Next Article in Journal
Pharmacokinetic Comparison of Chitosan-Derived and Biofermentation-Derived Glucosamine in Nutritional Supplement for Bone Health
Next Article in Special Issue
Pomegranate Extract Affects Gut Biofilm Forming Bacteria and Promotes Intestinal Mucosal Healing Regulating the Crosstalk between Epithelial Cells and Intestinal Fibroblasts
Previous Article in Journal
Interplay between Intestinal Bacterial Communities and Unicellular Parasites in a Morbidly Obese Population: A Neglected Trinomial
Previous Article in Special Issue
Akkermansia muciniphila Colonization Alleviating High Fructose and Restraint Stress-Induced Jejunal Mucosal Barrier Disruption
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bile Salt Hydrolase-Competent Probiotics in the Management of IBD: Unlocking the “Bile Acid Code”

by
Raffaella Maria Gadaleta
1,
Marica Cariello
1,
Lucilla Crudele
1 and
Antonio Moschetta
1,2,*
1
Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
2
INBB National Instituto for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(15), 3212; https://doi.org/10.3390/nu14153212
Submission received: 12 July 2022 / Revised: 2 August 2022 / Accepted: 3 August 2022 / Published: 5 August 2022

Abstract

:
Bile acid (BA) species and the gut microbiota (GM) contribute to intestinal mucosa homeostasis. BAs shape the GM and, conversely, intestinal bacteria with bile salt hydrolase (BSH) activity modulate the BA pool composition. The mutual interaction between BAs and intestinal microorganisms also influences mucosal barrier integrity, which is important for inflammatory bowel disease (IBD) pathogenesis, prevention and therapy. High levels of secondary BAs are detrimental for the intestinal barrier and increase the intestinal inflammatory response and dysbiosis. Additionally, a lack of BSH-active bacteria plays a role in intestinal inflammation and BA dysmetabolism. Thus, BSH-competent bacteria in probiotic formulations are being actively studied in IBD. At the same time, studies exploring the modulation of the master regulator of BA homeostasis, the Farnesoid X Receptor (FXR), in intestinal inflammation and how this impacts the GM are gaining significant momentum. Overall, the choice of probiotic supplementation should be a peculiar issue of personalized medicine, considering not only the disease but also the specific BA and metabolic signatures of a given patient.

1. Introduction

The entire gastrointestinal tract hosts the gut microbiota (GM), currently considered as an additional essential organ for living organisms. The GM’s physiological functions range from fermentation of non-digestible dietary fibers, to production of critical metabolites and immune-modulatory functions. The whole GM consists of different types of microorganisms, including bacteria, virus, fungi and even parasites. In the last two decades, studies have consistently shown the involvement of a dysbalanced gut bacterial composition in the pathogenesis of several diseases, while the role of virus and fungi is just starting to emerge. Intestinal bacteria could be categorized in six main phyla: Bacteroidota and Firmicutes make up to 90% of the total bacterial composition, while the remaining part is composed of Proteobacteria, Fusobacteria, Actinobacteria and Verrucomicrobia [1]. The host organism and its resident microbiota are in a constant mutual communication, regulated by sophisticated rules. A disproportionate number, reduced diversity or unbalanced communities of microorganisms (namely dysbiosis) decrease the ability of the host to resist infections, absorb nutrients, synthesize vitamins, and/or carry out peculiar metabolic activities [2], thereby contributing to the onset and progression of disease of the gut–liver axis [3], inflammatory [4], metabolic or autoimmune conditions [5] and even neurodegenerative disorders [6,7,8]. Despite the ‘big unknown’ in the GM field, it is universally accepted that the microbiome is unique for each individual and substantial differences are present even in identical twins [9]. In fact, the GM composition is not only determined genetically, but it is profoundly impacted by multiple factors including epigenetics, nutrition, drug administration, physical exercise, and general lifestyle.
In terms of metabolic functions, the GM produces short-chain fatty acids (SCFAs) [10] and synthesize vitamins, e.g., vitamin K and constituents of the vitamin B [11,12,13]. SCFAs, propionate, acetate and butyrate are produced by fiber fermentation and are involved in several metabolic functions, ranging from supplying energy and trophic factors to colonic cells [14], to modulating T regulatory cell functions [15,16] and exerting crucial physio-metabolic effects on several organs, including the brain [17,18]. Within the metabolic functions, a powerful emerging line of communication in organisms’ physiology is the relationship between the bile acid (BA) pool and the GM, as species-specific strains of bacteria are capable of metabolizing BAs [19].

2. Bile Acids: Between Digestion and Metabolic Signaling

BAs are digestive amphipathic compounds produced in the liver from the oxidation of cholesterol in two multistep biosynthetic pathways—the classic and the alternative pathway—leading to the production of primary BAs, mainly chenodeoxyxholic (CDCA) and cholic acid (CA) in humans, and CA and beta-muricholic acid (β-MCA) in mice. The classic pathway, regulated by the rate-limiting enzyme cholesterol 7 alpha-hydroxylase (Cyp7a1), produces CDCA and CA, while the alternative pathway is responsible for the production of intermediate oxidized cholesterol metabolites that are afterwards converted mainly to CDCA in hepatocytes. Before active secretion of BAs into the canalicular lumen, primary BAs are conjugated at the C24 carboxyl group with the aminoacids taurine or glycine [20]. This amidation process is very efficient and is important to make BAs less hydrophobic and, therefore, less cytotoxic, and more readily secretable into bile. Thus, the majority of BA species present in bile are conjugated. Once produced, BAs are stored in the gallbladder and, after a meal, they are secreted into the duodenum, where they flow and aid the absorption of lipophilic nutrients. After their journey along the intestinal tract performing their biological functions, BAs are then reabsorbed via combined mechanisms of active transport in the distal ileum and passive absorption along the whole intestine and are then re-circulated via the portal vein to the liver [21]. This process, known as enterohepatic circulation, occurs between 4 and 12 times per day in humans. Approximately 5% of BAs enter the colon, where they are mainly biotransformed by the GM and lost through feces. BA biotransformation carried out by the GM include subsequent steps of deconjugation, oxidation, epimerization, and also esterification and desulfatation [22,23]. Unconjugated BAs are generally more hydrophobic than the corresponding conjugated forms, and secondary BAs are more hydrophobic than the corresponding primaries; thus, unconjugated BAs and secondary BAs are more likely to cause noxious effects. A very recent work has shown that deletion of the gene encoding the enzyme catalyzing BA conjugation, the bile acid-CoA: amino acid N-acyltransferase (BAAT), causes metabolic changes, such as elevated plasma transaminases, lower lipid-soluble vitamin levels, higher fecal lipid content and lower intestinal BA levels, very similar to those reported in human BAAT mutations, together with an altered intestinal microbial composition [24].
The physiological effects of BAs have historically been attributed to their capacity of forming fat-solubilizing micelles aimed at aiding the absorption of dietary fats. Nowadays, it has become clear that in addition to this physico-chemical function, BAs behave as hormone-like signaling molecules and also interact with the GM. Therefore, the size and composition of the circulating BA pool play a crucial role not only in digestion, but also in regulating metabolic activities and shaping the gut microbial community [25]. Primary BAs exploit their endocrine function via binding to their cognate Farnesoid X Receptor (FXR) [26,27,28]. FXR is a nuclear receptor and transcription factor, highly expressed in the small intestine and liver acting as the master regulator of BA homeostasis. In fact, once activated by BAs, FXR regulates tissue-specific gene networks, orchestrating their synthesis, transport and metabolism (reviewed in [29]) in the gut–liver axis. In particular, via a negative feedback loop, activated FXR inhibits de novo BA hepatic synthesis, via the transactivation of one of its main intestinal targets, the enterokinefibroblast growth factor 15/19 (FGF15/19 in mouse and human, respectively) [30]. FGF15/19 is then secreted into the portal circulation and travels to the liver, where it binds to its cognate co-receptor heterodimer consisting of the fibroblast growth receptor 4 (FGFR4) and Klotho-beta (KLB), initiating a phosphorylation signaling cascade, ultimately resulting in CYP7A1 inhibition [31,32,33], hence suppressing de novo hepatic BA synthesis. In addition to this well-characterized pathway, BA-activated FXR also modulates BA cytotoxicity, aiming at creating a balanced BA pool that in physiological conditions and under proper nutritional cues does not harm the intestinal cell physiology. BA-activated FXR has also been shown to have anti-inflammatory [34,35,36] and antitumorigenic properties [37,38,39], and actively participates in the maintenance of intestinal barrier integrity [34,40]. Additionally, FGF19 was shown to have similar functions in mice and induce a reshape of the gut microbial community in the presence of an inflammatory trigger [41]. The ability of FXR and FGF19 to shape the circulating BA pool size and composition is critical for the relation BAs have with the GM community.

3. Bile Acid Deconjugationand BSH-Competent Microorganisms

The integrated metabolism of the BA pool and GM is among the most complex transgenomic biochemical interactions between the host and symbiotic microorganisms inhabiting it [42]. If one considers BA modulatory activities on cholesterol, triglycerides, glucose and energy homeostasis (reviewed in [43]), it is intuitive that gut bacteria-dependent BA biotransformation have crucial biological implications. Primary BAs synthesized and conjugated to glycine or taurine before secretion into the bile canaliculi are then transformed into secondary BAs, mainly deoxycholic acid (DCA), lithocolic acid (LCA) and β-muri-deoxycholic acid (β-MDCA; in mice) via bacteria with a peculiar activity: the bile salt hydrolase (BSH) activity [22,44,45]. The BSH catalyzes the “gateway” reaction in the bacterial metabolism of conjugated BAs hydrolyzing the C-24 N-acyl bond conjugating BA to glycine or taurine and is a prerequisite for the subsequent 7α/β-dehydroxylation [44,46,47,48] and 7α-dehydrogenation, ultimately resulting in BAs deconjugation and epimerization. GM-dependent BA metabolization mostly occurs in the terminal ileum and proximal colon, the focal colonization site of bacterial strains with BSH activity [22].
BSH activity was found in all major bacterial groups and archaeal species in the human gut [48]. Gram-positive gut bacteria, including strains of Lactobacilli [49,50], Bifidobacteria [51,52,53], Clostridium [54,55] and Enterococcus [56], display a highly heterogeneous distribution of BSH. On the contrary, BSH in Gram-negative bacterial strains has only been found in members of the phylum Bacteroidetes [48,57]. Recently published next-generation sequencing data on the human GM isolated from eleven populations from six continents unfolded the prevalence of BSH in 12 different phyla, including Bacteroidetes and Firmicutes [58]. Several strains, including Clostridium scindens, Clostridium hiranonis, and Clostridium hylemonae, have been discovered to be involved in the production of LCA and DCA in humans [23]. Lactobacilli [44,59], Bifidobacterium longum [53,60] and Pediococcus [61] have been shown to be BSH-competent species. Furthermore, pathogens such as Clostridium perfringens [62], Listeria monocytogenes [63], Brucella abortus [64], Enterococcus faecalis [65], and Xanthomonas [66] present with BSH activity. Last but not least, interesting findings have shown that bacteria isolated from marine sediments (Methanosarcina acetovorans) [48] and Antartican lakes (Planococcus antarticus) [67] display BSH activity.
In addition to the metabolic activities due to BSH-competent bacteria, there are other enzymatic biotransformations performed by gut microbes, such as the 7α/β-dehydroxylation, pyridine nucleotide-dependent hydroxysteroid dehydrogenization (operated by HSDHs enzymes), and conjugation of the cholate backbone with the aminoacids tyrosine, leucine and phenylalanine. The 7α/β-dehydroxylation is a multistep biotransformation carried out on free BAs and performed by a range of BA-inducible (bai) and bai-like genes [68]. Bai genes convert CA and CDCA to DCA and LCA, respectively, and have been identified in multiple species, mostly in Faecali catena contorta S122, Clostridium hiranonis and Clostridium scinden in humans [69] and Eggerthella lenta [70,71]. HSDHs performs the oxidation/reduction of hydroxy groups at the carbon atoms in positions 3, 7, and 12 of BAs [72], while the amide conjugation of cholate with aminoacids was recently found to be associated with Clostridium bolteae in mice fed a high-fat diet [73]; however, it is not currently clear what the function of such modified BA is.
FXR is differentially activated by the different BA species and their metabolites. Chenodeoxycholic Acid (CDCA) is the major FXR activator, followed by deoxycholic acid (DCA), lithocolic acid (LCA) and cholic acid (CA) [74]. Therefore, intestinal microorganisms not only act directly on BA species, but they also indirectly influence FXR transcriptional activity via the modulation of BA metabolism [75]. As a consequence, BA transformation by BSH-competent bacteria shapes the magnitude of BA-induced FXR activation in the gut [75,76,77,78]. Specifically, the major metabolic action of this specific group of bacteria is therefore to deconjugate intraluminal BAs, thus reducing the amount of conjugated BA (T/GCA and T/GCDCA) that can be absorbed by the enterocytes via the intestinal bile acid transporter (IBAT). These events translate into a de-activation of intestinal FXR (due to lower amounts of intracellular ligands) coupled with a significant increased fecal BA loss (up to 3 fold) (Figure 1). In this respect, one could speculate that the role of BSH-competent enteric bacterial would be similar to that of cholestyramine, which is able to bind intraluminal BA and increase their fecal release. Intriguingly, resins such ascholestyramine are nowadays used in clinic in BA-induced diarrhea in IBD and eventually BSH probiotics could complement this therapeutic strategy.
We have previously shown that enrichment of the gut milieu with BSH-competent species, such as Lactobacilli and Bifidobacteria, with VSL#3 probiotic administration in mice limits the availability of efficient FXR ligands and produces significant alterations in BA homeostasis that translates into enhanced fecal BA excretion, hepatic BA synthesis and biliary output [79], similar to what happens in mice receiving the BA sequestrant coleselavam [80]. Intriguingly, this phenomenon is abrogated in both Fxr- and Fgf15-null mice [79], indicating that the activation of the Fxr-Fgf15 duo is required for probiotic BSH-competent bacteria to promote fecal unconjugated BA loss and hepatic BA synthesis. For all these reasons, understanding how bacterial species affects BA metabolism and the FXR-FGF15/19 duo in the gut liver axis is a very relevant question gaining significant momentum from studies demonstrating that dysbiosis and BA dysmetabolism are associated with intestinal diseases.

4. Bile Acid-Dependent Shaping of Intestinal Microbial Community

The mutual interaction between the bileome and intestinal microorganisms has a direct influence on the integrity of the intestinal epithelial barrier. In fact, BAs flowing along the intestine control bacterial overgrowth and protect against intestinal mucosal damage. In particular, high levels of hydrophobic BAs have a direct antimicrobial activity mainly causing damage to the bacterial membrane [81,82]. On the one hand, Gram-negative bacteria, especially those belonging to Bacteroides, have been found to be more sensitive to free BAs [70,83]; on the other hand, Gram-positive bacteria seem to be more sensitive to tauro- and glyco-conjugated and primary BAs [59].
The tight interconnection between the GM and BA metabolism goes beyond BA intrinsic antimicrobial properties, as BAs indirectly shape the composition of GM by promoting the expression of genes fostering innate defense via FXR activation [40]. A lower level of BAs as a consequence of clinically and/or experimentally induced liver injuries can trigger intestinal bacterial overgrowth [84,85,86]. For example, surgical closure of the common bile duct in rodents results in bacterial overgrowth and translocation across the intestinal epithelial barrier, disruption of its integrity, and systematic infection that can be reversed by BA administration [87,88]. In the presence of an obstruction of bile flow, abnormal hepatic BA accumulation occurs and intestinal FXR signaling is abrogated [40]. In addition, devoiding the intestinal lumen of BA has a detrimental effect on the intestinal microbial community and causes mucosal injury [89]. Administration of BAs to both rodents and humans in conditions of biliary obstruction [88,90,91,92], as well as FXR activation [40] or FGF19 administration [41] in the presence of intestinal inflammatory triggers, counteracts intestinal mucosal damage and bacterial overgrowth and translocation. Rcent data from our group show that in a murine model of obstructed BA flow, FXR activation by its novel specific ligand TC-100 prevents intestinal mucosal damage, goblet cells and mucus depletion, and increases the expression of tight junctions by the mutual modulation of the GM and BA pool size and composition [93]. Both the peculiar enrichment of Akkermansia muciniphila and associated reduction in CA levels contribute to these beneficial effects [93]. BA-dependent activation of the Fxr-Fgf15/19 axis and the consequent inhibition of Cyp7a1, hence BA synthesis, is affected in rodents treated with antibiotics, germ-free and gnotobiotic animals [75,94,95,96,97,98] and mice lacking Fxr display ileal bacterial overgrowth [40] and a compromised epithelial barrier already at basal state [34,40].
Administration of the FXR-specific agonist obeticholic acid (OCA) in healthy volunteers was shown to inhibit endogenous BA synthesis while concomitantly enriching the GM with a number of Gram-positive strains, namely Streptococcus thermophilus, Lactobacillus casei and paracasei, Bifidobacterium breve, and Lactococcus lactis, boosting the notion that changes in the endogenous BA pool size and composition result in an FXR-dependent reshaping of the gut microbial composition [99]. In a mouse model of spontaneous colorectal cancer onset, the Apcmin/+ mouse model, administration of a diet supplemented with CA increased the abundance of opportunistic pathogens such as Prevotella and Desulfovibrio while decreasing the presence of beneficial bacteria, including Ruminococcus, Lactobacillus, and Roseburia [100]. A similar diet administration in rats, induced alterations in the composition of the GM; the phylum of Firmicutes predominated at the expense of Bacteroidetes and an expansion of several strains in the classes Clostridia and Erysipelotrichi occurred [76]. Concomitantly, the administered CA was efficiently transformed into DCA by a bacterial 7α-dehydroxylation reaction [76], suggesting that DCA could boost the reshaping of the GM composition due to its powerful antimicrobial activity. In line with this, mice fed with a diet supplemented with DCA display a lower abundance of BSH-competent Lactobacillus, Clostridium XI, and Clostridium XIV and a concomitant enriched presence of Parabacteroides and Bacteroides compared to controls [101]. Taken together, these data demonstrate that BA supplementation reshapes the composition of gut microbiota.

5. Bile Salt Hydrolase Activity and Probiotics

Dietary supplementation with probiotics was demonstrated to modify not only hosts’ BA pool but also their metabolism. BSH-competent bacteria in probiotic formulations are being actively studied for their BA composition-modifying properties and their ability to reduce serum cholesterol levels [102]. In fact, BSH-dependent BA deconjugation decreases the recycling of BAs promoting their de novo synthesis, thus reducing serum cholesterol levels [102]. Additionally, bacteria with BSH activity metabolize aminoacids produced from deconjugation as carbon, nitrogen and energy sources [44]. Using germ-free and conventionally raised mouse models, Joyce et al. demonstrated that gastrointestinal expression of BSH promotes BA deconjugation with concomitant alteration of the host’s immune homeostasis, energy metabolism and circadian rhythm [103]. In conventionally raised mice, BSH activity reduced weight gain, serum cholesterol, and hepatic triglyceride levels [103]. Expression of cloned BSH enzymes in the gastrointestinal tract of gnotobiotic or conventionally raised mice exhibited a gene expression profile characterized by Clock and Arntl induction, genes involved in circadian rhythm regulation and energy expenditure [103]. Mukherji et al. observed that the GM, Toll-like receptor expression on epithelial cells and circadian rhythm are tightly connected and perturbations of this interaction promote the development of a pre-diabetic syndrome [104]. Administration of the BSH-competent Lactobacillus reuteri NCIMB 30242® in human healthy volunteers improves unconjugated plasma BA profile highlighting the ability of this specific probiotic to modulate BA metabolism in humans [102]. Several studies conducted in mice and humans demonstrated the involvement of probiotics with Lactobacillus species such as Lactobacillus fermentum K73 and Lactobacillus plantarum 299v in the reduction of body weight and cholesterol levels due to their BSH competence [102,105]. Interestingly, it was shown that the yeast strain Saccharomyces Boulardii has BSH activity in vitro [106]. Daily administration of this yeast in hamsters fed a high-cholesterol diet decreased total plasma cholesterol levels and modified the GM composition [107]. In humans, administration of Saccaromices Boulardii for 8 weeks reduced remnant lipoprotein concentration, a predictive biomarker and potential therapeutic target in the prevention/treatment of coronary artery disease [108]. Additionally, mice administered with the probiotic VSL#3, containing the BSH-competent Lactobacillus acidophilus and Bifidobacterium infantis strains, modulates the BA profile by upregulating hepatic de novo BA synthesis [79] and their fecal excretion [79].

6. BSH-Competent Bacteriaand Probiotics Administrationin Inflammatory Bowel Disease

Patients affected by inflammatory bowel diseases (IBD) display low levels of BSH activity in the gut microbiota [109,110]. IBD is a chronic inflammatory condition of the gastrointestinal tract characterized by a dysregulation of the gut mucosal immune functions and a dysbiotic GM occurring in genetically susceptible hosts. IBD encompasses two major phenotypes: ulcerative colitis (UC) and Crohn’s disease (CD), which present with overlapping symptoms, despite being two distinct pathologies.
Previously published data have shown how secondary BAs are detrimental and co-responsible for the disruption of intestinal barrier integrity, increase of the inflammatory response and dysbiosis [101,111,112]. Interestingly, rectal instillation of DCA/LCA in different murine models resulted in anti-inflammatory benefits, partially due to TGR5 signaling [113], indicating a dual pro- and anti-inflammatory effect of DCA in different experimental contexts and administration routes. This strongly suggests that caution should be taken when translating mouse data to IBD patients’ bedside.
Analysis of data collected on an IBD multiomics database (The Integrative Human Microbiome Project), revealed that BAs are a prominent component of the network in relation to changes in the microbiome [114]. In particular, primary BAs (specifically CA, TCA and GCA) levels were higher, while secondary BAs levels were lower [114]. This was associated with an enrichment of Roseburia and strongly indicated a lack of BSH-competent bacteria IBD-related dysbiosis [114]. Franzosa et al. also demonstrated that levels of primary BAs were higher in IBD patients and potentially associated with the coherent microbial groups, while secondary BA levels were lower in CD patients [115]. In line with this, patients with UC pouch displayed low levels of secondary BAs and high levels of CDCA and this was consistently associated with lower levels of Ruminococcaceae and bai genes [113], indicating once again a lack of BSH-competent bacteria, hence a dysbiosis-dependent disturbance of BA metabolization. Beside unravelling the intertwined relation between BA species and BSH-competent bacteria in IBD pathogenesis, its clinical and therapeutic relevance is also being actively studied. Omics data revealed that specific GM composition in IBD patients predicts early remission response to anti-cytokine and anti-integrin therapies, via gut bacterial clusterization associated with secondary BAs enrichment [71]. Probiotic supplementation in these patients may represent a useful tool for the prevention, treatment and remission of the acute phase. In IBD, probiotics have a beneficial impact when are able to modulate the immune response, gut barrier integrity and GM composition [116]. Several studies have been conducted in animal models and IBD patients to find suitable probiotics in IBD, but a universal consensus in not available yet.
Data on the GM composition of IBD patients indicated a lower level of Firmicutes and a higher level of Proteobacteria [117]. Consistent with this, recent data have shown that BSH-competent Proteobacteria are increased, while BSH-competent Firmicutes are decreased in IBD patients [118]. In vitro studies and mouse models have shown that Faecalibacterium prausnitzii and Lactobacillus rhamnosus CNCM I-3690 ameliorate gut barrier integrity by increasing the expression of occludin and cadherin proteins [119,120]. Experimental colitis caused by Trinitrobenezenesulphonic acid (TNBS), increased Escherichia coli and Clostridium strains and reduced Bifidobacterium and Lactobacillus populations [121]. On the contrary, Bifidobacterium supplementation in murine experimental colitis, seems to have beneficial effects and decrease inflammation [122,123,124,125]. Bifidobacterium infantis administration reduced symptoms of colitis such as colonic edema and weight-loss, protected the epithelial cell layer, prevented goblet cell loss and modulated cytokine levels, specifically increasing the anti-inflammatory cytokine IL-10 and decreasing the pro-inflammatory one IL-1β [123,125,126]. In Sodium Dextran Sulphate (DSS)-induced murine colitis, administration of Bifidobacterium animalis and Bifidobacterium longum ameliorated inflammatory symptoms, preserved the colonic structure and decreased intestinal epithelial cell apoptosis and TNF-α levels [122,124]. Furthermore, in a TNBS-induced colitis mouse model, the influence of Bifidobacterium animalis and Lactobacillus reuteri administration on the immune system was studied [127]. Authors observed that Bifidobacterium animalis beneficial effects on the host were linked to the bone marrow-derived dendritic cell development and IL-17A secretion, whereas Lactobacillus reuteri administration promoted tolerogenic dendritic cells and Tregs population development [127]. Interestingly, two different formulations of VSL#3 have shown divergent metabolic and anti-inflammatory activities in mice subjected to DSS and TNBS colitis [128].
Clinical trials are ongoing in IBD patients with the aim to evaluate the effects of probiotics on the disease. In UC patients, several studies indicated that different Bifidobacterium strains are able to reduce pro-inflammatory interleukins and C-reactive protein (CRP) levels, sustaining the remission phase [129,130,131,132,133]. On the other hand, in CD patients, the administration of Saccaromyces boulardii promoted the maintenance of the remission phase [134]. Probio-Tec AB25, a cocktail of Lactobacillus acidophilus strain LA-5 and Bifidobacteriumanimalis subsp. lactis BB12, was administered to 32 UC patients, with no beneficial effects in maintaining remission [135]. VSL#3 is among the most common probiotic mixtures of proven efficiency on human IBD, and it contains different Lactobacilli, Bifidobacteria and Streptococcus thermophilus strains [136,137]. In a double-blind placebo-controlled trial, patients with the active form of UC received VSL#3 twice daily for 12 weeks. Authors observed remission characterized by the reduction of stool frequency and rectal bleeding [136]. In another clinical trial, VSL#3 was used as an adjuvant added to patients’ pharmaceutical treatment for 8 weeks. A reduction in rectal bleeding was observed, although no differences were found in endoscopic inflammatory scores, highlighting the importance of long-term use of probiotics [137]. Furthermore, the administration of VSL#3 in children with UC determined a significant remission of patients due to a reduction of IFN-γ and TNF-α levels together with a modulation of the GM [138]. Conversely, VSL#3 treatment in CD patients does not seem to have clear benefits [139], although further research is warranted. In these patients, supplementation with Faecalibacterium prausnitzii, instead, exhibited anti-inflammatory effects [140]. Table 1 and Table 2 summarize the ongoing and/or completed clinical trials in IBD patients administered with either probiotics or microbial metabolites.

7. Conclusions and Discussion

BA species and intestinal microorganisms both contribute to intestinal fitness, are tightly interconnected and communicate directly with each other and indirectly via mainly FXR and related signaling pathways. Despite the evident association between BSH-competent bacteria and BA pool composition, metagenomic analysis of IBD patients’ GM revealed that not all BSH-competent species are created equal and the fact that any GM metabolic activity is strainspecific should always be remembered. Therefore, further studies on probiotics administration and their effect on gut microbial reshaping and consequent BA metabolism modulation are needed. At the same time, these studies should be paralleled by the generation of data on how microbial reshaping can be achieved dueto the modulation of the BA pool size and composition, as it occurs when FXR- or FGF19-based drugs are used. Clinical use of probiotics containing BSH-competent species is currently ongoing in patients with IBD, pouchitis and diarrhea and could be also relevant in patients with leaky gut secondary to liver disease (such as intrahepatic cholestasis) or colorectal cancer. On the other hand, a few considerations about limitations and potential adverse effects related to the indiscriminate use of probiotics resulting in high levels of unconjugated bile acids, lipid malabsorption and steatorrhea should be made. Elevated concentrations of DCA and LCA, produced from unconjugated BAs, are considered a risk factor for colorectal cancer [149]. Experimental evidence hasdemonstrated that BSH activity promotes gut colonization by pathogenic bacterial strains such as Listeria monocytogenes and Brucella abortus [63]. Moreover, mice and humans display different BA profiles characterized by high levels of tauro-conjugated BAs and high levels of glyco-conjugated BAs, respectively [59,74]. In this scenario, previous data on the species-specific difference affecting the translation of data from mice to humans and confounding the selection of probiotics are actively being validated in clinical trials. The field is new and a greatereffort should be made in integrating the enormous amounts of data generated by all the available omic platforms. Overall, the choice of probiotic supplementation should be considered a peculiar issue of the personalized medicine, considering not only the disease but also the specific BA and metabolic signature of a given patient.

Author Contributions

Conceptualization, A.M. and R.M.G.; writing—original draft preparation, R.M.G. and M.C.; writing, L.C.; visualization, R.M.G.; supervision, A.M.; funding acquisition, A.M. All authors have read and agreed to the published version of the manuscript.

Funding

A.M. was funded by the Italian Association for Cancer Research—AIRC IG 2019 Id 23239; MIUR-PRIN 2017 Cod. 2017 J3E2W2.

Conflicts of Interest

Authors have no conflict of interest to declare.

References

  1. Rinninella, E.; Cintoni, M.; Raoul, P.; Lopetuso, L.R.; Scaldaferri, F.; Pulcini, G.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. Food Components and Dietary Habits: Keys for a Healthy Gut Microbiota Composition. Nutrients 2019, 11, 2393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pflughoeft, K.J.; Versalovic, J. Human Microbiome in Health and Disease. Annu. Rev. Pathol. 2012, 7, 99–122. [Google Scholar] [CrossRef] [PubMed]
  3. Ohtani, N.; Kawada, N. Role of the Gut-Liver Axis in Liver Inflammation, Fibrosis, and Cancer: A Special Focus on the Gut Microbiota Relationship. Hepatol. Commun. 2019, 3, 456–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Macia, L.; Thorburn, A.N.; Binge, L.C.; Mariño, E.; Rogers, K.E.; Maslowski, K.; Vieira, A.; Kranich, J.; Mackay, C.R. Microbial influences on epithelial integrity and immune function as a basis for inflammatory diseases. Immunol. Rev. 2012, 245, 164–176. [Google Scholar] [CrossRef] [PubMed]
  5. Jiao, Y.; Wu, L.; Huntington, N.D.; Zhang, X. Crosstalk between Gut Microbiota and Innate Immunity and Its Implication in Autoimmune Diseases. Front. Immunol. 2020, 11, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cerdó, T.; Diéguez, E.; Campoy, C. Impact of gut microbiota on neurogenesis and neurological diseases during infancy. Curr. Opin. Pharmacol. 2020, 50, 33–37. [Google Scholar] [CrossRef]
  7. Ghaisas, S.; Maher, J.; Kanthasamy, A. Gut microbiome in health and disease: Linking the microbiome–gut–brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2016, 158, 52–62. [Google Scholar] [CrossRef] [Green Version]
  8. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.-F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflamm. 2019, 16, 53. [Google Scholar] [CrossRef] [Green Version]
  9. Berry, S.E.; Valdes, A.M.; Drew, D.A.; Asnicar, F.; Mazidi, M.; Wolf, J. Human postprandial responses to food and potential for precision nutrition. Nat. Med. 2020, 26, 964–973. [Google Scholar] [CrossRef]
  10. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [Green Version]
  11. Albert, M.J.; Mathan, V.I.; Baker, S.J. Vitamin B12 synthesis by human small intestinal bacteria. Nature 1980, 283, 781–782. [Google Scholar] [CrossRef] [PubMed]
  12. LeBlanc, J.G.; Milani, C.; de Giori, G.S.; Sesma, F.; van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotechnol. 2013, 24, 160–168. [Google Scholar] [CrossRef] [PubMed]
  13. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  14. Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S. Microbiota and metabolic diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef] [PubMed]
  15. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
  16. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
  17. Dalile, B.; Van, O.L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
  18. Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef]
  19. Wahlström, A.; Sayin, S.I.; Marschall, H.-U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [Green Version]
  20. Falany, C.N.; Johnson, M.R.; Barnes, S.; Diasio, R.B. Glycine and taurine conjugation of bile acids by a single enzyme. Molecular cloning and expression of human liver bile acid CoA:amino acid N-acyltransferase. J. Biol. Chem. 1994, 269, 19375–19379. [Google Scholar] [CrossRef]
  21. Chiang, J.Y. Bile acid metabolism and signaling. Compr. Physiol. 2013, 3, 1191–1212. [Google Scholar] [PubMed] [Green Version]
  22. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006, 47, 241–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ridlon, J.M.; Harris, S.C.; Bhowmik, S.; Kang, D.-J.; Hylemon, P.B. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes 2016, 7, 22–39. [Google Scholar] [CrossRef] [Green Version]
  24. Alrehaili, B.D.; Lee, M.; Takahashi, S.; Novak, R.; Rimal, B.; Boehme, S.; Trammell, S.A.J.; Grevengoed, T.J.; Kumar, D.; Alnouti, Y.; et al. Bile acid conjugation deficiency causes hypercholanemia, hyperphagia, islet dysfunction, and gut dysbiosis in mice. Hepatol. Commun. 2022. [Google Scholar] [CrossRef] [PubMed]
  25. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B.; Bajaj, J.S. Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 2014, 30, 332–338. [Google Scholar] [CrossRef] [Green Version]
  26. Makishima, M.; Okamoto, A.Y.; Repa, J.J.; Tu, H.; Learned, R.M.; Luk, A.; Hull, M.V.; Lustig, K.D.; Mangelsdorf, D.J.; Shan, B. Identification of a Nuclear Receptor for Bile Acids. Science 1999, 284, 1362–1365. [Google Scholar] [CrossRef]
  27. Parks, D.J.; Blanchard, S.G.; Bledsoe, R.K.; Chandra, G.; Consler, T.G.; Kliewer, S.A.; Stimmel, J.B.; Willson, T.M.; Zavacki, A.M.; Moore, D.D.; et al. Bile Acids: Natural Ligands for an Orphan Nuclear Receptor. Science 1999, 284, 1365–1368. [Google Scholar] [CrossRef]
  28. Wang, H.; Chen, J.; Hollister, K.; Sowers, L.C.; Forman, B.M. Endogenous Bile Acids Are Ligands for the Nuclear Receptor FXR/BAR. Mol. Cell 1999, 3, 543–553. [Google Scholar] [CrossRef]
  29. Gadaleta, R.M.; Cariello, M.; Sabbà, C.; Moschetta, A. Tissue-specific actions of FXR in metabolism and cancer. Biochim. Biophys. Acta BBA Mol. Cell Biol. Lipids 2015, 1851, 30–39. [Google Scholar] [CrossRef]
  30. Inagaki, T.; Choi, M.; Moschetta, A.; Peng, L.; Cummins, C.L.; McDonald, J.G.; Luo, G.; Jones, S.A.; Goodwin, B.; Richardson, J.A.; et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005, 2, 217–225. [Google Scholar] [CrossRef] [Green Version]
  31. Kim, I.; Ahn, S.-H.; Inagaki, T.; Choi, M.; Ito, S.; Guo, G.L.; Kliewer, S.A.; Gonzalez, F.J. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J. Lipid Res. 2007, 48, 2664–2672. [Google Scholar] [CrossRef] [Green Version]
  32. Kong, B.; Wang, L.; Chiang, J.Y.; Zhang, Y.; Klaassen, C.D.; Guo, G.L. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology 2012, 56, 1034–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Repa, J.J.; Mangelsdorf, D.J. The Role of Orphan Nuclear Receptors in the Regulation of Cholesterol Homeostasis. Annu. Rev. Cell Dev. Biol. 2000, 16, 459–481. [Google Scholar] [CrossRef] [PubMed]
  34. Gadaleta, R.M.; Van Erpecum, K.J.; Oldenburg, B.; Willemsen, E.C.L.; Renooij, W.; Murzilli, S.; Klomp, L.W.J.; Siersema, P.D.; Schipper, M.E.; Danese, S.; et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 2011, 60, 463–472. [Google Scholar] [CrossRef] [PubMed]
  35. Gadaleta, R.M.; Oldenburg, B.; Willemsen, E.C.; Spit, M.; Murzilli, S.; Salvatore, L. Activation of bile salt nuclear receptor FXR is repressed by pro-inflammatory cytokines activating NF-kappaB signaling in the intestine. Biochim. Biophys. Acta 2011, 1812, 851–858. [Google Scholar] [CrossRef]
  36. Vavassori, P.; Mencarelli, A.; Renga, B.; Distrutti, E.; Fiorucci, S. The Bile Acid Receptor FXR Is a Modulator of Intestinal Innate Immunity. J. Immunol. 2009, 183, 6251–6261. [Google Scholar] [CrossRef] [Green Version]
  37. Cariello, M.; Zerlotin, R.; Pasculli, E.; Piccinin, E.; Peres, C.; Porru, E.; Roda, A.; Gadaleta, R.M.; Moschetta, A. Intestinal FXR Activation via Transgenic Chimera or Chemical Agonism Prevents Colitis-Associated and Genetically-Induced Colon Cancer. Cancers 2022, 14, 3081. [Google Scholar] [CrossRef]
  38. Maran, R.R.; Thomas, A.; Roth, M.; Sheng, Z.; Esterly, N.; Pinson, D.; Gao, X.; Zhang, Y.; Ganapathy, V.; Gonzalez, F.J.; et al. Farnesoid X Receptor Deficiency in Mice Leads to Increased Intestinal Epithelial Cell Proliferation and Tumor Development. J. Pharmacol. Exp. Ther. 2009, 328, 469–477. [Google Scholar] [CrossRef] [Green Version]
  39. Modica, S.; Murzilli, S.; Salvatore, L.; Schmidt, D.R.; Moschetta, A. Nuclear Bile Acid Receptor FXR Protects against Intestinal Tumorigenesis. Cancer Res. 2008, 68, 9589–9594. [Google Scholar] [CrossRef] [Green Version]
  40. Inagaki, T.; Moschetta, A.; Lee, Y.-K.; Peng, L.; Zhao, G.; Downes, M.; Yu, R.T.; Shelton, J.M.; Richardson, J.A.; Repa, J.J.; 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. [Google Scholar] [CrossRef] [Green Version]
  41. Gadaleta, R.M.; Garcia-Irigoyen, O.; Cariello, M.; Scialpi, N.; Peres, C.; Vetrano, S. Fibroblast Growth Factor 19 modulates intestinal microbiota and inflammation in presence of Farnesoid X Receptor. eBioMedicine 2020, 54, 102719. [Google Scholar] [CrossRef] [PubMed]
  42. Turroni, F.; Ribbera, A.; Foroni, E.; Van Sinderen, D.; Ventura, M. Human gut microbiota and bifidobacteria: From composition to functionality. Antonie Leeuwenhoek 2008, 94, 35–50. [Google Scholar] [CrossRef] [PubMed]
  43. Massafra, V.; van Mil, S.W.C. Farnesoid X receptor: A “homeostat” for hepatic nutrient metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 45–59. [Google Scholar] [CrossRef] [PubMed]
  44. Begley, M.; Hill, C.; Gahan, C.G. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 2006, 72, 1729–1738. [Google Scholar] [CrossRef] [Green Version]
  45. Patel, A.K.; Singhania, R.R.; Pandey, A.; Chincholkar, S.B. Probiotic Bile Salt Hydrolase: Current Developments and Perspectives. Appl. Biochem. Biotechnol. 2009, 162, 166–180. [Google Scholar] [CrossRef]
  46. Batta, A.K.; 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. [Google Scholar] [CrossRef]
  47. Geng, W.; Lin, J. Bacterial bile salt hydrolase: An intestinal microbiome target for enhanced animal health. Anim. Health Res. Rev. 2016, 17, 148–158. [Google Scholar] [CrossRef] [Green Version]
  48. Jones, B.V.; Begley, M.; Hill, C.; Gahan, C.G.M.; Marchesi, J.R. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc. Natl. Acad. Sci. USA 2008, 105, 13580–13585. [Google Scholar] [CrossRef] [Green Version]
  49. Bateup, J.M.; McConnell, M.A.; Jenkinson, H.F.; Tannock, G.W. Comparison of Lactobacillus strains with respect to bile salt hydrolase activity, colonization of the gastrointestinal tract, and growth rate of the murine host. Appl. Environ. Microbiol. 1995, 61, 1147–1149. [Google Scholar] [CrossRef] [Green Version]
  50. Corzo, G.; Gilliland, S.E. Bile Salt Hydrolase Activity of Three Strains of Lactobacillus acidophilus. J. Dairy Sci. 1999, 82, 472–480. [Google Scholar] [CrossRef]
  51. Grill, J.; Schneider, F.; Crociani, J.; Ballongue, J. Purification and Characterization of Conjugated Bile Salt Hydrolase from Bifidobacterium longum BB536. Appl. Environ. Microbiol. 1995, 61, 2577–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kim, G.-B.; Yi, S.-H.; Lee, B.H. Purification and Characterization of Three Different Types of Bile Salt Hydrolases from Bifidobacterium Strains. J. Dairy Sci. 2004, 87, 258–266. [Google Scholar] [CrossRef] [Green Version]
  53. Tanaka, H.; Hashiba, H.; Kok, J.; Mierau, I. Bile Salt Hydrolase of Bifidobacterium longum—Biochemical and Genetic Characterization. Appl. Environ. Microbiol. 2000, 66, 2502–2512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kishinaka, M.; Umeda, A.; Kuroki, S. High concentrations of conjugated bile acids inhibit bacterial growth of Clostridium perfringens and induce its extracellular cholylglycine hydrolase. Steroids 1994, 59, 485–489. [Google Scholar] [CrossRef]
  55. Masuda, N. Deconjugation of bile salts by Bacteroids and Clostridium. Microbiol. Immunol. 1981, 25, 1–11. [Google Scholar] [CrossRef]
  56. Franz, C.M.; Specht, I.; Haberer, P.; Holzapfel, W.H. Bile Salt Hydrolase Activity of Enterococci Isolated from Food: Screening and Quantitative Determination. J. Food Prot. 2001, 64, 725–729. [Google Scholar] [CrossRef]
  57. Yao, L.; Seaton, S.C.; Ndousse-Fetter, S.; Adhikari, A.A.; DiBenedetto, N.; Mina, A.; Banks, A.S.; Bry, L.; Devlin, A.S. A selective gut bacterial bile salt hydrolase alters host metabolism. eLife 2018, 7, e37182. [Google Scholar] [CrossRef]
  58. Song, Z.; Cai, Y.; Lao, X.; Wang, X.; Lin, X.; Cui, Y.; Kalavagunta, P.K.; Liao, J.; Jin, L.; Shang, J.; et al. Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. Microbiome 2019, 7, 9. [Google Scholar] [CrossRef] [Green Version]
  59. Begley, M.; Gahan, C.G.; Hill, C. The interaction between bacteria and bile. FEMS Microbiol. Rev. 2005, 29, 625–651. [Google Scholar] [CrossRef] [Green Version]
  60. Jarocki, P.; Targoński, Z. Genetic Diversity of Bile Salt Hydrolases among Human Intestinal Bifidobacteria. Curr. Microbiol. 2013, 67, 286–292. [Google Scholar] [CrossRef] [Green Version]
  61. Liang, L.; Yi, Y.; Lv, Y.; Qian, J.; Lei, X.; Zhang, G. A Comprehensive Genome Survey Provides Novel Insights into Bile Salt Hydrolase (BSH) in Lactobacillaceae. Molecules 2018, 23, 1157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Coleman, J.P.; Hudson, L.L. Cloning and characterization of a conjugated bile acid hydrolase gene from Clostridium perfringens. Appl. Environ. Microbiol. 1995, 61, 2514–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Dussurget, O.; Cabanes, D.; Dehoux, P.; Lecuit, M.; Buchrieser, C.; Glaser, P. 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. [Google Scholar] [CrossRef]
  64. Delpino, M.V.; Marchesini, M.I.; Estein, S.M.; Comerci, D.J.; Cassataro, J.; Fossati, C.A.; Baldi, P.C. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shankar, N.; Baghdayan, A.S.; Gilmore, M.S. Modulation of virulence within a pathogenicity island in vancomycin-resistant Enterococcus faecalis. Nature 2002, 417, 746–750. [Google Scholar] [CrossRef]
  66. Dean, M.; Cervellati, C.; Casanova, E.; Squerzanti, M.; Lanzara, V.; Medici, A.; De Laureto, P.P.; Bergamini, C.M. 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. [Google Scholar] [CrossRef] [Green Version]
  67. Margolles, A.; Gueimonde, M.; Sánchez, B. Genome Sequence of the Antarctic Psychrophile Bacterium Planococcusantarcticus DSM 14505. J. Bacteriol. 2012, 194, 4465. [Google Scholar] [CrossRef] [Green Version]
  68. Funabashi, M.; Grove, T.L.; Wang, M.; Varma, Y.; McFadden, M.E.; Brown, L.C.; Guo, C.; Higginbottom, S.; Almo, S.C.; Fischbach, M.A. A metabolic pathway for bile acid dehydroxylation by the gut microbiome. Nature 2020, 582, 566–570. [Google Scholar] [CrossRef]
  69. Jin, W.-B.; Li, T.-T.; Huo, D.; Qu, S.; Li, X.V.; Arifuzzaman, M.; Lima, S.F.; Shi, H.-Q.; Wang, A.; Putzel, G.G.; et al. Genetic manipulation of gut microbes enables single-gene interrogation in a complex microbiome. Cell 2022, 185, 547–562.e22. [Google Scholar] [CrossRef]
  70. Devlin, A.S.; Fischbach, M.A. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat. Chem. Biol. 2015, 11, 685–690. [Google Scholar] [CrossRef] [Green Version]
  71. Lee, J.W.J.; Plichta, D.; Hogstrom, L.; Borren, N.Z.; Lau, H.; Gregory, S.M.; Tan, W.; Khalili, H.; Clish, C.; Vlamakis, H.; et al. Multi-omics reveal microbial determinants impacting responses to biologic therapies in inflammatory bowel disease. Cell Host Microbe 2021, 29, 1294–1304.e4. [Google Scholar] [CrossRef] [PubMed]
  72. Doden, H.L.; Ridlon, J.M. Microbial Hydroxysteroid Dehydrogenases: From Alpha to Omega. Microorganisms 2021, 9, 469. [Google Scholar] [CrossRef] [PubMed]
  73. Quinn, R.A.; Melnik, A.V.; Vrbanac, A.; Fu, T.; Patras, K.A.; Christy, M.P.; Bodai, Z.; Belda-Ferre, P.; Tripathi, A.; Chung, L.K.; et al. Global chemical effects of the microbiome include new bile-acid conjugations. Nature 2020, 579, 123–129. [Google Scholar] [CrossRef]
  74. de Aguiar Vallim, T.Q.; Tarling, E.J.; Edwards, P.A. Pleiotropic roles of bile acids in metabolism. Cell Metab. 2013, 17, 657–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Sayin, S.I.; Wahlström, A.; Felin, J.; Jäntti, S.; Marschall, H.-U.; Bamberg, K.; Angelin, B.; Hyötyläinen, T.; Orešič, M.; Bäckhed, F. Gut Microbiota Regulates Bile Acid Metabolism by Reducing the Levels of Tauro-beta-muricholic Acid, a Naturally Occurring FXR Antagonist. Cell Metab. 2013, 17, 225–235. [Google Scholar] [CrossRef] [Green Version]
  76. Islam, K.S.; Fukiya, S.; Hagio, M.; Fujii, N.; Ishizuka, S.; Ooka, T.; Ogura, Y.; Hayashi, T.; Yokota, A. Bile Acid Is a Host Factor That Regulates the Composition of the Cecal Microbiota in Rats. Gastroenterology 2011, 141, 1773–1781. [Google Scholar] [CrossRef]
  77. Jones, M.L.; Martoni, C.J.; Ganopolsky, J.G.; Labbé, A.; Prakash, S. The human microbiome and bile acid metabolism: Dysbiosis, dysmetabolism, disease and intervention. Expert Opin. Biol. Ther. 2014, 14, 467–482. [Google Scholar] [CrossRef]
  78. Sagar, N.M.; Cree, I.A.; Covington, J.A.; Arasaradnam, R.P. The interplay of the gut microbiome, bile acids, and volatile organic compounds. Gastroenterol. Res. Pract. 2015, 2015, 398585. [Google Scholar] [CrossRef] [Green Version]
  79. Degirolamo, C.; Rainaldi, S.; Bovenga, F.; Murzilli, S.; Moschetta, A. Microbiota Modification with Probiotics Induces Hepatic Bile Acid Synthesis via Downregulation of the Fxr-Fgf15 Axis in Mice. Cell Rep. 2014, 7, 12–18. [Google Scholar] [CrossRef] [Green Version]
  80. Herrema, H.; Meissner, M.; van Dijk, T.H.; Brufau, G.; Boverhof, R.; Oosterveer, M.H. Bile salt sequestration induces hepatic de novo lipogenesis through farnesoid X receptor- and liver X receptor alpha-controlled metabolic pathways in mice. Hepatology 2010, 51, 806–816. [Google Scholar] [CrossRef]
  81. Coleman, R.; Lowe, P.J.; Billington, D. Membrane lipid composition and susceptibility to bile salt damage. Biochim. Biophys. Acta BBA Biomembr. 1980, 599, 294–300. [Google Scholar] [CrossRef]
  82. Taranto, M.P.; Perez-Martinez, G.; Font d, V. Effect of bile acid on the cell membrane functionality of lactic acid bacteria for oral administration. Res. Microbiol. 2006, 157, 720–725. [Google Scholar] [CrossRef] [PubMed]
  83. Watanabe, M.; Fukiya, S.; Yokota, A. Comprehensive evaluation of the bactericidal activities of free bile acids in the large intestine of humans and rodents. J. Lipid Res. 2017, 58, 1143–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Clements, W.D.; Parks, R.; Erwin, P.; Halliday, M.I.; Barr, J.; Rowlands, B.J. Role of the gut in the pathophysiology of extrahepatic biliary obstruction. Gut 1996, 39, 587–593. [Google Scholar] [CrossRef] [Green Version]
  85. Ilan, Y. Leaky gut and the liver: A role for bacterial translocation in nonalcoholic steatohepatitis. World J. Gastroenterol. 2012, 18, 2609–2618. [Google Scholar] [CrossRef] [PubMed]
  86. Wigg, A.J.; Roberts-Thomson, I.C.; Dymock, R.B.; McCarthy, P.J.; Grose, R.H.; Cummins, A.G. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut 2001, 48, 206–211. [Google Scholar] [CrossRef] [Green Version]
  87. Fouts, D.E.; Torralba, M.; Nelson, K.E.; Brenner, D.A.; Schnabl, B. Bacterial translocation and changes in the intestinal microbiome in mouse models of liver disease. J. Hepatol. 2012, 56, 1283–1292. [Google Scholar] [CrossRef] [Green Version]
  88. Lorenzo-Zúñiga, V.; Bartolí, R.; Planas, R.; Hofmann, A.F.; Viñado, B.; Hagey, L.R.; Hernández, J.M.; Mañé, J.; Alvarez-Gonzalez, M.A.; Ausina, V.; et al. Oral bile acids reduce bacterial overgrowth, bacterial translocation, and endotoxemia in cirrhotic rats. Hepatology 2003, 37, 551–557. [Google Scholar] [CrossRef]
  89. Berg, R.D. Bacterial translocation from the gastrointestinal tract. Adv. Exp. Med. Biol. 1999, 473, 11–30. [Google Scholar] [CrossRef]
  90. Cahill, C.J.; Pain, J.A.; Bailey, M.E. Bile salts, endotoxin and renal function in obstructive jaundice. Surg. Gynecol. Obstet. 1987, 165, 519–522. [Google Scholar]
  91. Ding, J.W.; Andersson, R.; Soltesz, V.; Willén, R.; Bengmark, S. The Role of Bile and Bile Acids in Bacterial Translocation in Obstructive Jaundice in Rats. Eur. Surg. Res. 1993, 25, 11–19. [Google Scholar] [CrossRef] [PubMed]
  92. Evans, H.J.; Torrealba, V.; Hudd, C.; Knight, M. The effect of preoperative bile salt administration on postoperative renal function in patients with obstructive jaundice. Br. J. Surg. 1982, 69, 706–708. [Google Scholar] [CrossRef] [PubMed]
  93. Marzano, M.; Fosso, B.; Colliva, C.; Notario, E.; Passeri, D.; Intranuovo, M. Farnesoid X receptor activation by the novel agonist TC-100 (3a, 7a, 11ß-Trihydroxy-6a-ethyl-5ß-cholan-24-oic Acid) preserves the intestinal barrier integrity and promotes intestinal microbial reshaping in a mouse model of obstructed bile acid flow. Biomed. Pharmacother. 2022, 153, 1–9. [Google Scholar] [CrossRef]
  94. Claus, S.P.; Tsang, T.M.; Wang, Y.; Cloarec, O.; Skordi, E.; Martin, F.-P.; Rezzi, S.; Ross, A.; Kochhar, S.; Holmes, E.; et al. Systemic multicompartmental effects of the gut microbiome on mouse metabolic phenotypes. Mol. Syst. Biol. 2008, 4, 219. [Google Scholar] [CrossRef] [Green Version]
  95. Hartmann, P.; Hochrath, K.; Horvath, A.; Chen, P.; Seebauer, C.T.; Llorente, C.; Wang, L.; Alnouti, Y.; Fouts, D.E.; Stärkel, P.; et al. Modulation of the intestinal bile acid/farnesoid X receptor/fibroblast growth factor 15 axis improves alcoholic liver disease in mice. Hepatology 2018, 67, 2150–2166. [Google Scholar] [CrossRef]
  96. Martin, F.-P.; Dumas, M.-E.; Wang, Y.; Legido-Quigley, C.; Yap, I.K.S.; Tang, H.; Zirah, S.; Murphy, G.M.; Cloarec, O.; Lindon, J.C.; et al. A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model. Mol. Syst. Biol. 2007, 3, 112. [Google Scholar] [CrossRef] [Green Version]
  97. Swann, J.R.; Want, E.J.; Geier, F.M.; Spagou, K.; Wilson, I.D.; Sidaway, J.E. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4523–4530. [Google Scholar] [CrossRef] [Green Version]
  98. Wostmann, B.S. Intestinal Bile Acids and Cholesterol Absorption in the Germfree Rat. J. Nutr. 1973, 103, 982–990. [Google Scholar] [CrossRef]
  99. Friedman, E.S.; Li, Y.; Shen, T.-C.D.; Jiang, J.; Chau, L.; Adorini, L.; Babakhani, F.; Edwards, J.; Shapiro, D.; Zhao, C.; et al. FXR-Dependent Modulation of the Human Small Intestinal Microbiome by the Bile Acid Derivative Obeticholic Acid. Gastroenterology 2018, 155, 1741–1752.e5. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, S.; Dong, W.; Liu, L.; Xu, M.; Wang, Y.; Liu, T.; Zhang, Y.; Wang, B.; Cao, H. Interplay between bile acids and the gut microbiota promotes intestinal carcinogenesis. Mol. Carcinog. 2019, 58, 1155–1167. [Google Scholar] [CrossRef] [Green Version]
  101. Xu, M.; Cen, M.; Shen, Y.; Zhu, Y.; Cheng, F.; Tang, L.; Hu, W.; Dai, N. Deoxycholic Acid-Induced Gut Dysbiosis Disrupts Bile Acid Enterohepatic Circulation and Promotes Intestinal Inflammation. Dig. Dis. Sci. 2021, 66, 568–576. [Google Scholar] [CrossRef] [PubMed]
  102. Jones, M.L.; Tomaro-Duchesneau, C.; Martoni, C.J.; Prakash, S. Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications. Expert Opin. Biol. Ther. 2013, 13, 631–642. [Google Scholar] [CrossRef] [PubMed]
  103. Joyce, S.A.; MacSharry, J.; Casey, P.G.; Kinsella, M.; Murphy, E.F.; Shanahan, F.; Hill, C.; Gahan, C.G.M. Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proc. Natl. Acad. Sci. USA 2014, 111, 7421–7426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Mukherji, A.; Kobiita, A.; Ye, T.; Chambon, P. Homeostasis in Intestinal Epithelium Is Orchestrated by the Circadian Clock and Microbiota Cues Transduced by TLRs. Cell 2013, 153, 812–827. [Google Scholar] [CrossRef] [Green Version]
  105. Angelakis, E.; Merhej, V.; Raoult, D. Related actions of probiotics and antibiotics on gut microbiota and weight modification. Lancet Infect. Dis. 2013, 13, 889–899. [Google Scholar] [CrossRef]
  106. Hernandez-Gomez, J.G.; Lopez-Bonilla, A.; Trejo-Tapia, G.; Avila-Reyes, S.V.; Jimenez-Aparicio, A.R.; Hernandez-Sanchez, H. In Vitro Bile Salt Hydrolase (BSH) Activity Screening of Different Probiotic Microorganisms. Foods 2021, 10, 674. [Google Scholar] [CrossRef]
  107. Briand, F.; Sulpice, T.; Giammarinaro, P.; Roux, X. Saccharomyces boulardii CNCM I-745 changes lipidemic profile and gut microbiota in a hamster hypercholesterolemic model. Benef. Microbes 2019, 10, 555–567. [Google Scholar] [CrossRef]
  108. Ryan, J.J.; Hanes, D.A.; Schafer, M.B.; Mikolai, J.; Zwickey, H. Effect of the Probiotic Saccharomyces boulardii on Cholesterol and Lipoprotein Particles in Hypercholesterolemic Adults: A Single-Arm, Open-Label Pilot Study. J. Altern. Complement. Med. 2015, 21, 288–293. [Google Scholar] [CrossRef] [Green Version]
  109. Duboc, H.; Rajca, S.; Rainteau, D.; Benarous, D.; Maubert, M.-A.; Quervain, E.; Thomas, G.; Barbu, V.; Humbert, L.; Despras, G.; et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut 2013, 62, 531–539. [Google Scholar] [CrossRef]
  110. Ogilvie, L.A.; Jones, B.V. Dysbiosis modulates capacity for bile acid modification in the gut microbiomes of patients with inflammatory bowel disease: A mechanism and marker of disease? Gut 2012, 61, 1642–1643. [Google Scholar] [CrossRef] [Green Version]
  111. Liu, L.; Dong, W.; Wang, S.; Zhang, Y.; Liu, T.; Xie, R.; Wang, B.; Cao, H. Deoxycholic acid disrupts the intestinal mucosal barrier and promotes intestinal tumorigenesis. Food Funct. 2018, 9, 5588–5597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Xu, M.; Shen, Y.; Cen, M.; Zhu, Y.; Cheng, F.; Tang, L.; Zheng, X.; Kim, J.J.; Dai, N.; Hu, W. Modulation of the Gut Microbiota-farnesoid X Receptor Axis Improves Deoxycholic Acid-induced Intestinal Inflammation in Mice. J. Crohn’s Colitis 2021, 15, 1197–1210. [Google Scholar] [CrossRef] [PubMed]
  113. Sinha, S.R.; Haileselassie, Y.; Nguyen, L.P.; Tropini, C.; Wang, M.; Becker, L.S. Dysbiosis-Induced Secondary Bile Acid Deficiency Promotes Intestinal Inflammation. Cell Host Microb. 2020, 27, 659–670. [Google Scholar] [CrossRef] [PubMed]
  114. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  115. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef]
  116. Jakubczyk, D.; Leszczyńska, K.; Górska, S. The Effectiveness of Probiotics in the Treatment of Inflammatory Bowel Disease (IBD)—A Critical Review. Nutrients 2020, 12, 1973. [Google Scholar] [CrossRef]
  117. Huttenhower, C.; Kostic, A.D.; Xavier, R.J. Inflammatory Bowel Disease as a Model for Translating the Microbiome. Immunity 2014, 40, 843–854. [Google Scholar] [CrossRef] [Green Version]
  118. Jia, B.; Park, D.; Hahn, Y.; Jeon, C.O. Metagenomic analysis of the human microbiome reveals the association between the abundance of gut bile salt hydrolases and host health. Gut Microbes 2020, 11, 1300–1313. [Google Scholar] [CrossRef]
  119. Carlsson, A.H.; Yakymenko, O.; Olivier, I.; Hakansson, F.; Postma, E.; Keita, A.V. Faecalibacteriumprausnitzii supernatant improves intestinal barrier function in mice DSS colitis. Scand. J. Gastroenterol. 2013, 48, 1136–1144. [Google Scholar] [CrossRef]
  120. Laval, L.; Martin, R.; Natividad, J.; Chain, F.; Miquel, S.; de Maredsous, C.D.; Capronnier, S.; Sokol, H.; Verdu, E.; van HylckamaVlieg, J.E.; et al. Lactobacillus rhamnosusCNCM I-3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrier hyper-permeability in mice. Gut Microbes 2015, 6, 1–9. [Google Scholar] [CrossRef] [Green Version]
  121. Traina, G.; Menchetti, L.; Rappa, F.; Casagrande-Proietti, P.; Barbato, O.; Leonardi, L.; Carini, F.; Piro, F.; Brecchia, G. Probiotic mixture supplementation in the preventive management of trinitrobenzenesulfonic acid-induced inflammation in a murine model. J. Biol. Regul. Homeost. Agents 2016, 30, 895–901. [Google Scholar] [PubMed]
  122. Chae, J.M.; Heo, W.; Cho, H.T.; Lee, D.H.; Kim, J.H.; Rhee, M.S.; Park, T.S.; Kim, Y.K.; Lee, J.H.; Kim, Y.J. Effects of Orally-Administered Bifidobacterium animalis subsp. lactis Strain BB12 on Dextran Sodium Sulfate-Induced Colitis in Mice. J. Microbiol. Biotechnol. 2018, 28, 1800–1805. [Google Scholar] [CrossRef] [PubMed]
  123. Duranti, S.; Gaiani, F.; Mancabelli, L.; Milani, C.; Grandi, A.; Bolchi, A.; Santoni, A.; Lugli, G.A.; Ferrario, C.; Mangifesta, M.; et al. Elucidating the gut microbiome of ulcerative colitis: Bifidobacteria as novel microbial biomarkers. FEMS Microbiol. Ecol. 2016, 92, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Elian, S.D.; Souza, E.L.; Vieira, A.T.; Teixeira, M.M.; Arantes, R.M.; Nicoli, J.R. Bifidobacterium longum subsp. infantis BB-02 attenuates acute murine experimental model of inflammatory bowel disease. Benef. Microb. 2015, 6, 277–286. [Google Scholar] [CrossRef]
  125. Javed, N.H.; Alsahly, M.B.; Khubchandani, J. Oral Feeding of Probiotic Bifidobacteriuminfantis: Colonic Morphological Changes in Rat Model of TNBS-Induced Colitis. Scientifica 2016, 2016, 9572596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Satish Kumar, C.S.; Kondal, R.K.; Reddy, A.G.; Vinoth, A.; Ch, S.R.; Boobalan, G. Protective effect of Lactobacillus plantarum 21, a probiotic on trinitrobenzenesulfonic acid-induced ulcerative colitis in rats. Int. Immunopharmacol. 2015, 25, 504–510. [Google Scholar] [CrossRef]
  127. Hrdý, J.; Alard, J.; Couturier-Maillard, A.; Boulard, O.; Boutillier, D.; Delacre, M.; Lapadatescu, C.; Cesaro, A.; Blanc, P.; Pot, B.; et al. Lactobacillus reuteri 5454 and Bifidobacterium animalis ssp. lactis 5764 improve colitis while differentially impacting dendritic cells maturation and antimicrobial responses. Sci. Rep. 2020, 10, 5345. [Google Scholar] [CrossRef] [Green Version]
  128. Biagioli, M.; Laghi, L.; Carino, A.; Cipriani, S.; Distrutti, E.; Marchianò, S.; Parolin, C.; Scarpelli, P.; Vitali, B.; Fiorucci, S. Metabolic Variability of a Multispecies Probiotic Preparation Impacts on the Anti-inflammatory Activity. Front. Pharmacol. 2017, 8, 505. [Google Scholar] [CrossRef] [Green Version]
  129. Hegazy, S.K.; El-Bedewy, M.M. Effect of probiotics on pro-inflammatory cytokines and NF-kappaB activation in ulcerative colitis. World J. Gastroenterol. 2010, 16, 4145–4151. [Google Scholar] [CrossRef]
  130. Kato, K.; Mizuno, S.; Umesaki, Y.; Ishii, Y.; Sugitani, M.; Imaoka, A.; Otsuka, M.; Hasunuma, O.; Kurihara, R.; Iwasaki, A.; et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment. Pharmacol. Ther. 2004, 20, 1133–1141. [Google Scholar] [CrossRef]
  131. Kruis, W.; Fric, P.; Pokrotnieks, J.; Lukas, M.; Fixa, B.; Kaščák, M.; Kamm, M.A.; Weismueller, J.; Beglinger, C.; Stolte, M.; et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 2004, 53, 1617–1623. [Google Scholar] [CrossRef] [PubMed]
  132. Matsuoka, K.; Uemura, Y.; Kanai, T.; Kunisaki, R.; Suzuki, Y.; Yokoyama, K.; Yoshimura, N.; Hibi, T. Efficacy of Bifidobacterium breve Fermented Milk in Maintaining Remission of Ulcerative Colitis. Dig. Dis. Sci. 2018, 63, 1910–1919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Tamaki, H.; Nakase, H.; Inoue, S.; Kawanami, C.; Itani, T.; Ohana, M. Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: A randomized, double-blinded, placebo-controlled multicenter trial. Dig. Endosc. 2016, 28, 67–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Garcia Vilela, E.; De Lourdes De Abreu Ferrari, M.; Oswaldo Da Gama Torres, H.; Guerra Pinto, A. Carolina Carneiro Aguirre, A.; Paiva Martins, F.; Sales Da Cunha, A. Influence of Saccharomyces boulardii on the intestinal permeability of patients with Crohn’s disease in remission. Scand. J. Gastroenterol. 2008, 43, 842–848. [Google Scholar] [CrossRef]
  135. Wildt, S.; Nordgaard, I.; Hansen, U.; Brockmann, E.; Rumessen, J.J. A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis. J. Crohns Colitis 2011, 5, 115–121. [Google Scholar] [CrossRef] [Green Version]
  136. Sood, A.; Midha, V.; Makharia, G.K.; Ahuja, V.; Singal, D.; Goswami, P.; Tandon, R.K. The Probiotic Preparation, VSL#3 Induces Remission in Patients with Mild-to-Moderately Active Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2009, 7, 1202–1209.e1. [Google Scholar] [CrossRef] [PubMed]
  137. Tursi, A.; Brandimarte, G.; Papa, A.; Giglio, A.; Elisei, W.; Giorgetti, G.M.; Forti, G.; Morini, S.; Hassan, C.; Pistoia, M.A.; et al. Treatment of Relapsing Mild-to-Moderate Ulcerative Colitis with the Probiotic VSL#3 as Adjunctive to a Standard Pharmaceutical Treatment: A Double-Blind, Randomized, Placebo-Controlled Study. Am. J. Gastroenterol. 2010, 105, 2218–2227. [Google Scholar] [CrossRef] [Green Version]
  138. Huynh, H.Q.; Debruyn, J.; Guan, L.; Diaz, H.; Li, M.; Girgis, S.; Turner, J.; Fedorak, R.; Madsen, K. Probiotic preparation VSL#3 induces remission in children with mild to moderate acute ulcerative colitis: A pilot study. Inflamm. Bowel Dis. 2009, 15, 760–768. [Google Scholar] [CrossRef]
  139. Fedorak, R.N.; Feagan, B.G.; Hotte, N.; Leddin, D.; Dieleman, L.A.; Petrunia, D.M. The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clin. Gastroenterol. Hepatol. 2015, 13, 928–935. [Google Scholar]
  140. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humaran, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  141. Pappa, H.M.; Mitchell, P.D.; Jiang, H.; Kassiff, S.; Filip-Dhima, R.; DiFabio, D.; Quinn, N.; Lawton, R.C.; Varvaris, M.; Van Straaten, S.; et al. Treatment of Vitamin D Insufficiency in Children and Adolescents with Inflammatory Bowel Disease: A Randomized Clinical Trial Comparing Three Regimens. J. Clin. Endocrinol. Metab. 2012, 97, 2134–2142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Pappa, H.M.; Mitchell, P.D.; Jiang, H.; Kassiff, S.; Filip-Dhima, R.; DiFabio, D.; Quinn, N.; Lawton, R.C.; Bronzwaer, M.E.S.; Koenen, K.; et al. Maintenance of Optimal Vitamin D Status in Children and Adolescents with Inflammatory Bowel Disease: A Randomized Clinical Trial Comparing Two Regimens. J. Clin. Endocrinol. Metab. 2014, 99, 3408–3417. [Google Scholar] [CrossRef] [PubMed]
  143. Kumari, M.; Khazai, N.B.; Ziegler, T.R.; Nanes, M.S.; Abrams, S.A.; Tangpricha, V. Vitamin D-mediated calcium absorption in patients with clinically stable Crohn’s disease: A pilot study. Mol. Nutr. Food Res. 2010, 54, 1085–1091. [Google Scholar] [CrossRef]
  144. Ananthakrishnan, A.N.; Khalili, H.; Higuchi, L.M.; Bao, Y.; Korzenik, J.R.; Giovannucci, E.L. Higher predicted vitamin D status is associated with reduced risk of Crohn’s disease. Gastroenterology 2012, 142, 482–489. [Google Scholar] [CrossRef] [Green Version]
  145. Cantorna, M.T.; Munsick, C.; Bemiss, C.; Mahon, B.D. 1,25-Dihydroxycholecalciferol Prevents and Ameliorates Symptoms of Experimental Murine Inflammatory Bowel Disease. J. Nutr. 2000, 130, 2648–2652. [Google Scholar] [CrossRef] [Green Version]
  146. Jørgensen, S.P.G.; Agnholt, J.; Glerup, H.; Lyhne, S.; Villadsen, G.E.; Hvas, C.L.; Bartels, L.E.; Kelsen, J.; Christensen, L.A.; Dahlerup, J.F. Clinical trial: Vitamin D3 treatment in Crohn’s disease—A randomized double-blind placebo-controlled study. Aliment. Pharmacol. Ther. 2010, 32, 377–383. [Google Scholar] [CrossRef]
  147. Khalili, H.; Huang, E.S.; Ananthakrishnan, A.N.; Higuchi, L.; Richter, J.M.; Fuchs, C.S. Geographical variation and incidence of inflammatory bowel disease among US women. Gut 2012, 61, 1686–1692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Dryden, G.W.; Lam, A.; Beatty, K.; Qazzaz, H.H.; McClain, C.J. A Pilot Study to Evaluate the Safety and Efficacy of an Oral Dose of (−)-Epigallocatechin-3-Gallate–Rich Polyphenon E in Patients with Mild to Moderate Ulcerative Colitis. Inflamm. Bowel Dis. 2013, 19, 1904–1912. [Google Scholar] [CrossRef]
  149. Ajouz, H.; Mukherji, D.; Shamseddine, A. Secondary bile acids: An underrecognized cause of colon cancer. World J. Surg. Oncol. 2014, 12, 164. [Google Scholar] [CrossRef] [Green Version]
Figure 1. BSH-competent bacteria activity in probiotics. In physiological conditions, there is a balance between the input of conjugated BA taken up in the enterocytes and activating FXR and the unconjugated BA fecal output. Enriching the intestinal milieu with BSH-competent intestinal microorganisms induces BA transformation that, in turn, shapes the magnitude of FXR activation in the enterocytes. This translates into increased fecal BA excretion and lower FXR transcriptional activation. (Bas = bile acids; CA = cholic acid; CDCA = chenodeoxycholic acid; FXR = Farnesoid X Receptors; green FXR indicates activated FXR; red FXR indicated de-activated FXR; purple globus indicates BSH-competent bacteria; black arrows pointing up indicate an increase of CA, CDCA and fecal excretion, bold green arrow pointing up—right panel—indicates increased deconjugation).
Figure 1. BSH-competent bacteria activity in probiotics. In physiological conditions, there is a balance between the input of conjugated BA taken up in the enterocytes and activating FXR and the unconjugated BA fecal output. Enriching the intestinal milieu with BSH-competent intestinal microorganisms induces BA transformation that, in turn, shapes the magnitude of FXR activation in the enterocytes. This translates into increased fecal BA excretion and lower FXR transcriptional activation. (Bas = bile acids; CA = cholic acid; CDCA = chenodeoxycholic acid; FXR = Farnesoid X Receptors; green FXR indicates activated FXR; red FXR indicated de-activated FXR; purple globus indicates BSH-competent bacteria; black arrows pointing up indicate an increase of CA, CDCA and fecal excretion, bold green arrow pointing up—right panel—indicates increased deconjugation).
Nutrients 14 03212 g001
Table 1. Clinical Trials exploiting the use of Probiotics in IBD.
Table 1. Clinical Trials exploiting the use of Probiotics in IBD.
Trial IdentifierTrail Phase (Status)TitleConditionsInterventions
NCT03266484Active, not recruitingEffect of a Probiotic Mixture on the Gut Microbiome and Fatigue in Patients With Quiescent Inflammatory Bowel DiseaseCrohn’s Disease
Ulcerative Colitis
-
Dietary Supplement: Probiotic Mixture
-
Dietary Supplement: Placebo
NCT01765998UnknownThe Effect of Probiotics on Exacerbation of Inflammatory Bowel Disease Exacerbation (Crohn’s Disease)Crohn’s Disease
-
Drug: Probiotic
-
Drug: Placebo
NCT01765439Active, not recruitingThe Effect of VSL#3 Probiotic Preparation on the Bile Acid Metabolism in Patients With Inflammatory Bowel DiseaseCrohn’s Disease
Ulcerative Colitis
Dietary Supplement: VSL#3
NCT00175292CompletedA Randomized Controlled Trial of VSL#3 for the Prevention of Endoscopic Recurrence Following Surgery for Crohn’s DiseaseCrohn’s Disease
Inflammatory Bowel Disease
Drug: Probiotic—VSL#3
NCT01078935UnknownThe Effect of Probiotics on the Rate of Recovery of Inflammatory Bowel Disease Exacerbation, Endothelial Function, and Markers of InflammationCrohn’s Disease
Ulcerative Colitis
-
Dietary Supplement: probiotics
-
Dietary Supplement: placebo
NCT01173588CompletedEffect of Yogurt Added With Bifidobacteria and Soluble Fiber on Bowel FunctionInflammatory Bowel DiseaseOther: yogurt added with bifidobacteria and soluble fiber (YBF)
NCT01479660UnknownRole of Healthy Bacteria in Ulcerative ColitisUlcerative Colitis
-
Other: Control
-
Drug: Probiotic
NCT00510978UnknownProbiotics in GastroIntestinal Disorders (PROGID)Crohn’s Disease
Ulcerative Colitis
-
Biological: Bifidobacterium infantis 35624
-
Biological: Lactobacillus salivarius UCC118
-
Biological: Placebo
NCT01632462UnknownA Prospective, Placebo Controlled, Double-Blind, Cross-over Study on the Effects of a Probiotic Preparation (VSL#3) on Metabolic Profile, Intestinal Permeability, Microbiota, Cytokines and Chemokines Expression and Other Inflammatory Markers in Pediatric Patients With Crohn’s DiseaseCrohn’s DiseaseDrug: VSL#3
NCT01548014UnknownThe Effect of a Probiotic Preparation (VSL#3) Plus Infliximab in Children With Crohn’s DiseaseCrohn’s Disease
-
Dietary Supplement: VSL#3
NCT00374374CompletedTreatment With Lactobacillus Rhamnosus and Lactobacillus Acidophilus for Patients With Active Colonic Crohn’s DiseaseCrohn’s Disease
-
Behavioral: Administration of probiotic
NCT03565939CompletedProbiotic Treatment of Ulcerative Colitis With Trichuris Suis Ova (TSO) (PROCTO)Ulcerative Colitis Chronic Moderate
-
Biological: Trichuris suis ova
-
Biological: Placebo
NCT00114465CompletedVSL#3 Versus Placebo in Maintenance of Remission in Crohn’s DiseaseCrohn’s Disease
-
Drug: VSL#3- Other: Placebo
NCT00944736CompletedEffect of VSL#3 on Intestinal Permeability in Pediatric Crohn’s DiseaseCrohn’s Disease
-
Dietary Supplement: VSL#3
-
Dietary Supplement: Placebo
NCT04842149RecruitingThe Effects of Bifidobacterium Breve Bif195 for Small Intestinal Crohn’s DiseaseCrohn’s Disease
-
Dietary Supplement: Bif195 capsules
-
Dietary Supplement: Placebo capsules
NCT04223479CompletedEffect of Probiotic Supplementation on the Immune System in Patients With Ulcerative Colitis in Amman, JordanUlcerative Colitis
-
Drug: Probiotic Formula Capsule
-
Drug: Placebos
NCT01698970CompletedEffect of the Consumption of a Probiotic Strain on the Prevention of Post-operative Recurrence in Crohn’s DiseaseCrohn’s Disease
-
Other: 1-Freeze-dried Probiotics provided in capsule (150 mg) containing 1.0 × 1010 colony forming unit per capsule (test)
-
Other: 2-excipients (150 mg) in capsule (control)
NCT01772615CompletedTreatment of Ulcerative Colitis With Ciprofloxacin and E. Coli NissleUlcerative Colitis
-
Drug: Ciprofloxacin
-
Dietary Supplement: E. coli Nissle
NCT02488954TerminatedInterest of Propionibacterium Freudenreichii for the Treatment of Mild to Moderate Ulcerative Colitis (EMMENTAL)Ulcerative ColitisOther: Probiotics in the form of cheese portion
NCT04305535UnknownImpact of an Oligomeric Diet in Intestinal Absorption and Inflammatory Markers in Patients With Crohn DiseaseCrohn Disease
Absorption; Disorder: Protein
Absorption; Disorder
Absorption; Disorder: Fat
Absorption; Disorder: Carbohydrate
Malnutrition
-
Dietary Supplement: Peptidic + Probiotic
-
Dietary Supplement: Peptidic + Placebo
-
Dietary Supplement: Polymeric + Placebo
NCT00305409CompletedSynbiotic Treatment in Crohn’s Disease PatientsCrohn’s DiseaseDrug: Synbiotic (Synergy I/B. longum)
NCT04804046RecruitingSynbiotics and Post-op Crohn’s DiseaseCrohn’s Disease
-
Dietary Supplement: Synbiotic
-
Other: Digestible Maltodextrin
NCT00803829CompletedSynbiotic Treatment of Ulcerative Colitis PatientsUlcerative Colitis
-
Other: Synbiotic (Synergy1/B. longum)
NCT00951548CompletedFood Supplementation With VSL#3 as a Support to Standard Pharmaceutical Therapy in Ulcerative ColitisUlcerative Colitis
-
Dietary Supplement: VSL#3
-
Dietary Supplement: Placebo
NCT00374725CompletedTreatment of Ulcerative Colitis With a Combination of Lactobacillus Rhamnosus and Lactobacillus Acidophilus.Ulcerative ColitisBehavioral: Administration of probiotic (L. rhamnosus and L. acidophilus)
NCT03415711TerminatedPRObiotic VSL#3® for Maintenance of Clinical and Endoscopic REMission in Ulcerative ColitisUlcerative Colitis
-
Dietary Supplement: VSL#3®
-
Drug: MesalamineDrug: Placebo
NCT04102852RecruitingLactobacillus Rhamnosus GG (ATCC 53103) in Mild-moderately Active UC PatientsUlcerative Colitis Chronic Mild
Ulcerative Colitis Chronic Moderate
Dietary Supplement: Lactobacillus rhamnosus GG ATCC 53103
NCT00367705UnknownVSL#3 Treatment in Children With Crohn’s DiseaseCrohn’s Disease
-
Dietary Supplement: VSL#3®
-
Dietary Supplement: Placebo
NCT04969679CompletedAdditive Effect of Probiotics (Mutaflor®) in Patients With Ulcerative Colitis on 5-ASA TreatmentUlcerative Colitis
-
Drug: E. coli Nissle 1917 (Mutaflor®)
-
Drug: Placebo
NCT00268164TerminatedLactobacillus Acidophilus and Bifidobacterium Animalis Subsp. Lactis, Maintenance Treatment in Ulcerative ColitisUlcerative ColitisDrug: lactobacilus acidophilus & bifidobacterium animalis/lactis
Table 2. Clinical Trials exploiting the use of Microbial Metabolites in IBD.
Table 2. Clinical Trials exploiting the use of Microbial Metabolites in IBD.
Trial IdentifierTrail Phase (Status)TitleConditionsInterventions
Short Chain Fatty Acids
NCT05456763CompletedButyrate in Pediatric Inflammatory Bowel DiseaseIBD
-
Dietary Supplement: sodium butyrate
-
Other: placebo
Tryptophan Metabolites
NCT04089501CompletedThe Role of the Pregnane X Receptor (PXR) in Indole Signaling and Intestinal Permeability in Inflammatory Bowel DiseaseIBD
-
Diagnostic Test: Stool collection
-
Diagnostic Test: Biopsy collection
Bile Acid Metabolites
NCT03724175RecruitingThe Role of Secondary Bile Acids in Intestinal InflammationUlcerative Colitis
Pouchitis
-
Drug: ursodiol (ursodeoxycholic acid, UDCA)
Vitamines
NCT03145896UnknownThe Correlation Between Anemia of Chronic Diseases, Hepcidin and Vitamin D in IBD PatientsIBDDietary Supplement: vitamin D
NCT03162432CompletedHigh Dose Interval Vitamin D Supplementation in Patients With IBD Receiving RemicadeIBD
Ulcerative Colitis
Crohn Disease
Drug: Vitamin D3
NCT02076750CompletedWeekly Vitamin D in Pediatric IBDIBD
Skin Pigmentation
Dietary Supplement: Vitamin D3 (cholecalciferol)
NCT00621257Terminated
Has Results
[141,142]
Vitamin D Levels in Children With IBDIBD
Crohn’s Disease
Ulcerative Colitis
-
Dietary Supplement: ergocalciferol
-
Dietary Supplement: Cholecalciferol
NCT02256605CompletedVitamin D3 Supplementation in Pediatric IBD: Weely vs Daily Dosing RegimensIBDDietary Supplement: Vitamin D-3
NCT04225819RecruitingAdjunctive Treatment With Vitamin D3 in Patients With Active IBDIBD
Crohn Disease
Ulcerative Colitis
Vitamin D3 Deficiency
-
Dietary Supplement: Vitamin D3
-
Other: Placebo
NCT03496246UnknownVitamin D Status in Inflammatory Bowel Disease (vdsinibd)Vitamin D Deficiency
IBD
-
Diagnostic Test: serum total 25(OH) vitamin D
-
Diagnostic Test: complete blood count (CBC)
-
Diagnostic Test: serum calcium level
-
Diagnostic Test: erythrocyte sedimentation rate (ESR)
-
Diagnostic Test: C-reactive protein (CRP)
-
Diagnostic Test: serum creatinine
-
Diagnostic Test: serum albumin level
-
Diagnostic Test: seum alanine aminotransferase
-
Diagnostic Test: serum potassium level
-
Diagnostic Test: serum phosphurus level
NCT04991324Not yet recruitingCholecalciferol Comedication in IBD—the 5C-study (5C)IBDDrug: Vitamin D3
NCT04828031RecruitingVitamin D Regulation of Gut Specific B Cells and Antibodies Targeting Gut Bacteria in Inflammatory Bowel DiseaseIBD
Ulcerative Colitis
Crohn Disease
Drug: Vitamin D
NCT04331639RecruitingHigh Dose Interval Vitamin D Supplementation in Patients With Inflammatory Bowel Disease Receiving Biologic TherapyIBD
Crohn Disease
Ulcerative Colitis
Vitamin D Deficiency
Dietary Supplement: vitamin D3
NCT01877577CompletedSupplementation of Vitamin D3 in Patients With Inflammatory Bowel Diseases and Hypovitaminosis DCrohn’s Disease
Ulcerative Colitis
Dietary Supplement: Vitamin D3
NCT00742781Completed
Has Results
Vitamin D Supplementation in Crohn’s PatientsIBDDietary Supplement: Vitamin D
NCT01121796UnknownInfluence of Vitamin D on Inflammatory Bowel Disease RemissionIBD
-
Dietary Supplement: Vitamin D
-
Other: Water or milk
-
Dietary Supplement: Vitamin D enriched milk
NCT01792388CompletedVitd and Barrier Function in IBDCrohn’s Disease
-
Dietary Supplement: Vitamin D
-
Dietary Supplement: Soya Bean oil
NCT00152841TerminatedEffect of Iron and Vitamin E Supplementation on Disease Activity in Patients With Either Crohn’s Disease or Ulcerative ColitisCrohn’s Disease
Ulcerative Colitis
Mild or Moderate Anaemia
-
Drug: Iron supplement 300–600 mg/day
-
Drug: Vitamin E 800 IU
NCT00114803CompletedNasal Calcitonin in the Treatment of Bone Mineral Loss in Children and Adolescents With Inflammatory Bowel DiseaseUlcerative Colitis
Crohn’s Disease
Drug: Calcitonin nasal spray (salmon)
NCT04913467RecruitingEffect of Ileocolonic Delivered Vitamins and an Anti-Inflammatory Diet on Crohn’s Disease and Healthy VolunteersCrohn Disease
-
Other: Groningen Anti-Inflammatory Diet (GrAID)
-
Dietary Supplement: ColoVit capsule
-
Other: ColoPulse-placebo capsule
NCT03718182UnknownCan Vitamin D Supplementation in People With Crohn’s Disease Improve Symptoms as an Adjunct Therapy?Crohn Disease
Vitamin D Deficiency
Dietary Supplement: Cholecalciferol
NCT02615288CompletedHigh Dose Vitamin D3 in Crohn’s DiseaseCrohn DiseaseDietary Supplement: Vitamin D3
NCT01692808CompletedBioavailability of Vitamin D in Children and Adolescents With Crohn’s DiseaseCrohn’s Disease
-
Drug: Vitamin D3 3000 UI daily
-
Drug: Vitamin D3 4000 UI daily
NCT02186275CompletedThe Vitamin D in Pediatric Crohn’s DiseaseCrohn’s Disease
-
Drug: Vitamin D3: 3000 or 4000 UI/day then 2000 UI/day
-
Drug: Vitamin D3 800 UI/day then 800 UI/day
NCT03999580RecruitingThe Vitamin D in Pediatric Crohn’s Disease (ViDiPeC-2)Crohn DiseaseDrug: vitamin D3
NCT01235325CompletedThe Effect of Vitamin K Supplementation on Bone Health in Adult Crohn’s Disease PatientsSupplementation
Bone Health
Crohn’s Disease
-
Dietary Supplement: phylloquinone (vitamin K1)
-
Dietary Supplement: placebo
NCT00132184UnknownVitamin D Treatment for Crohn’s DiseaseCrohn DiseaseDrug: Vitamin D
NCT01369667CompletedVitamin D Supplementation in Adult Crohn’s DiseaseCrohn Disease
-
Dietary Supplement: Vitamin D3
-
Other: Placebo
NCT01864616TerminatedThe Impact of Vitamin D on Disease Activity in Crohn’s DiseaseCrohn DiseaseDietary Supplement: Vitamin D3
NCT00427804Completed
Has Results [143]
Tumor Necrosis Factor Decreases Vitamin D Dependant Calcium AbsorptionRheumatoid Arthritis
Crohn’s Disease
Drug: calcitriol
NCT04308850Not yet recruitingExploring the Effects of Vitamin D Supplementation on the Chronic Course of Patients With Crohn’s Disease With Vitamin D DeficiencyCrohn’s Disease
Vitamin D Deficiency
Vitamin D Supplement
Drug: Vitamin D drops
NCT04134065UnknownThe Effect of Vitamin D in Crohn’s DiseaseCrohn’s Disease
Vitamin D Deficiency
-
Drug: Vitamin D
-
Drug: Placebo oral capsule
NCT02704624UnknownEffects of Supplementation of Vitamin D in Patients With Crohn’s DiseaseCrohn Disease
Vitamin D Deficiency
Fatigue
Sarcopenia
Muscle Weakness
Disorder of Bone Density and Structure, Unspecified
-
Dietary Supplement: Vitamin D
-
Other: Placebo
NCT02208310Terminated
Has Results [144,145,146,147]
Trial of High Dose Vitamin D in Patient’s With Crohn’s DiseaseCrohn’s Disease
Vitamin D Deficiency
-
Drug: Cholecalciferol 10,000 IU
-
Drug: Cholecalciferol 400 IU
NCT03615378TerminatedMaintenance Dosing of Vitamin D in Crohn’s DiseaseCrohns Disease
Vitamin D Deficiency
-
Dietary Supplement: 5000 IU D3
-
Dietary Supplement: 1000 IU D3
-
Dietary Supplement: Placebo
NCT04309058UnknownObservation of the Effect of Vitamin D Supplementation on Chronic Course of Patients With Ulcerative Colitis Based on Vitamin D Receptor Fok I Gene PolymorphismUlcerative Colitis
Vitamin D Deficiency
Vitamin D Supplement
Drug: Vitamin D drops
NCT01046773TerminatedVitamin D Supplementation as Non-toxic Immunomodulation in Children With Crohn’s DiseaseCrohn’s Disease
Vitamin D Deficiency
Drug: Cholecalciferol
NCT04276649CompletedA Retrospective Analysis: the Influence of Caltrate Supplement on the Effect of Mesalazine in Ulcerative ColitisUlcerative Colitis
Vitamin D Deficiency
Vitamin D Supplement
Drug: Caltrate
NCT04259060Not yet recruitingHydroxocobalamin Approach for Reducing of Calprotectin With Butyrate for Ulcerative Colitis RemissionUlcerative Colitis
-
Drug: Hydroxocobalamin with Butyrate
-
Drug: Placebo with Butyrate
Sulfur-Containing Metabolites
NCT01282905CompletedHydrogen Sulfide Detoxification and Butyrate Metabolism in Ulcerative ColitisUlcerative Colitis/
NCT04474561CompletedReduced Sulfur Diet in Ulcerative Colitis PatientsUlcerative Colitis
Diet Habit
Other: Reduced sulfur diet
Polyphenol Metabolites
NCT00718094Completed
Has Results [148]
Pilot Study of Green Tea Extract (Polyphenon E®)in Ulcerative ColitisMild to Moderately Active Ulcerative Colitis
-
Drug: Polyphenon E®
-
Drug: Placebo Oral Tablet
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gadaleta, R.M.; Cariello, M.; Crudele, L.; Moschetta, A. Bile Salt Hydrolase-Competent Probiotics in the Management of IBD: Unlocking the “Bile Acid Code”. Nutrients 2022, 14, 3212. https://doi.org/10.3390/nu14153212

AMA Style

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. https://doi.org/10.3390/nu14153212

Chicago/Turabian Style

Gadaleta, Raffaella Maria, Marica Cariello, Lucilla Crudele, and Antonio Moschetta. 2022. "Bile Salt Hydrolase-Competent Probiotics in the Management of IBD: Unlocking the “Bile Acid Code”" Nutrients 14, no. 15: 3212. https://doi.org/10.3390/nu14153212

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop