Bile salt hydrolase: a key player in gut microbiota and its implications for metabolic dysfunction-associated steatotic liver disease
Abstract
The rising prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant global public health challenge. Bile acids (BAs), synthesized in the liver and further metabolized in the gut, are essential in maintaining host metabolic homeostasis. Bile salt hydrolase (BSH), an enzyme produced by the gut microbiota, catalyzes the hydrolysis of conjugated BAs, thus regulating the balance between primary and secondary BAs. Growing evidence suggests that BSH activity is intricately linked to the pathogenesis of MASLD. This review comprehensively examines the structural and functional properties of BSH enzymes, their distribution among gut microbial communities, and current methodologies for assessing BSH expression and activity. Furthermore, it highlights the alterations in BSH observed in MASLD and explores the potential mechanistic pathways involved, offering a foundation for the development of novel diagnostic and therapeutic strategies.
Keywords
INTRODUCTION
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease globally, characterized by hepatic fat accumulation that can progress from simple steatosis [metabolic dysfunction-associated steatosis (MASL)] to steatohepatitis [metabolic dysfunction-associated steatohepatitis (MASH)], fibrosis, and even cirrhosis and hepatocellular carcinoma[1]. MASLD is linked not only to liver-related complications but also to extrahepatic manifestations such as cardiovascular disease, chronic kidney disease, and certain cancers[2]. With a global prevalence of approximately 38.77%, MASLD has become a major global public health concern, imposing a heavy burden on individuals and society[3]. Therefore, the development of effective strategies for the prevention, screening, and treatment of MASLD is of great importance[4].
Bile acid (BA) metabolism and its interaction with the gut microbiota play important roles in modulating host immunity and influencing the pathogenesis of MASLD[5,6]. Primary bile acids (PBAs) are synthesized from cholesterol in the liver via both classical and alternative pathways. These PBAs are conjugated with taurine or glycine to form conjugated bile acids (conBAs), which are stored in the gallbladder and released into the intestine upon food intake to facilitate lipid digestion. In the intestine, the gut microbiota converts PBAs into secondary bile acids (secBAs), primarily deoxycholic acid (DCA) and lithocholic acid (LCA). Both PBAs and secBAs can activate receptors such as the farnesol X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5), which regulate glucose and lipid metabolism as well as immune responses[7].
During the microbial transformation of BAs, certain gut bacteria that express bile salt hydrolase (BSH) enzymes can deconjugate conBAs into unconjugated bile acids (unconBAs), initiating further modifications such as dehydroxylation and epimerization[5]. As such, BSH is often regarded as the “gatekeeper” of BA modifications in the gut[8], with significant influence on host metabolic processes including lipid digestion and cholesterol metabolism. Recent studies have highlighted close correlations between BSH activity and metabolic diseases, suggesting that BSH could serve as a therapeutic target in MASLD[9-11]. This review provides an overview of the structural features and functions of BSH, its distribution among gut bacteria, and current evaluation methods. It also examines changes in microbial BSH activity during the progression of MASLD, offering insights into the complex relationship between BSH, BA metabolic balance, and MASLD pathogenesis, with the goal of identifying novel therapeutic strategies for this widespread disease.
BILE SALT HYDROLASE
Molecular structure of BSH
BSH, classified as EC 3.5.1.24 in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, is an enzyme that hydrolyzes the amide bond in conjugated bile acids, converting them into free bile acids and amino acids. It belongs to the N-terminal nucleophile (Ntn) hydrolase family and typically consists of
Metabolic function of BSH
BSH plays a pivotal role in BA metabolism by deconjugating bile salts, thereby altering their physicochemical properties and influencing their absorption and reabsorption in the intestine. Deconjugation is a key step in the enterohepatic circulation of BAs, as it affects the composition of the BA pool and impacts processes such as lipid emulsification, cholesterol metabolism, and gut microbial ecology. BSH is traditionally understood to function by hydrolyzing amide bonds in conBAs, releasing free BAs and their respective amino acid residues[5]. However, recent studies have uncovered that BSH can also catalyze the conjugation of unconBAs with amines to form bacterial bile acid amidates (BBAAs). This discovery challenges the conventional understanding of BSH functions and provides new insights into its broader role in BA metabolism[14].
Distribution of BSH in the gut microbiota
BSH is widely distributed among gut bacteria species, with activity levels varying depending on bacterial strain and environmental conditions. BSH genes have been identified in Lactobacillus, Bifidobacterium, Clostridium, Enterococcus, Bacteroides, Listeria, Brucella, and Xanthomonas, which are typically associated with high BSH activity[15]. Advances in next-generation sequencing and bioinformatics have significantly enhanced our understanding of the BSH gene in the human gut microbiome. These studies indicate that BSH genes are predominantly expressed in members of the phyla Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Euryarchaeota, with Lactobacillus species showing the highest BSH activity in the human gut[9,16]. Despite these findings, the structural and functional diversity of BSH enzymes across different microbial taxa remains insufficiently understood.
Evaluation methods for changes in gut microbial BSH
The expression and activity of microbial BSH are typically evaluated by measuring changes in the composition and concentration of the BA pool, BSH gene expression levels, or in vitro enzymatic activity. Four common tools are used for these measurements: mass spectrometry, ninhydrin colorimetry, microbial sequencing, and activity probes [Table 1]. Mass spectrometry offers high sensitivity and specificity, enabling the precise quantification of BSH activity by measuring the concentrations of conBAs and unconBAs to determine the hydrolysis rate[17,18]. Ninhydrin colorimetry is a traditional method that detects amino acids released by BSH activity through a colorimetric reaction[19]. Although less specific and accurate than modern techniques, it remains a simple and cost-effective approach for evaluating enzyme activity in vitro. Microbial sequencing can infer changes in BSH expression by analyzing homologous gene sequences, copy numbers, or the relative abundance of BSH-expressing bacterial taxa[9-11,16,20-22]. Moreover, recent developments in activity-based probes have significantly improved the precision of BSH activity measurement[23,24]. Probes such as Ch-AOMK bind specifically to BSH active sites and enhance quantitative analysis via mass spectrometry. These probes hold great potential for non-invasive diagnostics and therapeutic monitoring in conditions such as inflammatory bowel disease (IBD)[24].
Evaluation methods for intestinal BSH activity
Method | Target compounds/microbes | Sample | BSH activity indicators | Pros and cons | References |
Mass spectrometry | Bile acid profiles (e.g., GCDCA, CDCA) | Cecal contents, serum, feces | Ratio of CDCA to TCDCA | Pros: high accuracy, high throughput Cons: expensive | [17,18] |
Ninhydrin colorimetry | Free amino acids released from conjugated BAs | Feces | Rate of CA generation | Pros: cost-effective Cons: low specificity | [19] |
Microbial sequencing | |||||
16s rRNA | Bacteroides, Clostridium, Lactobacillus | Feces | Relative abundance of representative BSH-producing bacteria | Pros: established method Cons: indirect reflection of BSH gene expression | [20] |
Bacteroides, Lactobacillus, Bifidobacterium | [21] | ||||
Bifidobacterium, Lactobacillus | [22] | ||||
16s rRNA | Various species | Feces | Relative abundance of BSH genes | Pros: easy to perform Cons: limited by database coverage and update frequency | [9,10] |
Metagenomics | Various species | Feces | Relative abundance of BSH homologous sequences | Pros: high accuracy, comprehensive analysis Cons: poor data quality and parameter consistency | [9,16] |
Activity probe | Ch-AOMK, BAL | Bacteria, feces | Fluorescence intensity | Pros: high sensitivity Cons: expensive | [23,24] |
CHANGES IN MICROBIAL BSH IN MASLD
Microbial BSH plays a pivotal role in the pathophysiology of MASLD. MASLD is characterized by excessive fat accumulation in the liver, and recent evidence suggests that alterations in gut microbial BSH may influence disease progression through modulation of BA metabolism. This section summarizes the changes in microbial BSH across the disease spectrum, from MASLD to its more severe forms including MASH and liver fibrosis [Table 2].
Changes in BSH expression or activity across different stages of MASLD
BSH changea | Subjects | Ethnicity/animal model | Intervention | Group | BSH activity evaluation method | Microbiome sequencing method | References | |
Metabolic dysfunction-associated steatotic liver disease (MASLD) | ||||||||
↓ | Adults | American | / | mild MASLD (72) vs. Control (308) | BSH gene abundance | Metagenomics | [9,59,60] | |
↓ | Children | Asian | / | MASLD (32) vs. HC (36) | Microbial relative abundance, KEGG analysis | Metagenomics | [25] | |
↑ | Adults | American | / | MASLD (57) vs. HC (18) | Microbial relative abundance | 16s rRNA | [11] | |
↓ | Mice | C57BL/6J | / | HFD (6) vs. Chow (9) | Microbial relative abundance | 16s rRNA | [22] | |
↓ | Mice | C57BL/6J | Drug | HFD + IsA (6) vs. Chow vs. HFD (6) | In vitro assay | 16s rRNA | [28] | |
↓ | Mice | C57BL/6J | Drug | HFD (8) vs. CN (8) | Microbial relative abundance | 16s rRNA | [29] | |
↓ | Mice | C57BL/6J | Drug | ZKY (6) vs. HFD (6) vs. Con (6) | BSH gene copy number | 16s rRNA | [26] | |
↑ | Rats | Wistar | BSH inhibitor | AAA-10 (8) vs. Vehicle (8) | In vitro assay | / | [31] | |
↑ | Mice | C57BL/6J | BSH inhibitor | CAPE (5) vs. Vehicle (5) | In vitro assay | 16s rRNA | [17] | |
↑ | Mice | C57BL/6J | Drug | PCPE (8) vs. HFD (8) | In vitro assay | 16s rRNA | [30] | |
Metabolic-Associated Steatohepatitis (MASH) | ||||||||
↓ | Mice | C57BL/6J | Probiotics | ALDH2-/--MCD (5) vs. WT-MCD (5) | In vitro assay | 16s rRNA | [32] | |
↓ | Mice | C57BL/6J | Drug | OCA (8) vs. Vehicle (8) | In vitro assay | 16s rRNA | [34] | |
↑ | Mice | C57BL/6J | / | HFHC (8) vs. HF (8) vs. Con (8) | Microbial relative abundance | 16s rRNA | [33] | |
Advanced liver diseases (Fibrosis and cirrhosis) | ||||||||
↓ | Adults | Asian | / | Hepatic fibrosis (17) vs. HC (10) | Microbial relative abundance, bile acid profile analysis | 16s rRNA | [18] | |
↓ | Adults | American | / | Advanced MASLD (Fibrosis,14) vs. Control (308) | BSH gene abundance | Metagenomics | [9,59,60] | |
↓ | Adults | Asian | / | Liver cirrhosis (114) vs. Control (123) | BSH gene abundance | Metagenomics | [9,38] | |
↓ | Mice | C57BL/6J | Probiotics | TAA (6) vs. Control (6) | Bile acids profile analysis | 16s rRNA | [18] | |
↓ | Mice | C57BL/6J | / | Tlr4-/- (8) vs. WT (10) | Microbial relative abundance | 16s rRNA | [36] | |
↓ | Mice | C57BL/6J | Drug | DDC + KH (5) vs. DDC+H2O (5) | Microbial relative abundance | 16s rRNA | [35] | |
↓ | Mice | C57BL/6J | Drug | DDC (5) vs. Control (5) | In vitro assay | 16s rRNA | [19] | |
↓ | Mice | C57BL/6J | Probiotics | BDL + LGG (7) vs. BDL (7) | In vitro assay | / | [37] |
MASLD
Both clinical and animal studies consistently report a significant reduction in microbial BSH in MASLD. Two clinical studies from the USA and China showed that a significantly reduced abundance of BSH genes in the gut microbiome of MASLD patients[9]. Additionally, a metagenomic study in children with MASLD reported that the conversion of PBAs to SecBAs was inhibited, which was supported by a marked decrease in BSH-expressing genera (Bacteroides and Eubacterium)[25]. Numerous animal studies have further validated the reduction of BSH in MASLD mouse models. Modulating BA metabolism by targeting
However, not all findings are consistent. Some studies have reported increased microbial BSH expression in MASLD. In these cases, reducing BSH expression or suppressing BSH activity was found to increase the conBA/unconBA ratio, reduce cholesterol levels, and limit lipid accumulation. Elevated microbial BSH activity may raise concentrations of certain cytotoxic unconBAs, such as glycodeoxycholic acid,
MASH
MASH represents a more severe and progressive form of MASLD, characterized by liver inflammation and hepatocellular damage, and is closely associated with an increased risk of liver fibrosis. Although research on changes in BSH activity in MASH is limited, existing studies suggest that BSH expression is closely related to the metabolic functions of specific bacterial taxa. These bacteria utilize BSH to modulate the composition and concentration of the BA pool, particularly unconBAs. For example, reduced BSH expression in MASH mice has been associated with a decreased abundance of Lactobacillus in the gut microbiome. Supplementation with Lactobacillus has been shown to promote the formation of LCA, which can activate the FXR signaling pathway, thereby improving liver inflammation and fat accumulation[32].
Conversely, other studies have reported a significant increase in BSH-expressing bacteria, such as Bacteroides, Clostridium, and Lactobacillus, in MASH mice fed a high-fat, high-cholesterol diet. This increase was positively correlated with elevated levels of unconBAs in the liver. Among these, excessive levels of DCA and chenodeoxycholic acid (CDCA) were found to significantly upregulate the expression of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), exacerbating inflammation and hepatic steatosis[33]. Furthermore, the enrichment of Bacteroides has been linked to enhanced hydrolysis of TconBAs, resulting in the overproduction of CDCA in mice fed a Western diet. This, in turn, induces mitochondrial reactive oxygen species (ROS) accumulation and lipid peroxidation in the liver in a dose-dependent manner[34].
Liver fibrosis and cirrhosis
As the disease progresses, excessive extracellular matrix accumulation can drive the progression of MASH to advanced liver fibrosis and ultimately cirrhosis. Existing studies have consistently reported decreased microbial BSH gene expressions in liver fibrosis, leading to a significant accumulation of conBAs in the intestine. This accumulation may inhibit the FXR/FGF-15 signaling pathway, thereby aggravating liver damage[19]. Notably, the enrichment of BSH-expressing genera such as Erysipelotrichaceae, Lactobacillus, Clostridium, and Bifidobacterium[35,36], as well as probiotic interventions with BSH-active strains[18,37], has shown beneficial effects in reversing liver fibrosis. For example, one study reported that the fecal CDCA/taurochenodeoxycholic acid (TCDCA) ratio was significantly lower in patients with liver fibrosis compared to healthy individuals, suggesting reduced microbial BSH activity[18]. The study also observed a marked decrease in Parabacteroides distasonis, a bacterium known for high BSH expression. Supplementation with P. distasonis alleviated liver fibrosis by enhancing BSH activity and promoting TCDCA hydrolysis, thereby reducing hepatotoxicity. Similarly, probiotic treatment with Lactobacillus rhamnosus GG (LGG) demonstrated therapeutic effects in murine models of liver fibrosis[37]. LGG supplementation increased microbial BSH activity, facilitating BA deconjugation and promoting BA excretion via feces and urine, finally preventing BA-induced liver fibrosis by enhancing intestinal FXR-FGF-15 signaling. Additionally, LGG promoted the deconjugation of
Causes of contradictory changes in BSH
The inconsistent findings regarding BSH activity in MASLD may be attributed to several factors. First, clinical and animal studies differ in sample sources, detection methods, and disease modeling approaches. Second, the heterogeneity of MASLD itself may lead to dynamic changes in BSH activity among different populations. Third, BSH-expressing bacteria may exhibit strain-specific functional differences; for example, Lactobacillus and Bacteroides strains may behave differently in the context of MASLD[8]. Song et al. reported that while atherosclerotic patients exhibited reduced total BSH abundance in the gut microbiota, the relative abundance of high-activity BSH subtypes was significantly increased, whereas low-activity subtypes were decreased[16]. Lastly, structural variations and substrate specificity among microbial BSH enzymes may produce divergent effects on BA metabolism (i.e., the relative composition and absolute concentration of both conBAs and unconBAs). For example, several studies have reported an increase in pro-inflammatory DCA and a decrease in hepatoprotective UDCA in MASLD. To summarize, these contradictory findings underscore the complexity of microbial BSH’s role in MASLD development and the need for further mechanistic research to clarify its function and therapeutic potential.
BIOLOGICAL MECHANISMS UNDERLYING BACTERIAL BSH-DRIVEN THERAPEUTIC INTERVENTIONS
This section summarizes three main biological mechanisms through which targeting microbial BSH may contribute to the prevention and treatment of MASLD [Figure 1].
Figure 1. Potential therapeutic role of targeting microbial BSH expression in MASLD. MASLD: Metabolic dysfunction-associated steatotic liver disease; BSH: bile salt hydrolase; T-β-MCA: tauro-β-muricholic acid; FXR: farnesol X receptor; conBAs: conjugated bile acids; unconBAs: unconjugated bile acids; BAT: brown adipose tissue.
Microbial BSH-mediated modulation of BA metabolism
Regulating BA deconjugation through BSH represents a promising therapeutic approach for managing MASLD. Probiotics, prebiotics, and BA sequestrants have shown potential in modulating BA metabolism and thereby improving liver health in MASLD patients[39]. Compared to conBAs, unconBAs are more hydrophobic and less soluble, facilitating their excretion or utilization by gut bacteria[40]. This process triggers a feedback mechanism that stimulates the synthesis of primary BAs, thereby enhancing cholesterol utilization[41]. Probiotics offer unique advantages in MASLD management, with BSH activity playing a crucial role in their colonization and growth in the gastrointestinal tract[42]. Several well-characterized
Modulation of FXR and TGR5 signaling pathways to improve glucose and lipid metabolism
FXR and TGR5 are two key BA receptors involved in regulating energy expenditure, reducing inflammation, and maintaining gut barrier integrity, making them attractive therapeutic targets for MASLD[7]. Microbial BSH influences the composition and concentration of secBAs, which can in turn modulate FXR and TGR5 signaling pathways.
Importantly, FXR exhibits tissue-specific effects - acting differently in the liver and intestine. Intestinal BSH inhibition can lead to the accumulation of endogenous FXR antagonists, thereby helping to restore metabolic balance in the gut. Several studies have reported that BSH inhibitors[17], antioxidants[46], and natural compounds[47] can reduce microbial BSH expression and/or activity in mouse models, resulting in increased levels of T-β-MCA, an endogenous FXR antagonist. This suppression of FXR signaling in ileal epithelial cells reduces ceramide synthesis and promotes GLP-1 secretion, ultimately improving metabolic outcomes. Conversely, supplementation with BSH-active Lactobacillus plantarum increases CDCA levels, which suppress hepatic lipogenesis and insulin resistance[48] via FXR signaling. Activation of hepatic FXR inhibits triglyceride production by downregulating the SREBP-1c lipogenesis pathway[49]. Additionally, FXR agonists such as obeticholic acid have been shown to promote brown fat differentiation and energy metabolism[50].
TGR5 also plays an important role in glucose and lipid metabolism and in mitigating inflammation in MASLD. Bariatric surgery, known for its metabolic benefits, increases levels of intestinal and circulating taurine-conjugated BAs, which in turn activate FXR and TGR5 signaling, thereby stimulating adaptive thermogenesis[51]. Salidroside has been found to alleviate lipid accumulation and inflammatory injury in MASH mice by increasing the abundance of BSH-expressing bacteria and decreasing conjugated BA levels, thereby enhancing downstream FXR and TGR5 activation[52]. Interestingly, oral administration of live Parabacteroides distasonis has been shown to alleviate inflammatory arthritis. This effect was attributed to its BSH-derived metabolites, 3-oxoLCA and isoLCA, which act as TGR5 agonists and promote M2 macrophage polarization[53].
Inhibiting NLRP3 to alleviate inflammation
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is an intracellular sensor implicated in the pathogenesis of various metabolic diseases, including MASLD. BAs can dose-dependently activate the NLRP3 inflammasome, leading to IL-1β secretion and liver fibrosis[54]. This activation occurs through mechanisms such as potassium efflux and ROS generation. In mouse models, reduced BSH activity lowers levels of secondary BAs like nor-deoxycholic acid (NorDCA), which in turn suppresses NLRP3 activation and mitigates liver inflammation[55]. Sun et al. found that Bacteroides dorei BDX-01 enhanced BSH protein expression in the intestines of mice, increasing the β-MCA to T-β-MCA ratio and decreasing NLRP3 and IL-1β expression in the colon[56]. These changes significantly alleviated intestinal inflammation. Notably,
CHALLENGES AND FUTURE DIRECTIONS
As the “gatekeeper” responsible for initiating secondary modifications of BAs in the gut, BSH plays a pivotal role in linking the gut microbiome with BA metabolism. Although no consensus has been reached regarding the precise pattern of BSH changes in individuals with MASLD, it is evident that such changes are closely related to disease progression. Most studies have demonstrated that intestinal BSH expression or activity tends to decline in more advanced stages of MASLD, such as the onset of hepatitis or fibrosis. These changes primarily affect the ratio of conjugated to unconjugated BAs, thereby disrupting the dynamic balance of the BA metabolic network. This, in turn, influences enterohepatic circulation and various metabolic signaling pathways.
Challenges in evaluating BSH activity in MASLD
Several factors contribute to the current uncertainties regarding the role of BSH in MASLD progression. First, the degree of disease severity varies among individuals, leading to differences in gut microbiome composition and BA profiles. Clinical studies have shown that MASLD patients with steatohepatitis exhibit significantly elevated serum levels of taurocholic acid (TCA), glycocholic acid (GCA), and taurolithocholic acid (TLCA) compared to those with simple steatosis[57]. The worsening of hepatic steatosis, inflammation, and ballooning degeneration is closely linked to the increase in these conBAs. Second, because BAs serve diverse biological functions, microbial BSH directly influences the levels of both beneficial and harmful secondary BAs. Consequently, studies focusing on specific BAs may yield varying conclusions regarding the relationship between BSH activity and disease progression. Third, the BA pool in mice contains a higher proportion of primary BAs and a broader range of secondary BAs than in humans[5], leading to potential discrepancies in BSH-related findings across species. Lastly, different methodologies have been employed to measure BSH expression and activity, with varying levels of sensitivity and accuracy, making direct comparison across studies challenging.
Therapeutic implications and potential interventions targeting BSH
Studying the role of microbial BSH in host metabolism presents methodological challenges due to the variability in gut microbiota composition and the complexity of BA metabolism. Longitudinal studies are therefore necessary to understand the long-term effects of microbial BSH on MASLD progression and to identify reliable biomarkers for disease monitoring. In addition, personalized approaches that account for individual differences in gut microbiota and metabolic responses will be critical for the development of effective BSH-targeted therapies.
Recent advances in microbiome research, metagenomics, and metabolomics are opening new avenues for understanding the role of BSH in MASLD and for developing targeted interventions. BSH not only supports bacterial colonization and survival in the gastrointestinal tract but also significantly affects host glucose and lipid metabolism and the regulation of inflammation. Its regulatory effects are mediated through BAs, especially via alterations in the composition and spatial distribution of secondary BAs, which interact with specific receptors and the NLRP3 inflammasome. These interactions indirectly influence the synthesis and secretion of key metabolic regulators such as cholesterol, ceramides, and GLP-1, contributing to the restoration of metabolic balance. This highlights the potential of BSH as a therapeutic target for MASLD. Consequently, the use of probiotics or natural small-molecule compounds to modulate microbial BSH gene expression or enzyme activity has become a focal point of research aimed at improving MASLD-related metabolic disorders[28].
Although extensive studies have explored the structure, function, and substrate specificity of various BSH enzymes, much remains to be discovered about gut bacteria and their functions[58]. With ongoing advancements in sequencing technologies, additional BSH variants are expected to be identified. Understanding the enzymatic properties of these variants will be crucial for developing personalized probiotics and more effective diagnostic and therapeutic tools.
CONCLUSION
Gut microbial BSH activity tends to decline during the development and progression of MASLD due to factors such as changes in gut microbiota composition, host metabolic status, inflammation, and dietary influences. BSH plays a critical role in maintaining BA metabolic balance and significantly impacts the progression and management of MASLD. A deeper understanding of the intricate relationship between gut microbiota, BSH, and MASLD will provide valuable insights for the development of targeted therapeutic strategies. Further research is needed to clarify the mechanisms by which BSH changes influence liver health and to devise effective interventions for MASLD. Targeting BSH represents a promising approach for modulating BA metabolism and improving metabolic health, highlighting the crucial role of the gut microbiota in disease management.
DECLARATIONS
Acknowledgments
Graphic elements used in the figures were obtained from BioGDP (https://www.biogdp.com) and assembled by the authors.
Authors’ contributions
Drafted and revised the manuscript: Zhao W, Ni Y, Wang H, Zheng M
Conceived and designed this project: Ni Y
Availability of data and materials
Not applicable.
Financial support and sponsorship
This work was supported by grants from the National Natural Science Foundation of China (82170583 and 81900510) and the National Key Research and Development Program of China (2021YFC2701900).
Conflicts of interest
All authors declared that there are no conflicts of interest.
Ethical approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Copyright
© The Author(s) 2025.
REFERENCES
1. Loomba R, Friedman SL, Shulman GI. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell. 2021;184:2537-64.
2. Miao L, Targher G, Byrne CD, Cao YY, Zheng MH. Current status and future trends of the global burden of MASLD. Trends Endocrinol Metab. 2024;35:697-707.
3. Chan KE, Koh TJL, Tang ASP, et al. Global prevalence and clinical characteristics of metabolic-associated fatty liver disease: a meta-analysis and systematic review of 10 739 607 individuals. J Clin Endocrinol Metab. 2022;107:2691-700.
4. Sharpton SR, Schnabl B, Knight R, Loomba R. Current concepts, opportunities, and challenges of gut microbiome-based personalized medicine in nonalcoholic fatty liver disease. Cell Metab. 2021;33:21-32.
5. Collins SL, Stine JG, Bisanz JE, Okafor CD, Patterson AD. Bile acids and the gut microbiota: metabolic interactions and impacts on disease. Nat Rev Microbiol. 2023;21:236-47.
6. Lee MH, Nuccio SP, Mohanty I, et al. How bile acids and the microbiota interact to shape host immunity. Nat Rev Immunol. 2024;24:798-809.
8. Foley MH, Walker ME, Stewart AK, et al. Bile salt hydrolases shape the bile acid landscape and restrict Clostridioides difficile growth in the murine gut. Nat Microbiol. 2023;8:611-28.
9. Jia B, Park D, Hahn Y, Jeon CO. Metagenomic analysis of the human microbiome reveals the association between the abundance of gut bile salt hydrolases and host health. Gut Microbes. 2020;11:1300-13.
10. Sun L, Zhang Y, Cai J, et al. Bile salt hydrolase in non-enterotoxigenic Bacteroides potentiates colorectal cancer. Nat Commun. 2023;14:755.
11. Smirnova E, Muthiah MD, Narayan N, et al. Metabolic reprogramming of the intestinal microbiome with functional bile acid changes underlie the development of NAFLD. Hepatology. 2022;76:1811-24.
12. Begley M, Hill C, Gahan CGM. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol. 2006;72:1729-38.
13. Xu F, Hu XJ, Singh W, Geng W, Tikhonova IG, Lin J. The complex structure of bile salt hydrolase from Lactobacillus salivarius reveals the structural basis of substrate specificity. Sci Rep. 2019;9:12438.
14. Rimal B, Collins SL, Tanes CE, et al. Bile salt hydrolase catalyses formation of amine-conjugated bile acids. Nature. 2024;626:859-63.
15. Bustos AY, de Valdez GF, Fadda S, Taranto MP. New insights into bacterial bile resistance mechanisms: the role of bile salt hydrolase and its impact on human health. Food Res Int. 2018;112:250-62.
16. 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.
17. Zhong XC, Liu YM, Gao XX, et al. Caffeic acid phenethyl ester suppresses intestinal FXR signaling and ameliorates nonalcoholic fatty liver disease by inhibiting bacterial bile salt hydrolase activity. Acta Pharmacol Sin. 2023;44:145-56.
18. Zhao Q, Dai MY, Huang RY, et al. Parabacteroides distasonis ameliorates hepatic fibrosis potentially via modulating intestinal bile acid metabolism and hepatocyte pyroptosis in male mice. Nat Commun. 2023;14:1829.
19. Liu X, Li J, Shi M, et al. Melatonin improves cholestatic liver disease via the gut-liver axis. J Pineal Res. 2024;76:e12929.
20. Wan Y, Yuan J, Li J, et al. Unconjugated and secondary bile acid profiles in response to higher-fat, lower-carbohydrate diet and associated with related gut microbiota: a 6-month randomized controlled-feeding trial. Clin Nutr. 2020;39:395-404.
21. Jiao N, Baker SS, Chapa-Rodriguez A, et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut. 2018;67:1881-91.
22. He X, Gao X, Hong Y, et al. High fat diet and high sucrose intake divergently induce dysregulation of glucose homeostasis through distinct gut microbiota-derived bile acid metabolism in mice. J Agric Food Chem. 2024;72:230-44.
23. Parasar B, Zhou H, Xiao X, Shi Q, Brito IL, Chang PV. Chemoproteomic profiling of gut microbiota-associated bile salt hydrolase activity. ACS Cent Sci. 2019;5:867-73.
24. Khodakivskyi PV, Lauber CL, Yevtodiyenko A, et al. Noninvasive imaging and quantification of bile salt hydrolase activity: from bacteria to humans. Sci Adv. 2021;7:eaaz9857.
25. Yu J, Zhang H, Chen L, Ruan Y, Chen Y, Liu Q. Disease-associated gut microbiota reduces the profile of secondary bile acids in pediatric nonalcoholic fatty liver disease. Front Cell Infect Microbiol. 2021;11:698852.
26. Li Y, Wang H, He X, et al. Zhi-Kang-Yin formula attenuates high-fat diet-induced metabolic disorders through modulating gut microbiota-bile acids axis in mice. Chin Med. 2024;19:145.
27. Jia W, Rajani C, Zheng X, Jia W. Hyocholic acid species improve glucose homeostasis through a distinct TGR5 and FXR signaling mechanism. J Mol Cell Biol. 2021;13:460-2.
28. Zhao WW, Xiao M, Wu X, et al. Ilexsaponin A1 Ameliorates diet-induced nonalcoholic fatty liver disease by regulating bile acid metabolism in mice. Front Pharmacol. 2021;12:771976.
29. Xu H, Yuan M, Niu K, et al. Involvement of bile acid metabolism and gut microbiota in the amelioration of experimental metabolism-associated fatty liver disease by nobiletin. Molecules. 2024;29:976.
30. Li X, Zhao W, Xiao M, et al. Penthorum chinense Pursh. extract attenuates non-alcholic fatty liver disease by regulating gut microbiota and bile acid metabolism in mice. J Ethnopharmacol. 2022;294:115333.
31. Li DK, Chaudhari SN, Lee Y, et al. Inhibition of microbial deconjugation of micellar bile acids protects against intestinal permeability and liver injury. Sci Adv. 2022;8:eabo2794.
32. Li ZM, Kong CY, Mao YQ, et al. Host ALDH2 deficiency aggravates nonalcoholic steatohepatitis through gut-liver axis. Pharmacol Res. 2023;196:106902.
33. Gao X, Lin X, Xin Y, et al. Dietary cholesterol drives the development of nonalcoholic steatohepatitis by altering gut microbiota mediated bile acid metabolism in high-fat diet fed mice. J Nutr Biochem. 2023;117:109347.
34. Zhuge A, Li S, Yuan Y, et al. Microbiota-induced lipid peroxidation impairs obeticholic acid-mediated antifibrotic effect towards nonalcoholic steatohepatitis in mice. Redox Biol. 2023;59:102582.
35. Shen B, Zhou C, Gu T, et al. Kuhuang alleviates liver fibrosis by modulating gut microbiota-mediated hepatic IFN signaling and bile acid synthesis. Front Pharmacol. 2022;13:1080226.
36. Takimoto Y, Chu PS, Nakamoto N, et al. Myeloid TLR4 signaling promotes post-injury withdrawal resolution of murine liver fibrosis. iScience. 2023;26:106220.
37. Liu Y, Chen K, Li F, et al. Probiotic Lactobacillus rhamnosus GG prevents liver fibrosis through inhibiting hepatic bile acid synthesis and enhancing bile acid excretion in mice. Hepatology. 2020;71:2050-66.
38. Qin N, Yang F, Li A, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature. 2014;513:59-64.
39. Kobyliak N, Abenavoli L, Falalyeyeva T, et al. Beneficial effects of probiotic combination with omega-3 fatty acids in NAFLD: a randomized clinical study. Minerva Med. 2018;109:418-28.
40. Fogelson KA, Dorrestein PC, Zarrinpar A, Knight R. The gut microbial bile acid modulation and its relevance to digestive health and diseases. Gastroenterology. 2023;164:1069-85.
41. Chen ML, Yi L, Zhang Y, et al. Resveratrol attenuates trimethylamine- N -oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. mBio. 2016;7:e02210-15.
42. Pontes KSDS, Guedes MR, Cunha MRD, et al. Effects of probiotics on body adiposity and cardiovascular risk markers in individuals with overweight and obesity: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr. 2021;40:4915-31.
43. Agolino G, Pino A, Vaccalluzzo A, et al. Bile salt hydrolase: the complexity behind its mechanism in relation to lowering-cholesterol lactobacilli probiotics. J Funct Foods. 2024; 120:106357.
44. Kumar R, Grover S, Batish VK. Bile salt hydrolase (Bsh) activity screening of lactobacilli: in vitro selection of indigenous lactobacillus strains with potential bile salt hydrolysing and cholesterol-lowering ability. Probiotics Antimicro Prot. 2012;4:162-72.
45. Yang Y, Liu Y, Zhou S, Huang L, Chen Y, Huan H. Bile salt hydrolase can improve Lactobacillus plantarum survival in gastrointestinal tract by enhancing their adhesion ability. FEMS Microbiol Lett. 2019;366:fnz100.
46. Li F, Jiang C, Krausz KW, et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun. 2013;4:2384.
47. Huang F, Zheng X, Ma X, et al. Theabrownin from Pu-erh tea attenuates hypercholesterolemia via modulation of gut microbiota and bile acid metabolism. Nat Commun. 2019;10:4971.
48. Liang C, Zhou XH, Gong PM, et al.
49. Watanabe M, Houten SM, Wang L, et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J Clin Invest. 2004;113:1408-18.
50. Zhang H, Dong M, Liu X. Obeticholic acid ameliorates obesity and hepatic steatosis by activating brown fat. Exp Ther Med. 2021;22:991.
51. Münzker J, Haase N, Till A, et al. Functional changes of the gastric bypass microbiota reactivate thermogenic adipose tissue and systemic glucose control via intestinal FXR-TGR5 crosstalk in diet-induced obesity. Microbiome. 2022;10:96.
52. Zhang J, Zhou J, He Z, et al. Salidroside attenuates NASH through regulating bile acid-FXR/TGR5 signaling pathway via targeting gut microbiota. Int J Biol Macromol. 2025;307:142276.
53. Sun H, Guo Y, Wang H, et al. Gut commensal Parabacteroides distasonis alleviates inflammatory arthritis. Gut. 2023;72:1664-77.
54. Gong Z, Zhou J, Zhao S, et al. Chenodeoxycholic acid activates NLRP3 inflammasome and contributes to cholestatic liver fibrosis. Oncotarget. ;7:83951-63.
55. Yue H, Li Y, Cai W, Bai X, Dong P, Wang J. Antarctic krill peptide alleviates liver fibrosis via downregulating the secondary bile acid mediated NLRP3 signaling pathway. Food Funct. 2022;13:7740-9.
56. Sun X, Chen Z, Yu L, et al. Bacteroides dorei BDX-01 alleviates DSS-induced experimental colitis in mice by regulating intestinal bile salt hydrolase activity and the FXR-NLRP3 signaling pathway. Front Pharmacol. 2023;14:1205323.
57. Puri P, Daita K, Joyce A, et al. The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids. Hepatology. 2018;67:534-48.
58. Liu C, Du MX, Abuduaini R, et al. Enlightening the taxonomy darkness of human gut microbiomes with a cultured biobank. Microbiome. 2021;9:119.
59. Abu-Ali GS, Mehta RS, Lloyd-Price J, et al. Metatranscriptome of human faecal microbial communities in a cohort of adult men. Nat Microbiol. 2018;3:356-66.
Cite This Article

How to Cite
Download Citation
Export Citation File:
Type of Import
Tips on Downloading Citation
Citation Manager File Format
Type of Import
Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.
Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.
About This Article
Special Issue
Copyright
Data & Comments
Data

Comments
Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at [email protected].