit P, Beuerlein G, Pandol SJ, Ammon HV. Cl- secretion induced by bile salts. A study of the mechanism of action based on a cultured colonic epithelial cell line. J Clin Invest 1989;84:945-953.
    Pubmed KoreaMed CrossRef
  124. Keating N, Mroz MS, Scharl MM, et al. Physiological concentrations of bile acids down-regulate agonist induced secretion in colonic epithelial cells. J Cell Mol Med 2009;13(8B):2293-2303.
    Pubmed KoreaMed CrossRef
  125. Kirwan WO, Smith AN, Mitchell WD, Falconer JD, Eastwood MA. Bile acids and colonic motility in the rabbit and the human. Gut 1975;16:894-902.
    Pubmed KoreaMed CrossRef
  126. Flynn M, Hammond P, Darby C, Taylor I. Effects of bile acids on human colonic motor function in vitro. Digestion 1982;23:211-216.
    Pubmed CrossRef
  127. Shiff SJ, Soloway RD, Snape WJ Jr. Mechanism of deoxycholic acid stimulation of the rabbit colon. J Clin Invest 1982;69:985-992.
    Pubmed KoreaMed CrossRef
  128. Armstrong DN, Krenz HK, Modlin IM, Ballantyne GH. Bile salt inhibition of motility in the isolated perfused rabbit terminal ileum. Gut 1993;34:483-488.
    Pubmed KoreaMed CrossRef
  129. Romero F, Frediani-Neto E, Paiva TB, Paiva AC. Role of Na+/Ca++ exchange in the relaxant effect of sodium taurocholate on the guinea-pig ileum smooth muscle. Naunyn Schmiedebergs Arch Pharmacol 1993;348:325-331.
    Pubmed CrossRef
  130. Alemi F, Poole DP, Chiu J, et al. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice. Gastroenterology 2013;144:145-154.
    Pubmed KoreaMed CrossRef
  131. Rao AS, Wong BS, Camilleri M, et al. Chenodeoxycholate in females with irritable bowel syndrome-constipation: a pharmacodynamic and pharmacogenetic analysis. Gastroenterology 2010;139:1549-1558, 1558, e1.
    Pubmed KoreaMed CrossRef
  132. Bazzoli F, Malavolti M, Petronelli A, Barbara L, Roda E. Treatment of constipation with chenodeoxycholic acid. J Int Med Res 1983;11:120-123.
    Pubmed CrossRef
  133. Edwards CA, Brown S, Baxter AJ, Bannister JJ, Read NW. Effect of bile acid on anorectal function in man. Gut 1989;30:383-386.
    Pubmed KoreaMed CrossRef
  134. Li WT, Luo QQ, Wang B, et al. Bile acids induce visceral hypersensitivity via mucosal mast cell-to-nociceptor signaling that involves the farnesoid X receptor/nerve growth factor/transient receptor potential vanilloid 1 axis. FASEB J 2019;33:2435-2450.
    Pubmed CrossRef
  135. Castro J, Harrington AM, Lieu T, et al. Activation of pruritogenic TGR5, MrgprA3, and MrgprC11 on colon-innervating afferents induces visceral hypersensitivity. JCI Insight 2019;4:e131712.
    Pubmed KoreaMed CrossRef
  136. Wei W, Wang HF, Zhang Y, Zhang YL, Niu BY, Yao SK. Altered metabolism of bile acids correlates with clinical parameters and the gut microbiota in patients with diarrhea-predominant irritable bowel syndrome. World J Gastroenterol 2020;26:7153-7172.
    Pubmed KoreaMed CrossRef
  137. Zhao L, Yang W, Chen Y, et al. A Clostridia-rich microbiota enhances bile acid excretion in diarrhea-predominant irritable bowel syndrome. J Clin Invest 2020;130:438-450.
    Pubmed KoreaMed CrossRef
  138. Wei W, Wang H, Zhang Y, et al. Faecal bile acids and colonic bile acid membrane receptor correlate with symptom severity of diarrhoea-predominant irritable bowel syndrome: a pilot study. Dig Liver Dis 2021;53:1120-1127.
    Pubmed CrossRef
  139. Wong BS, Camilleri M, Carlson P, et al. Increased bile acid biosynthesis is associated with irritable bowel syndrome with diarrhea. Clin Gastroenterol Hepatol 2012;10:1009-1015, e3.
    Pubmed KoreaMed CrossRef
  140. Shin A, Camilleri M, Vijayvargiya P, et al. Bowel functions, fecal unconjugated primary and secondary bile acids, and colonic transit in patients with irritable bowel syndrome. Clin Gastroenterol Hepatol 2013;11:1270-1275, e1.
    Pubmed KoreaMed CrossRef
  141. James SC, Fraser K, Young W, et al. Concentrations of fecal bile acids in participants with functional gut disorders and healthy controls. Metabolites 2021;11:612.
    Pubmed KoreaMed CrossRef
  142. Camilleri M, Acosta A, Busciglio I, et al. Effect of colesevelam on faecal bile acids and bowel functions in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther 2015;41:438-448.
    Pubmed KoreaMed CrossRef
  143. Wedlake L, A'Hern R, Russell D, Thomas K, Walters JR, Andreyev HJ. Systematic review: the prevalence of idiopathic bile acid malabsorption as diagnosed by SeHCAT scanning in patients with diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther 2009;30:707-717.
    Pubmed CrossRef
  144. Farooqui N, Elhence A, Shalimar. A current understanding of bile acids in chronic liver disease. J Clin Exp Hepatol 2022;12:155-173.
    Pubmed KoreaMed CrossRef
  145. Kullak-Ublick GA, Paumgartner G, Berr F. Long-term effects of cholecystectomy on bile acid metabolism. Hepatology 1995;21:41-45.
    Pubmed CrossRef
  146. Berr F, Stellaard F, Pratschke E, Paumgartner G. Effects of cholecystectomy on the kinetics of primary and secondary bile acids. J Clin Invest 1989;83:1541-1550.
    Pubmed KoreaMed CrossRef
  147. Clayton PT. Inborn errors of bile acid metabolism. J Inherit Metab Dis 1991;14:478-496.
    Pubmed CrossRef
  148. Bove KE, Heubi JE, Balistreri WF, Setchell KD. Bile acid synthetic defects and liver disease: a comprehensive review. Pediatr Dev Pathol 2004;7:315-334.
    Pubmed CrossRef
  149. Reichert MC, Hall RA, Krawczyk M, Lammert F. Genetic determinants of cholangiopathies: molecular and systems genetics. Biochim Biophys Acta Mol Basis Dis 2018;1864(4 Pt B):1484-1490.
    Pubmed CrossRef
  150. Felzen A, Verkade HJ. The spectrum of progressive familial intrahepatic cholestasis diseases: update on pathophysiology and emerging treatments. Eur J Med Genet 2021;64:104317.
    Pubmed CrossRef


This Article

e-submission

Archives

Aims and Scope