Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-19T02:12:25.775Z Has data issue: false hasContentIssue false

Bacterial bile salt hydrolase: an intestinal microbiome target for enhanced animal health

Published online by Cambridge University Press:  03 February 2017

Wenjing Geng
Affiliation:
Department of Animal Science, The University of Tennessee, Tennessee, USA
Jun Lin*
Affiliation:
Department of Animal Science, The University of Tennessee, Tennessee, USA
*
*Corresponding author. E-mail: jlin6@utk.edu
Rights & Permissions [Opens in a new window]

Abstract

To effectively mitigate antimicrobial resistance in the agricultural ecosystem, there is an increasing pressure to reduce and eliminate the use of in-feed antibiotics for growth promotion and disease prevention in food animals. However, limiting antibiotic use could compromise animal production efficiency and health. Thus, there is an urgent need to develop effective alternatives to antibiotic growth promoters (AGPs). Increasing evidence has shown that the growth-promoting effect of AGPs was highly correlated with the reduced activity of bile salt hydrolase (BSH), an intestinal bacterial enzyme that has a negative impact on host fat digestion and energy harvest; consistent with this finding, the population of Lactobacillus species, the major intestinal BSH-producer, was significantly reduced in response to AGP use. Thus, BSH is a key mechanistic microbiome target for developing novel alternatives to AGPs. Despite recent significant progress in the characterization of diverse BSH enzymes, research on BSH is still in its infancy. This review is focused on the function of BSH and its significant impacts on host physiology in human beings, laboratory animals and food animals. The gaps in BSH-based translational microbiome research for enhanced animal health are also identified and discussed.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2017 

Introduction

Antibiotic use clearly serves as a selective driving force to enrich antimicrobial resistance (AMR) genes and promote the emergence of antibiotic-resistant bacterial pathogens (Davies, Reference Davies, Atlas and Maloy2014). Thus, reducing or eliminating the use of in-feed antibiotics in healthy animals has been a worldwide trend to effectively mitigate AMR and protect food safety. US Food and Drug Administration recently implemented a new policy to recommend a voluntary withdrawal of medically important antibiotic from routine animal production practices by December 2016. Therefore, there is an urgent need to develop effective strategies to maintain animal productivity and health without relying on in-feed antibiotics.

Food animal producers have manipulated intestinal microbiota for more than 60 years to increase feed efficiency and body weight gain through the routine use of low-dose antibiotics as feed additives, called antibiotic growth promoters (AGPs). With the aid of culture-independent molecular approaches, investigations of the effect of AGPs on intestinal microbiota have been initiated in different food animals, including poultry and swine (Lin, Reference Lin2014). These microbiome studies have shed light on the mechanism of mode of action of AGPs and on the development of novel alternatives to AGPs. Specifically, data indicate that the body weight gain in food animals is inversely related to the activity of bile salt hydrolase (BSH) as well as the abundance of potent BSH-producing bacteria in the intestine (Lin, Reference Lin2014). Because the BSH enzymes produced by intestinal bacteria catalyze deconjugation of conjugated bile acids, an essential gateway reaction in the metabolism of bile acids which play an important role in host fat metabolism, energy harvest and body weight gain (Begley et al., Reference Begley, Hill and Gahan2006; Joyce et al., Reference Joyce, Shanahan, Hill and Gahan2014b), we propose that BSH is a key mechanistic microbiome target for developing novel alternatives to AGPs, such as BSH inhibitors for enhanced animal production and health. This article reviews recent progress on BSH research, with emphasis on BSH functions and its impact on host physiology.

Bile acids

Primary bile acids are de novo synthesized from cholesterol in the liver and are conjugated to either glycine or taurine to form conjugated bile acids (Appleby and Walters, Reference Appleby and Walters2014; Schaap et al., Reference Schaap, Trauner and Jansen2014; Camilleri and Gores, Reference Camilleri and Gores2015). The amphipathic characteristic of conjugated bile acid helps dietary lipids or fat-soluble vitamins form micelles, which facilitate their metabolism by pancreatic enzymes prior to their absorption (de Aguiar Vallim et al., Reference De Aguiar Vallim, Tarling and Edwards2013). Thus, conjugated bile acids are more efficient than unconjugated bile acids for emulsification and digestion of dietary lipids or lipid soluble nutrients (Hofmann and Mysels, Reference Hofmann and Mysels1992; Ridlon et al., Reference Ridlon, Kang and Hylemon2006). Following synthesis, bile salts are stored and concentrated in the gallbladder. Upon food consumption, chyme from partly digested food is expelled from stomach into the duodenum, acids and partially digested fat stimulate the secretion of secretin and cholecystokinin (CCK) (Begley et al., Reference Begley, Gahan and Hill2005). Subsequently, CCK stimulates the contraction of the gallbladder, and leads to the release of bile salts from the gallbladder into the small intestine for lipid digestion (Johnson, Reference Johnson1998). In animals without a gallbladder, such as horses and rats, bile salts continuously flow directly from the liver to the duodenum via the bile duct.

After reaching the ileum, bile salts are taken up into enterocytes via efficient membrane transporters, further absorbed into the portal vein to get back to the liver and finally re-secreted into bile; this process is called enterohepatic circulation (Vlahcevic et al., Reference Vlahcevic, Heuman and Hylemon1996; Roberts et al., Reference Roberts, Magnusson, Burczynski and Weiss2002; Begley et al., Reference Begley, Hill and Gahan2006; Ridlon et al., Reference Ridlon, Kang and Hylemon2006; Russell, Reference Russell2009). In human beings, approximately 400–800 mg of bile salts daily are subjected to microbial transformations in the intestine (Vlahcevic et al., Reference Vlahcevic, Heuman and Hylemon1996). Among various bile salt transformations, deconjugation of conjugated bile salts is the gateway reaction for bile alteration and is a prerequisite for all sterol transformation (Batta et al., Reference Batta, Salen, Arora, Shefer, Batta and Person1990; Kim and Lee, Reference Kim and Lee2005). Notably, in addition to a direct digestive role in the emulsification of dietary fats in the intestine, bile acids can act as signaling molecules to affect energy metabolism, bile acids enterohepatic circulation, host cholesterol level, and triglyceride and glucose homeostasis (Joyce et al., Reference Joyce, Shanahan, Hill and Gahan2014b). In particular, unconjugated bile acids have been shown to specifically interact with orphan nuclear hormone receptors such as farnesoid X receptor (FXR) and G-protein-coupled receptor TGR5 (Gupta et al., Reference Gupta, Stravitz, Dent and Hylemon2001; Qiao et al., Reference Qiao, Han, Fang, Park, Gupta, Gilfor, Amorino, Valerie, Sealy and Engelhardt2003; Houten et al., Reference Houten, Watanabe and Auwerx2006; Inagaki et al., Reference Inagaki, Moschetta, Lee, Peng, Zhao, Downes, Ruth, Shelton, Richardson and Repa2006; Evans et al., Reference Evans, Mahaney, Borges-Marcucci, Lai, Wang, Krueger, Gardell, Huard, Martinez and Vlasuk2009).

Bile salt hydrolase

The BSH enzyme produced by intestinal bacteria catalyzes deconjugation of conjugated bile acids by hydrolyzing the amide bond and producing free amino acids and unconjugated bile acids; this is an essential gateway reaction in the metabolism of bile acids in the small intestine (Begley et al., Reference Begley, Hill and Gahan2006). BSH enzyme belongs to the choloylglycine hydrolase (EC 3.5.1.24) family. Phylogenetic analysis indicated that BSH was derived from the wider Ntn_CGH-like family of proteins, specifically penicillin V acylase (Kumar et al., Reference Kumar, Brannigan, Prabhune, Pundle, Dodson, Dodson and Suresh2006; Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008).

BSH enzymes from various sources differ in activity, substrate specificity, and optimal temperature and pH for enzymatic activity (Begley et al., Reference Begley, Hill and Gahan2006). Molecular weights of the BSH subunit range from 28 to 50 kDa, and optimal pH for BSH activity is slightly acidic, ranging from 3.5 to 6. Most identified BSH enzymes still display activity at temperatures up to 60°C. Many identified BSH enzymes have a narrow substrate spectrum and display much higher activity in hydrolyzing glycine-conjugated bile salts than taurine-conjugated bile salts (Coleman and Hudson, Reference Coleman and Hudson1995; Smet et al., Reference Smet, Hoorde, Woestyne, Christiaens and Verstraete1995; Tanaka et al., Reference Tanaka, Hashiba, Kok and Mierau2000; Kim et al., Reference Kim, Miyamoto, Meighen and Lee2004; Liong and Shah, Reference Liong and Shah2005; Pavlović et al., Reference Pavlović, Stankov and Mikov2012). However, some BSH enzymes show a preference for taurine-conjugated bile salts, such as two BSH enzymes in Lactobacillus jonsonii PF01 (Chae et al., Reference Chae, Valeriano, Kim and Kang2013) and the BSH enzymes from five lactobacilli strains (Jiang et al., Reference Jiang, Hang, Zhang, Liu, Li and Yang2010). Recently, a potent BSH enzyme was identified and characterized from a chicken Lactobacillus salivarius strain; this BSH displayed potent hydrolysis activity towards both glycol-conjugated and taurine-conjugated bile salts (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012). It has been proposed that BSH enzymes recognize conjugated bile acids on both amino acid moieties and the cholate steroid nucleus (Begley et al., Reference Begley, Hill and Gahan2006). Not surprisingly, substrate preferences of BSH may differ under different pH, likely due to pH-mediated structural changes (Corzo and Gilliland, Reference Corzo and Gilliland1999).

To date, structural basis of BSH function is still largely unknown. Crystal structures of the BSH enzymes from only three specific species, Bifidobacterium longum, Clostridium perfrigens, and L. salivarius have been reported (Rossocha et al., Reference Rossocha, Schultz-Heienbrok, von Moeller, Coleman and Saenger2005; Kumar et al., Reference Kumar, Brannigan, Prabhune, Pundle, Dodson, Dodson and Suresh2006; Xu et al., Reference Xu, Guo, Hu and Lin2016). The 1.90 Å crystal structure of the L. salivarius BSH was recently determined by molecular replacement using the starting model of C. perfringens BSH (Xu et al., Reference Xu, Guo, Hu and Lin2016). Comparative structural analysis of the L. salivarius BSH also identified potential residues contributing to catalysis and substrate specificity. Together, unlike the binding pocket in other BSHs such as the C. perfrigens BSH that shows an open entrance with shallow bottom, a panel of unique residues in the L. salivarius BSH make this BSH display narrow entrance of the binding pocket and the increased inner capacity of the binding pocket, which may enable substrates to sit deeply in the pocket with different conformation and lead to the broad spectrum of specificity (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012; Xu et al., Reference Xu, Guo, Hu and Lin2016). Previous comparative genomics and structural studies have identified some conserved, catalytically important residues in the active site of BSH (Cys2, Arg 16, Asp19, Asn79, Asn171, and Arg224); however, this conclusion was primarily based on the comparison of BSH structure with penicillin V acylase (Begley et al., Reference Begley, Hill and Gahan2006; Kumar et al., Reference Kumar, Brannigan, Prabhune, Pundle, Dodson, Dodson and Suresh2006; Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012). To date, Cys2 is the only residue that has been subjected to site-directed mutagenesis and validated for its essential role in BSH activity (Kumar et al., Reference Kumar, Brannigan, Prabhune, Pundle, Dodson, Dodson and Suresh2006). Therefore, future in-depth structural analysis of the unique L. salivarius BSH (e.g. in complex with specific substrate) in conjunction with comprehensive amino acid substitution mutagenesis would help to discover residues critical in catalysis and understand why this BSH displayed potent catalytic activity toward a broad spectrum of substrates including both glycol-conjugated and taurine-conjugated bile salts.

BSH-producing bacteria in the intestine

BSH enzymes have been identified in diverse bacterial species from different sources (Summarized in Table 1). Among the BSH-producing organisms, most of them are Gram-positive bacteria, except two from the Gram-negative genus, Bacteroides (Stellwag and Hylemon, Reference Stellwag and Hylemon1976; Masuda, Reference Masuda1981; Lambert et al., Reference Lambert, Bongers, de Vos and Kleerebezem2008). Jones et al. (Reference Jones, Begley, Hill, Gahan and Marchesi2008) performed a functional and comparative metagenomic analysis of BSH activity in the human intestinal microbiome and showed a high level of redundancy of BSH distribution in the human intestine ecosystem; most BSH activity was distributed in all major phyla within intestinal microbiota (primarily Firmicutes, followed by Bacterioidetes and Actinobacteria) and across two domains of life (Bacteria and Archaea in the intestine) (Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008).

Table 1. The BSH enzymes identified in bacteria from various sources

a Molecular mass of tetramer.

ATCC = American type culture collection, JCM = Japanese collection of microorganisms, CGMCC = China general microbiological culture collection center, NRRL = Northern regional research laboratory, the agricultural research service culture collection, ND = not determined.

BSH genes are particularly abundant in lactic acid fermenting probiotics, such as lactobacilli and bifidobacteria, which are the species most commonly used as probiotics due to their health-promoting activities (Reviewed by Begley et al., Reference Begley, Hill and Gahan2006). As shown in Table 1, BSH activity and corresponding enzymes have been identified primarily in lactic acid bacteria isolated from the gastrointestinal tract, which include but are not limited to L. salivarius, Lactobacillus acidophilus, Lactobacillus johnsonii, Lactobacillus plantarum, Bifidobacterium longum, Bifidobacterium bifidum, Bifidobacterium adolescentis, and Bifidobacterium animalis. BSH genes are either located in the chromosome or in mobile element, such as the megaplasmid identified in L. salivarius UCC118 (Claesson et al., Reference Claesson, Li, Leahy, Canchaya, van Pijkeren, Cerdeño-Tárraga, Parkhill, Flynn, O'Sullivan and Collins2006). It is not unusual that multiple BSH homologs, which are not identical, could be present in a single intestinal bacterial strain (Begley et al., Reference Begley, Hill and Gahan2006; Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012). It has been speculated that BSH genes may be acquired horizontally among intestinal microorganisms (Begley et al., Reference Begley, Hill and Gahan2006). However, there is no compelling evidence demonstrating horizontal transfer of BSH genes in intestinal microorganisms.

Jones et al. (Reference Jones, Begley, Hill, Gahan and Marchesi2008) also have determined that active BSH enzymes are restricted to intestinal microorganisms, suggesting that BSH activity plays a role in in vivo adaptation of intestinal microorganisms in the gastrointestinal environment and in the mutualism between intestinal microbiota and animal hosts (Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008). Physiological advantages of BSH for bacterial producers themselves are still not well understood. One popular opinion is that BSH activity contributes to the resistance of commensal bacteria towards bile salts, a natural antimicrobial present in the intestine (Begley et al., Reference Begley, Hill and Gahan2006). For example, it has been demonstrated that BSH activity plays an important role in the bile resistance and intestinal colonization of Listeria innocua in a mouse model (Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008). However, the unconjugated bile salts resulting from BSH hydrolysis could still display antimicrobial activity; thus, there are contradictory findings about contribution of BSH activity to bile tolerance in intestinal probiotic bacteria (Begley et al., Reference Begley, Hill and Gahan2006). At present, there is no convincing in vivo evidence demonstrating that BSH enzyme contributes to bile resistance in probiotic bacteria, such as lactobacilli. Fang et al. (Reference Fang, Li, Bumann, Raftis, Casey, Cooney, Walsh and O'Toole2009) demonstrated that production of BSH does not determine the bile resistance level in L. salivarius, the dominant Lactobacillus species present in animal intestine (Fang et al., Reference Fang, Li, Bumann, Raftis, Casey, Cooney, Walsh and O'Toole2009). In addition to this popular hypothesis, there are some other opinions about the roles of BSH in bacterial physiology based on some evidence in certain commensal bacteria. For example, it has been proposed that hydrolysis of conjugated bile acids by BSH can provide cellular carbon, nitrogen, sulfur as well as energy source for some bacteria species (Vlahcevic et al., Reference Vlahcevic, Heuman and Hylemon1996; Tanaka et al., Reference Tanaka, Hashiba, Kok and Mierau2000; Ridlon et al., Reference Ridlon, Kang and Hylemon2006). BSH may also trigger the influx of cholesterol or bile into bacterial cells and increase membrane electrochemical characteristics, which may facilitate some microorganisms to inhabit in the gastrointestinal epithelium in the host via immune evasion (Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008; Mukherji and Prabhune, Reference Mukherji and Prabhune2015).

The impact of bacterial BSH activity on host physiology

Despite the lack of understanding of the benefits of BSH for BSH-producing bacteria, it has been well recognized that intestinal BSH plays an important role in host lipid metabolism, dietary energy harvest and body weight gain because BSH catalyzes the gateway reaction in the metabolism of bile acids in the intestine (Begley et al., Reference Begley, Hill and Gahan2006; Jones et al., Reference Jones, Begley, Hill, Gahan and Marchesi2008; Joyce et al., Reference Joyce, Shanahan, Hill and Gahan2014b). To date, functional research on the relationship between bacterial BSH and host physiology/health have been primarily focused on human probiotics using laboratory animal model systems. There are very limited efforts to determine the impact of intestinal bacterial BSH activity on growth and health in food animals (Feighner and Dashkevicz, Reference Feighner and Dashkevicz1988; Knarreborg et al., Reference Knarreborg, Lauridsen, Engberg and Jensen2004; Guban et al., Reference Guban, Korver, Allison and Tannock2006; Lin, Reference Lin2011). The following paragraphs summarize findings from laboratory animal studies and human trials, which shed light on future directions for food animal health research.

Host lipid metabolism, cholesterol, and body weight

As children and adults are increasingly becoming overweight and obese, obesity-associated diseases will increase (Kahn et al., Reference Kahn, Hull and Utzschneider2006; Van Gaal et al., Reference Van Gaal, Mertens and Christophe2006). Recent studies have indicated that intestinal microbiota are implicated in obesity in people (Tremaroli and Bäckhed, Reference Tremaroli and Bäckhed2012); however, key microbial functions influencing host energy harvest remain to be clearly elucidated. The BSH enzyme has been increasingly recognized as a critical intestinal microbiome target for developing intervention strategy to control obesity.

Given that the bile acids have dual digestive and signaling roles in the host, intestinal BSH plays an important role in host metabolism and energy harvest; BSH activity has significant impacts on host physiology by disturbing conjugated bile acid-mediated fat metabolism and endocrine functions (Begley et al., Reference Begley, Hill and Gahan2006; Patel et al., Reference Patel, Singhania, Pandey and Chincholkar2010; Jones et al., Reference Jones, Martoni, Ganopolsky, Labbé and Prakash2014; Joyce et al., Reference Joyce, Shanahan, Hill and Gahan2014b). Recent probiotics studies have already shown that oral administration of BSH-producing lactobacilli could affect lipid metabolism, consequently reducing body weight and/or cholesterol level in human beings (Jones et al., Reference Jones, Tomaro-Duchesneau, Martoni and Prakash2013), rats (Pato et al., Reference Pato, Surono and Hosono2004; Kumar et al., Reference Kumar, Grover and Batish2011), mice (Park et al., Reference Park, Ahn, Park, Huh, Yoo, Yu, Sung, McGregor and Choi2013, Reference Park, Oh and Cha2014; Miyoshi et al., Reference Miyoshi, Ogawa, Higurashi and Kadooka2014), and pigs (De Smet et al., Reference De Smet, De Boever and Verstraete1998).

Molecular and cellular studies also provided new insights into underlying mechanisms of the effect of BSH enzyme on host lipid metabolism and energy harvest. Clearly, unconjugated bile acids, directly resulting from BSH activity, are less effective than conjugated bile acids in the emulsification of dietary fat and consequently affect lipid absorption and metabolism. However, unconjugated bile acids could exert more profound impacts on host energy harvest both locally and systemically. Farnesoid X receptor (FXR), which is preferentially stimulated by unconjugated bile acids, not only regulate lipogenesis and triglyceride synthesis (Watanabe et al., Reference Watanabe, Houten, Wang, Moschetta, Mangelsdorf, Heyman, Moore and Auwerx2004; Li et al., Reference Li, Jiang, Krausz, Li, Albert, Hao, Fabre, Mitchell, Patterson and Gonzalez2013), but also regulate glucose homeostasis by increasing glycogen synthesis (Zhang et al., Reference Zhang, Lee, Barrera, Lee, Vales, Gonzalez, Willson and Edwards2006; Caron et al., Reference Caron, Samanez, Dehondt, Ploton, Briand, Lien, Dorchies, Dumont, Postic and Cariou2013) or decreasing glycolysis (Caron et al., Reference Caron, Samanez, Dehondt, Ploton, Briand, Lien, Dorchies, Dumont, Postic and Cariou2013). Using a pig model, Pereira-Fantini et al. (Reference Pereira-Fantini, Lapthorne, Joyce, Dellios, Wilson, Fouhy, Thomas, Scurr, Hill and Gahan2014) examined the impact of BSH-mediated bile acid dysmetabolism on FXR signaling pathways and clinical outcomes and showed that alterations in bile acid composition may have contributed to the observed disturbance in FXR-mediated signaling pathways (Pereira-Fantini et al., Reference Pereira-Fantini, Lapthorne, Joyce, Dellios, Wilson, Fouhy, Thomas, Scurr, Hill and Gahan2014).

Notably, obesity development is a complex physiological issue. The BSH-mediated bile salt metabolism is only one of several potential mechanisms by which the microbiota affect host energy harvest and weight gain (Walker and Parkhill, Reference Walker and Parkhill2013). The studies described above only provide indirect evidence supporting the role of BSH-producing probiotics or BSH-mediated bile metabolism in host lipid metabolism and energy harvest. Direct and controlled approaches are required in order to obtain complete understanding of BSH-mediated regulation of host weight gain and lipid metabolism.

Recently, using a controlled system in conjunction with a mouse model, Joyce et al. (Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a) obtained the first direct evidence demonstrating that manipulation of in situ BSH activity alone significantly influenced lipid metabolism, signaling functions, and weight gain (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a). Briefly, two well characterized L. salivarius BSH enzymes were cloned into an E. coli host strain (MG1655). The recombinant Escherichia coli constructs could effectively colonize the gastrointestinal tract of mice with expression of high level of BSH activity. Colonization of germ-free mice with such BSH-producing E. coli strain elevated intestinal BSH activity and resulted in local bile acids deconjugation with concomitant reduced levels in body weight and cholesterol, alternations in lipid metabolism, signaling functions, local and systemic transcriptome profiles in the pathways governing lipid metabolism (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a). Notably, in conventionally raised mice, enhanced in situ BSH activity also caused local bile acid deconjugation, reduced mouse weight gain, lowered serum cholesterol level, and reduced liver triglyceride level, which further demonstrates that BSH is a key mechanism through which the microbiota modulates host lipid metabolism and dietary energy harvest (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a). In addition to its ability to alter local (gastrointestinal) functions, BSH activity could systemically affect host physiology such that the BSH activity-mediated bile acids can interact with transporters (e.g. Abcg5/8) and regulators (e.g. FXR regulon, Fiaf) that lead to change in body mass (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a).

Other physiological process

The BSH-mediated unconjugated bile acids also affect immune homeostasis because of their ability to modulate a panel of effectors in the intestine, such as inducible nitric oxide synthase (iNOS) (Inagaki et al., Reference Inagaki, Moschetta, Lee, Peng, Zhao, Downes, Ruth, Shelton, Richardson and Repa2006), the antimicrobial peptide RegIIIγ produced by intestinal paneth cells (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a), and dendritic cell differentiation (Ichikawa et al., Reference Ichikawa, Takayama, Yoneno, Kamada, Kitazume, Higuchi, Matsuoka, Watanabe, Itoh and Kanai2012; Joyce et al., Reference Joyce, Shanahan, Hill and Gahan2014b). In addition to the pathway via intestinal FXR, unconjugated bile acids also affect TGR5-mediated adipose tissue development and weight loss (Watanabe et al., Reference Watanabe, Houten, Mataki, Christoffolete, Kim, Sato, Messaddeq, Harney, Ezaki and Kodama2006; Svensson et al., Reference Svensson, Olsson, Andersson-Assarsson, Taube, Pereira, Froguel and Jacobson2013). Interestingly, Joyce et al. (Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a) also observed that enhanced in situ BSH activity reversed the expression pattern of genes responsible for regulating circadian rhythm (e.g., Dbp) and other genes central to circadian clock (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a). Finally, unconjugated bile acids can also alter intestinal microbiota, consequently may exert more complex impacts on host (Inagaki et al., Reference Inagaki, Moschetta, Lee, Peng, Zhao, Downes, Ruth, Shelton, Richardson and Repa2006; Islam et al., Reference Islam, Fukiya, Hagio, Fujii, Ishizuka, Ooka, Ogura, Hayashi and Yokota2011).

Potential adverse effects due to high-level BSH activity in the intestine

High-level BSH activity would result in a large proportion of unconjugated bile acids, which can lead to malabsorption of lipid and may cause steatorrhea in the host (Kim and Lee, Reference Kim and Lee2005). Recent research also indicated that deconjugation of bile salts by BSH-producing lactobacilli is an important factor leading to short bowel syndrome due to abnormal lipid metabolism and a disrupted bile acid profile (Bongaerts et al., Reference Bongaerts, Severijnen, Tangerman, Verrips and Tolboom2000; Choi et al., Reference Choi, Lew, Yeo, Nair Parvathy and Liong2014).

BSH-mediated deconjugation of bile salts can increase bile recovery from passive absorption across the colonic epithelium by making bile salts more hydrophobic, which may also cause some adverse effects. For example, a high concentration of secondary bile acids in blood and feces, that are produced by a multistep of 7α-dehydroxylation reaction from unconjugated bile acids, are proposed to be related to the pathogenesis of cholesterol gallstone diseases as well as colon cancer (van Faassen et al., Reference Van Faassen, Bol, van Dokkum, Pikaar, Ockhuizen and Hermus1987; Färkkilä and Miettinen, Reference Färkkilä and Miettinen1990; Marteau and Rambaud, Reference Marteau and Rambaud1993; McGarr et al., Reference McGarr, Ridlon and Hylemon2005; Venneman and van Erpecum, Reference Venneman and van Erpecum2010; Ou et al., Reference Ou, Carbonero, Zoetendal, DeLany, Wang, Newton, Gaskins and O'Keefe2013). Secondary bile acids may increase the risk of cancer by increasing oxidative stress and associated DNA damage (Cooke et al., Reference Cooke, Evans, Dizdaroglu and Lunec2003; Bernstein et al., Reference Bernstein, Bernstein, Payne, Dvorakova and Garewal2005). The sulfonic acid moiety in unconjugated bile acids could be reduced and dissimilated to hydrogen sulfide, which is highly toxic and can increase colon cell turnover (Christl et al., Reference Christl, Eisner, Dusel, Kasper and Scheppach1996; Corzo and Gilliland, Reference Corzo and Gilliland1999; Lie et al., Reference Lie, Clawson, Godchaux and Leadbetter1999; Laue et al., Reference Laue, Friedrich, Ruff and Cook2001; Ridlon et al., Reference Ridlon, Kang and Hylemon2006). Hydrogen sulfide is a potent inhibitor of colonic butyrate metabolism, which is a key nutrient and regulator of cell turnover (Christl et al., Reference Christl, Eisner, Dusel, Kasper and Scheppach1996; Van Eldere et al., Reference Van Eldere, Celis, De Pauw, Lesaffre and Eyssen1996). Hydrogen sulfide can also reduce apoptosis in colon cancer cells by preventing the function of a chemo-preventative agent β-phenylethyl isothiocyanate (PEITC) (Rose et al., Reference Rose, Moore, Ming, Nam, Armstrong and Whiteman2005).

Target BSH for enhanced animal production and health

In contrast to the significant progress on BSH research for human health described above, little information exists concerning BSH and BSH-producing bacteria in food animals. Some early studies evaluated direct usage of bile salts as a feed additive to improve feed efficiency due to the well-recognized role of bile salts in fat digestion (Kussaibati et al., Reference Kussaibati, Guillaume and Leclercq1982; Reinhart et al., Reference Reinhart, Mahan and Cera1988). In chickens, supplementation of bile salts in the diet increased the absorption of fatty acids, but had no influence on chickens with fat-free diet (Kussaibati et al., Reference Kussaibati, Guillaume and Leclercq1982). Presence of bile salts in the diet also increased fat digestibility in swine after the weaning period (Reinhart et al., Reference Reinhart, Mahan and Cera1988). Although the findings from these studies are encouraging, bile salts have not been adopted by the feed industry as feed additives to improve growth performance of food animals, likely due to the issues of cost, availability, and complex biotransformation of bile salts in the gastrointestinal tract.

AGPs are defined as a group of antibiotics used in feed at sub-therapeutic level to improve average daily weight gain and feed efficiency in food animals. This husbandry technique has been practiced since the 1950s. However, use of AGPs has been associated with the emergence of antibiotic-resistant human pathogens of animal origins. Therefore, ending the use of AGPs is a worldwide trend to protect public health. Effective alternatives to AGPs are urgently needed to maintain current animal production levels without threatening public health. Recent animal studies on the effect of AGP usage on intestinal microbiome indicate that the enhanced feed efficiency and body weight gain in food animals due to AGP usage is inversely related to the BSH activity as well as the abundance of potent BSH-producers in the intestine (Lin, Reference Lin2014).

As early as in 1980s, Feighner and Dashkevicz (Reference Feighner and Dashkevicz1987) reported that use of AGP reduced intestinal BSH activity in poultry and they proposed that inhibition of BSH activity would promote feed efficiency and weight gain in food animals. In this early study, a radiochemical method was successfully developed to directly determine BSH activity in intestinal contents; however, the method used in this study was technically challenging and time consuming (Feighner and Dashkevicz, Reference Feighner and Dashkevicz1987). Notably, the standard BSH activity assay widely used is not feasible for examining fecal BSH activity because of the high levels of background caused by free amino acids in intestinal contents. To date, fecal bile acid profile is an acceptable indicator for evaluating BSH activity in the intestinal contents. Consistent with the finding by Feighner and Dashkevicz (Reference Feighner and Dashkevicz1987), Knarreborg et al. (Reference Knarreborg, Lauridsen, Engberg and Jensen2004) also observed AGP usage reduced concentration of unconjugated bile salts in the intestine of broilers by using reversed-phase HLPC method, which led to an enhanced bioavailability of α-tocopheryl acetate. In multiple pen trials, Guban et al. (Reference Guban, Korver, Allison and Tannock2006) further confirmed that AGP treatment improved weight gain and fat digestibility in broilers, decreased population levels of L. salivarius, and significantly reduced BSH activity in the intestine, which was reflected by a decreased pool of deconjugated bile salts in ileal contents using a HPLC method. In pigs, De Smet et al. (Reference De Smet, De Boever and Verstraete1998) observed that oral administration of the L. reuteri with BSH activity influenced host lipid metabolism and decreased total and LDL-cholesterol concentrations. Du Toit et al. (Reference Du Toit, Franz, Dicks, Schillinger, Haberer, Warlies, Ahrens and Holzapfel1998) also had a similar finding in a minipig feeding trial using BSH-positive probiotic mix. However, both of these pig studies (De Smet et al., Reference De Smet, De Boever and Verstraete1998; Du Toit et al., Reference Du Toit, Franz, Dicks, Schillinger, Haberer, Warlies, Ahrens and Holzapfel1998) lack determination of intestinal BSH activity, which is needed to rule out potential pleotropic effects resulting from the treatment with BSH-producing probiotics.

Regarding response of intestinal microbiota to AGPs, a key issue for us to understand the mode of action of AGP, culture-independent molecular approaches have been used to examine the effect of AGPs on intestinal microbiota in poultry and swine; to date, more than ten papers have been published in this field (Lin, Reference Lin2014). Not surprisingly, long-term supplementation of diet with AGPs significantly affected the microbial ecology in the intestine in all reported studies. However, the specific bacteria or environmental niche changes that are meaningful and are linked to the desired phenotype of enhanced growth performance need to be clarified. In-depth comparative analysis of these animal microbiome studies led to an interesting finding: in most chicken and swine studies, use of AGP reduced the population of Lactobacillus species, the major BSH-producing bacteria in the animal intestine (Begley et al., Reference Begley, Hill and Gahan2006; Lin, Reference Lin2014). The independent findings from these food animal studies, together with those from human BSH research summarized above, are like jigsaw pieces which seem to be scattered but are in fact tightly interrelated. Therefore, it was proposed that BSH is a key mechanistic microbiome target for developing novel alternatives to AGPs and this hypothesis prompted us to identify and characterize a potent BSH enzyme from a chicken L. salivarius probiotic strain (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012). Interestingly, copper and zinc compounds displayed a potent inhibitory effect on BSH enzyme activity in this study, which not only provides scientific evidence to understand the mode of action of high dietary concentrations of copper/zinc for growth promotion, but also strongly supports our hypothesis that BSH inhibitors may serve as promising alternatives to AGPs (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012). Subsequently, by taking advantage of the unique feature of the L. salivarius BSH enzyme (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012), an efficient high-throughput screening system was successfully developed and used to discover BSH inhibitors (Smith et al., Reference Smith, Zeng and Lin2014). Unlike many BSH enzymes from other bacteria that have narrow substrate spectrum, the L. salivarius BSH displayed a potent hydrolysis activity towards both glycol-conjugated and taurine-conjugated bile salts. The broad substrates specificity nature of this BSH makes it an ideal candidate for screening desired BSH inhibitors. This hypothesis is further tested by our recent study showing the identified BSH inhibitors also exhibited potent inhibitory effects on a phylogenetically distant BSH from L. acidophilus (Lin et al., Reference Lin, Negga, Zeng and Smith2014).

Despite the recognized AMR issues associated with antibiotic usage in food animals, animal industries still heavily rely on antibiotics due to the lack of practical and consistent antibiotic alternative approaches. Solely limiting antibiotics without providing effective alternatives would compromise animal production and health. BSH inhibitors are promising alternatives to AGPs for enhanced feed efficiency and growth performance. Successful development of effective non-antibiotic BSH inhibitor feed additives could reduce the dependence on in-feed antibiotics for growth promotion, consequently mitigating AMR pressure in agriculture ecosystems, a significant and timely issue impacting animal health and food safety.

Other types of antibiotic-alternative products, such as probiotics, prebiotics, and organic acids, have drawn wide attention and have been developed and used to alter intestinal microbiota for improving animal health and production (Dibner and Richards, Reference Dibner and Richards2005; Lin, Reference Lin2014). However, very limited data are available to scientifically justify the choice of specific bacterial species or products for growth promotion and results are inconsistent from independent studies (Dibner and Richards, Reference Dibner and Richards2005). For example, although probiotics containing Lactobacillus are well recognized for their beneficial effects on boosting host immunity, these probiotics could have a negative impact on host lipid metabolism due to BSH production. Specifically, in a large pen trial, Sharifi et al. (Reference Sharifi, Dibamehr, Lotfollahian and Baurhoo2012) observed that supplementation of a 7-bacterial species probiotic (Protexin) to fat-rich diets significantly reduced body weight gain, fat digestibility, and feed conversion in broilers. Moreover, using a different 5-bacterial species competitive exclusion probiotic product, Mountzouris et al. (Reference Mountzouris, Tsitrsikos, Palamidi, Arvaniti, Mohnl, Schatzmayr and Fegeros2010) also observed similar inferior feed conversion efficiency and reduced fat digestibility in response to probiotic treatment in broilers. These investigators have proposed that the enrichment of the intestinal microflora, particularly lactobacilli, due to probiotic supplementation caused enhanced BSH activity in the intestine, leading to detrimental effects on lipid metabolism and growth performance of broilers. Therefore, improved knowledge in the role of BSH and BSH-producing bacteria will help design rationally tailored probiotics that will enhance animal health and performance. For example, the BSH inhibitors could also be used together with certain BSH-producing probiotics to maximize the beneficial effect of the probiotics by mitigating their potential negative impact on host fat digestion. This approach may further help animal production industries optimize existing probiotic and prebiotic additives for enhanced feed efficiency, growth performance and profitability.

Conclusions and research gaps

Antibiotics have been heavily used for animal farming to maintain animal production and health. However, farm use of antibiotics is a driving force to enrich AMR genes (called the ‘resistome’) in various niches and to promote pools of resistant pathogenic bacteria, raising food safety and public health concerns (Davies, Reference Davies, Atlas and Maloy2014; Perry et al., Reference Perry, Westman and Wright2014). To effectively mitigate AMR in agricultural systems, a reduction in the use of antibiotics in farming is imperative. Thus, intensive efforts are critically needed to develop effective non-antibiotic growth promotion strategies that can be practically implemented by animal producers. Recent microbiome studies have provided compelling evidence that BSH is a key mechanistic microbiome target for developing novel alternatives to AGPs. Development of BSH inhibitor-based non-antibiotic feed additives directly addresses the nutrition concern (feed efficiency/growth rate) that prevents animal industries from reducing antibiotic usage. In addition to benefitting healthy animals under routine management, the weight-enhancing BSH inhibitors may also help sick animals better harvest dietary energy while combatting infectious diseases or environmental/production stress.

Despite the significant role of bacterial BSH activity in host lipid metabolism and energy harvest, research on BSH is still in its infancy. In particular, little effort has been placed on characterization of BSH enzymes and/or BSH-producing bacteria in food animals. Several significant gaps remain in knowledge associated with BSH in food animal production and health. Filling these gaps will not only directly benefit animal health but also provide insights and likely new model systems for human health research, leading to novel ‘One Health’ measures for enhanced animal production, food safety, and human nutrition.

  • Ecology of BSH enzymes and BSH-producing bacteria in the intestine. To date, only a limited number of BSH enzymes have been identified in the intestinal bacteria isolated from food animals (Table 1). With the aid of next generation sequencing technologies and bioinformatics tools, functional and comparative metagenomic analyses of intestinal BSH in food animals are warranted and will provide a better picture of the diversity and function of BSH in the intestine. Information in conjunction with other phenotypic examinations would improve our understanding on the role of BSH in the symbiotic relationship between the gastrointestinal microbiome and animal host. Given that specific BSH enzyme(s) and corresponding BSH-producing bacteria may serve as biomarkers for health statuses of animal hosts, understanding the ecology of BSH enzymes and BSH-producing bacteria in the intestine would facilitate the development of diagnostics to evaluate the health status of animals and people.

  • Comprehensive evaluation using a controlled system together with a new model system is still critically needed to provide new mechanistic information for the role of BSH in host energy harvest and weight gain. Given the increasing awareness of important roles of microbiota in intestine health, development of specifically tailored probiotics is a logical strategy for practical application, but this approach needs an in-depth understanding of the molecular, physiological, and ecological features of probiotic organisms in order to select and design probiotics for safe, effective administration for specific purposes. To date, there are not any studies using BSH-negative and BSH-overproducing probiotic organisms to definitively link BSH activity to the specific phenotype and their impacts on host animals and native microbiomes. This is likely due to the challenge for manipulating BSH activity in commensal organisms for specific laboratory animal hosts and to the lack of public acceptance of using genetically modified organisms (GMOs) in human trials. While this concern has been partly addressed with a recent E. coli knock-in model (Joyce et al., Reference Joyce, MacSharry, Casey, Kinsella, Murphy, Shanahan, Hill and Gahan2014a), manipulating BSH activity of a natural intestinal commensal organism in an animal model would be a better approach. Recent characterization of L. salivarius as a potent BSH producer (Wang et al., Reference Wang, Zeng, Mo, Smith, Guo and Lin2012) provides an excellent opportunity to address this issue using a food animal model system, because genetic tools to manipulate L. salivarius have been well established. Such research efforts would enable us to better manage body weight by manipulating microbiota in people and animals.

  • Developing alternatives to AGPs by inhibiting BSH activity in the intestine. In addition to discovering more novel BSH inhibitors, comprehensive animal trials are essential to further evaluate and select desired BSH inhibitors. It is likely that prolonged use of a particular BSH inhibitor could lead to negative physiological consequences due to pleotropic effects of specific inhibitor and complexity of host physiology. For example, because BSH inhibitors are expected to improve lipid metabolism, it is important to examine if energy harvest and weight gain is partitioned adequately and not skewed toward excess fat deposition, which would be undesirable for both animal producers and consumers. In addition, it is also warranted to examine how inhibition of BSH activity affects the bile profile, as well as the gastrointestinal microbial community and all the implications that these changes hold for animal health and productivity.

  • Structural basis of BSH function. Given ecological diversity of BSH in the intestinal microbiome, structure analyses of BSH enzymes from various species are highly warranted, which would reveal critical residues in catalysis and provide key information on the substrates selectivity of BSH enzymes. Clearly, such basic studies also will directly facilitate future translational research, such as using molecular docking to develop desired BSH inhibitors for growth promotion in food animals.

Acknowledgment

Work in our laboratory was supported by a University of Tennessee AgResearch Innovation Grant and a University of Tennessee Research Foundation Technology Maturation Fund.

References

Appleby, RN and Walters, JR (2014). The role of bile acids in functional GI disorders. Neurogastroenterology & Motility 26: 10571069.CrossRefGoogle ScholarPubMed
Aries, V and Hill, M (1970). Degradation of steroids by intestinal bacteria I. Deconjugation of bile salts. Biochimica et Biophysica Acta (BBA)-Lipids and Lipid Metabolism 202: 526534.CrossRefGoogle ScholarPubMed
Batta, A, Salen, G, Arora, R, Shefer, S, Batta, M and Person, A (1990). Side chain conjugation prevents bacterial 7-dehydroxylation of bile acids. Journal of Biological Chemistry 265: 1092510928.CrossRefGoogle ScholarPubMed
Begley, M, Gahan, CG and Hill, C (2005). The interaction between bacteria and bile. FEMS Microbiology Reviews 29: 625651.CrossRefGoogle ScholarPubMed
Begley, M, Hill, C and Gahan, CG (2006). Bile salt hydrolase activity in probiotics. Applied and Environmental Microbiology 72: 17291738.CrossRefGoogle ScholarPubMed
Bernstein, H, Bernstein, C, Payne, C, Dvorakova, K and Garewal, H (2005). Bile acids as carcinogens in human gastrointestinal cancers. Mutation Research/Reviews in Mutation Research 589: 4765.CrossRefGoogle ScholarPubMed
Bi, J, Fang, F, Lu, S, Du, G and Chen, J (2013). New insight into the catalytic properties of bile salt hydrolase. Journal of Molecular Catalysis B: Enzymatic 96: 4651.CrossRefGoogle Scholar
Bongaerts, GP, Severijnen, RS, Tangerman, A, Verrips, A and Tolboom, JJ (2000). Bile acid deconjugation by Lactobacilli and its effects in patients with a short small bowel. Journal of Gastroenterology 35: 801804.CrossRefGoogle ScholarPubMed
Camilleri, M and Gores, GJ (2015). Therapeutic targeting of bile acids. American Journal of Physiology-Gastrointestinal and Liver Physiology 309: G209G215.CrossRefGoogle ScholarPubMed
Caron, S, Samanez, CH, Dehondt, H, Ploton, M, Briand, O, Lien, F, Dorchies, E, Dumont, J, Postic, C and Cariou, B (2013). Farnesoid X receptor inhibits the transcriptional activity of carbohydrate response element binding protein in human hepatocytes. Molecular and Cellular Biology 33: 22022211.CrossRefGoogle ScholarPubMed
Chae, J, Valeriano, V, Kim, GB and Kang, DK (2013). Molecular cloning, characterization and comparison of bile salt hydrolases from Lactobacillus johnsonii PF01. Journal of Applied Microbiology 114: 121133.CrossRefGoogle ScholarPubMed
Choi, S-B, Lew, L-C, Yeo, S-K, Nair Parvathy, S and Liong, M-T (2014). Probiotics and the BSH-related cholesterol lowering mechanism: a Jekyll and Hyde scenario. Critical Reviews in Biotechnology 35: 392401.CrossRefGoogle Scholar
Christl, SU, Eisner, H-D, Dusel, G, Kasper, H and Scheppach, W (1996). Antagonistic effects of sulfide and butyrate on proliferation of colonic mucosa. Digestive Diseases and Sciences 41: 24772481.CrossRefGoogle ScholarPubMed
Claesson, MJ, Li, Y, Leahy, S, Canchaya, C, van Pijkeren, JP, Cerdeño-Tárraga, AM, Parkhill, J, Flynn, S, O'Sullivan, GC and Collins, JK (2006). Multireplicon genome architecture of Lactobacillus salivarius . Proceedings of the National Academy of Sciences of the United States of America 103: 67186723.CrossRefGoogle ScholarPubMed
Coleman, JP and Hudson, LL (1995). Cloning and characterization of a conjugated bile acid hydrolase gene from Clostridium perfringens . Applied and Environmental Microbiology 61: 25142520.CrossRefGoogle ScholarPubMed
Cooke, MS, Evans, MD, Dizdaroglu, M and Lunec, J (2003). Oxidative DNA damage: mechanisms, mutation, and disease. FASEB Journal 17: 11951214.CrossRefGoogle ScholarPubMed
Corzo, G and Gilliland, S (1999). Bile salt hydrolase activity of three strains of Lactobacillus acidophilus . Journal of Dairy Science 82: 472480.CrossRefGoogle ScholarPubMed
Damodharan, K, Lee, YS, Palaniyandi, SA, Yang, SH and Suh, J-W (2015). Preliminary probiotic and technological characterization of Pediococcus pentosaceus strain KID7 and in vivo assessment of its cholesterol-lowering activity. Frontiers in Microbiology 6: 768. doi: 10.3389/fmicb.2015.00768.CrossRefGoogle ScholarPubMed
Davies, J (2014). Antibiotic resistance in and from nature. In: Atlas, RM and Maloy, S (eds) One Health: People, Animals, and the Environment. Washington, DC: American Society for Microbiology, pp. 185194.Google Scholar
De Aguiar Vallim, TQ, Tarling, EJ and Edwards, PA (2013). Pleiotropic roles of bile acids in metabolism. Cell Metabolism 17: 657669.CrossRefGoogle ScholarPubMed
Dean, M, Cervellati, C, Casanova, E, Squerzanti, M, Lanzara, V, Medici, A, de Laureto, PP and Bergamini, CM (2002). Characterization of cholylglycine hydrolase from a bile-adapted strain of Xanthomonas maltophilia and its application for quantitative hydrolysis of conjugated bile salts. Applied and Environmental Microbiology 68: 31263128.CrossRefGoogle ScholarPubMed
De Smet, I, De Boever, P and Verstraete, W (1998). Cholesterol lowering in pigs through enhanced bacterial bile salt hydrolase activity. British Journal of Nutrition 79: 185194.CrossRefGoogle ScholarPubMed
Dibner, J and Richards, J (2005). Antibiotic growth promoters in agriculture: history and mode of action. Poultry Science 84: 634643.CrossRefGoogle ScholarPubMed
Dong, Z, Zhang, J, Lee, B, Li, H, Du, G and Chen, J (2012). A bile salt hydrolase gene of Lactobacillus plantarum BBE7 with high cholesterol-removing activity. European Food Research and Technology 235: 419427.CrossRefGoogle Scholar
Dussurget, O, Cabanes, D, Dehoux, P, Lecuit, M, Buchrieser, C, Glaser, P and Cossart, P (2002). Listeria monocytogenes bile salt hydrolase is a PrfA-regulated virulence factor involved in the intestinal and hepatic phases of listeriosis. Molecular Microbiology 45: 10951106.CrossRefGoogle ScholarPubMed
Du Toit, M, Franz, C, Dicks, L, Schillinger, U, Haberer, P, Warlies, B, Ahrens, F and Holzapfel, W (1998). Characterisation and selection of probiotic lactobacilli for a preliminary minipig feeding trial and their effect on serum cholesterol levels, faeces pH and faeces moisture content. International Journal of Food Microbiology 40: 93104.CrossRefGoogle ScholarPubMed
Evans, MJ, Mahaney, PE, Borges-Marcucci, L, Lai, K, Wang, S, Krueger, JA, Gardell, SJ, Huard, C, Martinez, R and Vlasuk, GP (2009). A synthetic farnesoid X receptor (FXR) agonist promotes cholesterol lowering in models of dyslipidemia. American Journal of Physiology-Gastrointestinal and Liver Physiology 296: G543G552.CrossRefGoogle ScholarPubMed
Fang, F, Li, Y, Bumann, M, Raftis, EJ, Casey, PG, Cooney, JC, Walsh, MA and O'Toole, PW (2009). Allelic variation of bile salt hydrolase genes in Lactobacillus salivarius does not determine bile resistance levels. Journal of Bacteriology 191: 57435757.CrossRefGoogle Scholar
Färkkilä, M and Miettinen, TA (1990). Lipid metabolism in bile acid malabsorption. Annals of Medicine 22: 513.CrossRefGoogle ScholarPubMed
Feighner, SD and Dashkevicz, MP (1987). Subtherapeutic levels of antibiotics in poultry feeds and their effects on weight gain, feed efficiency, and bacterial cholyltaurine hydrolase activity. Applied and Environmental Microbiology 53: 331336.CrossRefGoogle ScholarPubMed
Feighner, SD and Dashkevicz, MP (1988). Effect of dietary carbohydrates on bacterial cholyltaurine hydrolase in poultry intestinal homogenates. Applied and Environmental Microbiology 54: 337342.CrossRefGoogle ScholarPubMed
Gopal-Srivastava, R and Hylemon, PB (1988). Purification and characterization of bile salt hydrolase from Clostridium perfringens . Journal of Lipid Research 29: 10791085.CrossRefGoogle ScholarPubMed
Grill, J, Schneider, F, Crociani, J and Ballongue, J (1995). Purification and characterization of conjugated bile salt hydrolase from Bifidobacterium longum BB536. Applied and Environmental Microbiology 61: 25772582.CrossRefGoogle ScholarPubMed
Gu, X-C, Luo, X-G, Wang, C-X, Ma, D-Y, Wang, Y, He, Y-Y, Li, W, Zhou, H and Zhang, T-C (2014). Cloning and analysis of bile salt hydrolase genes from Lactobacillus plantarum CGMCC No. 8198. Biotechnology Letters 36: 975983.CrossRefGoogle ScholarPubMed
Guban, J, Korver, D, Allison, G and Tannock, G (2006). Relationship of dietary antimicrobial drug administration with broiler performance, decreased population levels of Lactobacillus salivarius, and reduced bile salt deconjugation in the ileum of broiler chickens. Poultry Science 85: 21862194.CrossRefGoogle ScholarPubMed
Gupta, S, Stravitz, RT, Dent, P and Hylemon, PB (2001). Down-regulation of cholesterol 7α-hydroxylase (CYP7A1) gene expression by bile acids in primary rat hepatocytes is mediated by the c-Jun N-terminal kinase pathway. Journal of Biological Chemistry 276: 1581615822.CrossRefGoogle ScholarPubMed
Hofmann, AF and Mysels, KJ (1992). Bile acid solubility and precipitation in vitro and in vivo: the role of conjugation, pH, and Ca2+ ions. Journal of Lipid Research 33: 617626.CrossRefGoogle ScholarPubMed
Houten, SM, Watanabe, M and Auwerx, J (2006). Endocrine functions of bile acids. EMBO Journal 25: 14191425.CrossRefGoogle ScholarPubMed
Huang, Y, Wang, X, Wang, J, Wu, F, Sui, Y, Yang, L and Wang, Z (2013). Lactobacillus plantarum strains as potential probiotic cultures with cholesterol-lowering activity. Journal of Dairy Science 96: 27462753.CrossRefGoogle ScholarPubMed
Ichikawa, R, Takayama, T, Yoneno, K, Kamada, N, Kitazume, MT, Higuchi, H, Matsuoka, K, Watanabe, M, Itoh, H and Kanai, T (2012). Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway. Immunology 136: 153162.CrossRefGoogle Scholar
Inagaki, T, Moschetta, A, Lee, Y-K, Peng, L, Zhao, G, Downes, M, Ruth, TY, Shelton, JM, Richardson, JA and Repa, JJ (2006). Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proceedings of the National Academy of Sciences of the United States of America 103: 39203925.CrossRefGoogle ScholarPubMed
Islam, KS, Fukiya, S, Hagio, M, Fujii, N, Ishizuka, S, Ooka, T, Ogura, Y, Hayashi, T and Yokota, A (2011). Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 141: 17731781.CrossRefGoogle ScholarPubMed
Jiang, J, Hang, X, Zhang, M, Liu, X, Li, D and Yang, H (2010). Diversity of bile salt hydrolase activities in different lactobacilli toward human bile salts. Annals of Microbiology 60: 8188.CrossRefGoogle Scholar
Johnson, L (1998). Bile secretion and gallbladder function. Essential Medical Physiology 2: 465471.Google Scholar
Jones, BV, Begley, M, Hill, C, Gahan, CG and Marchesi, JR (2008). Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proceedings of the National Academy of Sciences of the United States of America 105: 1358013585.CrossRefGoogle ScholarPubMed
Jones, ML, Tomaro-Duchesneau, C, Martoni, CJ and Prakash, S (2013). Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications. Expert Opinion on Biological Therapy 13: 631642.CrossRefGoogle ScholarPubMed
Jones, ML, Martoni, CJ, Ganopolsky, JG, Labbé, A and Prakash, S (2014). The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opinion on Biological Therapy 14: 467482.CrossRefGoogle ScholarPubMed
Joyce, SA, MacSharry, J, Casey, PG, Kinsella, M, Murphy, EF, Shanahan, F, Hill, C and Gahan, CG (2014a). Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proceedings of the National Academy of Sciences of the United States of America 111: 74217426.CrossRefGoogle ScholarPubMed
Joyce, SA, Shanahan, F, Hill, C and Gahan, CG (2014b). Bacterial bile salt hydrolase in host metabolism: potential for influencing gastrointestinal microbe-host crosstalk. Gut Microbes 5: 669674.CrossRefGoogle ScholarPubMed
Kahn, SE, Hull, RL and Utzschneider, KM (2006). Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444: 840846.CrossRefGoogle ScholarPubMed
Kawamoto, K, Horibe, I and Uchida, K (1989). Purification and characterization of a new hydrolase for conjugated bile acids, chenodeoxycholyltaurine hydrolase, from Bacteroides vulgatus . Journal of Biochemistry 106: 10491053.CrossRefGoogle ScholarPubMed
Kim, G and Lee, BH (2005) Biochemical and molecular insights into bile salt hydrolase in the gastrointestinal microflora-a review. Asian Australasian Journal of Animal Sciences 18: 1505.CrossRefGoogle Scholar
Kim, G-B, Miyamoto, CM, Meighen, EA and Lee, BH (2004). Cloning and characterization of the bile salt hydrolase genes (bsh) from Bifidobacterium bifidum strains. Applied and Environmental Microbiology 70: 56035612.CrossRefGoogle ScholarPubMed
Kim, G-B, Brochet, M and Lee, BH (2005). Cloning and characterization of a bile salt hydrolase (bsh) from Bifidobacterium adolescentis . Biotechnology Letters 27: 817822.CrossRefGoogle ScholarPubMed
Knarreborg, A, Lauridsen, C, Engberg, RM and Jensen, SK (2004). Dietary antibiotic growth promoters enhance the bioavailability of α-tocopheryl acetate in broilers by altering lipid absorption. Journal of Nutrition 134: 14871492.CrossRefGoogle ScholarPubMed
Kumar, R, Grover, S and Batish, VK (2011). Hypocholesterolaemic effect of dietary inclusion of two putative probiotic bile salt hydrolase-producing Lactobacillus plantarum strains in Sprague–Dawley rats. British Journal of Nutrition 105: 561573.CrossRefGoogle ScholarPubMed
Kumar, RS, Brannigan, JA, Prabhune, AA, Pundle, AV, Dodson, GG, Dodson, EJ and Suresh, C (2006). Structural and functional analysis of a conjugated bile salt hydrolase from Bifidobacterium longum reveals an evolutionary relationship with penicillin V acylase. Journal of Biological Chemistry 281: 3251632525.CrossRefGoogle ScholarPubMed
Kussaibati, R, Guillaume, J and Leclercq, B (1982). The effects of endogenous energy, type of diet, and addition of bile salts on true metabolizable energy values in young chicks. Poultry Science 61: 22182223.CrossRefGoogle ScholarPubMed
Lambert, JM, Bongers, RS, de Vos, WM and Kleerebezem, M (2008). Functional analysis of four bile salt hydrolase and penicillin acylase family members in Lactobacillus plantarum WCFS1. Applied and Environmental Microbiology 74: 47194726.CrossRefGoogle ScholarPubMed
Laue, H, Friedrich, M, Ruff, J and Cook, AM (2001). Dissimilatory sulfite reductase (desulfoviridin) of the taurine-degrading, non-sulfate-reducing bacterium Bilophila wadsworthia RZATAU contains a fused DsrB-DsrD subunit. Journal of Bacteriology 183: 17271733.CrossRefGoogle ScholarPubMed
Lepercq, P, Relano, P, Cayuela, C and Juste, C (2004). Bifidobacterium animalis strain DN-173 010 hydrolyses bile salts in the gastrointestinal tract of pigs. Scandinavian Journal of Gastroenterology 39: 12661271.CrossRefGoogle ScholarPubMed
Li, F, Jiang, C, Krausz, KW, Li, Y, Albert, I, Hao, H, Fabre, KM, Mitchell, JB, Patterson, AD and Gonzalez, FJ (2013). Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nature Communications 4: 2384. doi: 10.1038/ncomms3384.CrossRefGoogle ScholarPubMed
Li, Y, Raftis, E, Canchaya, C, Fitzgerald, GF, van Sinderen, D and O'Toole, PW (2006). Polyphasic analysis indicates that Lactobacillus salivarius subsp. salivarius and Lactobacillus salivarius subsp. salicinius do not merit separate subspecies status. International Journal of Systematic and Evolutionary Microbiology 56: 23972403.CrossRefGoogle Scholar
Lie, TJ, Clawson, ML, Godchaux, W and Leadbetter, ER (1999). Sulfidogenesis from 2-aminoethanesulfonate (taurine) fermentation by a morphologically unusual sulfate-reducing bacterium, Desulforhopalus singaporensis sp. nov. Applied and Environmental Microbiology 65: 33283334.CrossRefGoogle ScholarPubMed
Lin, J (2011). Effect of antibiotic growth promoters on intestinal microbiota in food animals: a novel model for studying the relationship between gut microbiota and human obesity? Frontiers in Cellular and Infection Microbiology 2: 53. doi: 10.3389/ fmicb.2011.00053.Google ScholarPubMed
Lin, J (2014). Antibiotic growth promoters enhance animal production by targeting intestinal bile salt hydrolase and its producers. Frontiers in Microbiology 5: 33. doi: 10.3389/fmicb.2014.00033.CrossRefGoogle ScholarPubMed
Lin, J, Negga, R, Zeng, X and Smith, K (2014). Effect of bile salt hydrolase inhibitors on a bile salt hydrolase from Lactobacillus acidophilus . Pathogens 3: 947956.CrossRefGoogle ScholarPubMed
Liong, M and Shah, N (2005). Acid and bile tolerance and cholesterol removal ability of lactobacilli strains. Journal of Dairy Science 88: 5566.CrossRefGoogle ScholarPubMed
Lundeen, SG and Savage, DC (1990). Characterization and purification of bile salt hydrolase from Lactobacillus sp. strain 100-100. Journal of Bacteriology 172: 41714177.CrossRefGoogle ScholarPubMed
Marteau, P and Rambaud, J-C (1993). Potential of using lactic acid bacteria for therapy and immunomodulation in man. FEMS Microbiology Reviews 12: 207220.CrossRefGoogle ScholarPubMed
Masuda, N (1981). Deconjugation of bile salts by Bacteroides and Clostridium . Microbiology and Immunology 25: 111.CrossRefGoogle Scholar
McAuliffe, O, Cano, RJ and Klaenhammer, TR (2005). Genetic analysis of two bile salt hydrolase activities in Lactobacillus acidophilus NCFM. Applied and Environmental Microbiology 71: 49254929.CrossRefGoogle ScholarPubMed
McGarr, SE, Ridlon, JM and Hylemon, PB (2005). Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. Journal of Clinical Gastroenterology 39: 98109.Google ScholarPubMed
Miyoshi, M, Ogawa, A, Higurashi, S and Kadooka, Y (2014). Anti-obesity effect of Lactobacillus gasseri SBT2055 accompanied by inhibition of pro-inflammatory gene expression in the visceral adipose tissue in diet-induced obese mice. European Journal of Nutrition 53: 599606.CrossRefGoogle ScholarPubMed
Mountzouris, K, Tsitrsikos, P, Palamidi, I, Arvaniti, A, Mohnl, M, Schatzmayr, G and Fegeros, K (2010). Effects of probiotic inclusion levels in broiler nutrition on growth performance, nutrient digestibility, plasma immunoglobulins, and cecal microflora composition. Poultry Science 89: 5867.CrossRefGoogle ScholarPubMed
Mukherji, R and Prabhune, A (2015). Possible correlation between bile salt hydrolysis and AHL deamidation: Staphylococcus epidermidis RM1, a potent quorum quencher and bile salt hydrolase producer. Applied Biochemistry and Biotechnology 176: 140150.CrossRefGoogle ScholarPubMed
Nair, P, Gordon, M and Reback, J (1967). The enzymatic cleavage of the carbon-nitrogen bond in 3α, 7α, 12α-trihydroxy-5β-cholan-24-oylglycine. Journal of Biological Chemistry 242: 711.CrossRefGoogle Scholar
Oh, H-K, Lee, JY, Lim, SJ, Kim, MJ, Kim, G-B, Kim, JH, Hong, S-K and Kang, D-K (2008). Molecular cloning and characterization of a bile salt hydrolase from Lactobacillus acidophilus PF01. Journal of Microbiology and Biotechnology 18: 449456.Google ScholarPubMed
Ou, J, Carbonero, F, Zoetendal, EG, DeLany, JP, Wang, M, Newton, K, Gaskins, HR and O'Keefe, SJ (2013). Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. American Journal of Clinical Nutrition 98: 111120.CrossRefGoogle ScholarPubMed
Park, D-Y, Ahn, Y-T, Park, S-H, Huh, C-S, Yoo, S-R, Yu, R, Sung, M-K, McGregor, RA and Choi, M-S (2013). Supplementation of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 in diet-induced obese mice is associated with gut microbial changes and reduction in obesity. PloS ONE 8: e59470.CrossRefGoogle ScholarPubMed
Park, JE, Oh, SH and Cha, YS (2014). Lactobacillus plantarum LG42 isolated from gajami sik-hae decreases body and fat pad weights in diet-induced obese mice. Journal of Applied Microbiology 116: 145156.CrossRefGoogle ScholarPubMed
Patel, AK, Singhania, RR, Pandey, A and Chincholkar, SB (2010). Probiotic bile salt hydrolase: current developments and perspectives. Applied Biochemistry and Biotechnology 162: 166180.CrossRefGoogle Scholar
Pato, U, Surono, IS and Hosono, A (2004). Hypocholesterolemic effect of indigenous dadih lactic acid bacteria by deconjugation of bile salts. Asian-Australasian Journal of Animal Sciences 17: 17411745.CrossRefGoogle Scholar
Pavlović, N, Stankov, K and Mikov, M (2012). Probiotics-interactions with bile acids and impact on cholesterol metabolism. Applied Biochemistry and Biotechnology 168: 18801895.CrossRefGoogle ScholarPubMed
Pereira-Fantini, PM, Lapthorne, S, Joyce, SA, Dellios, NL, Wilson, G, Fouhy, F, Thomas, SL, Scurr, M, Hill, C and Gahan, CG (2014). Altered FXR signalling is associated with bile acid dysmetabolism in short bowel syndrome-associated liver disease. Journal of Hepatology 61: 11151125.CrossRefGoogle ScholarPubMed
Perry, JA, Westman, EL and Wright, GD (2014). The antibiotic resistome: what's new? Current Opinion in Microbiology 21: 4550.CrossRefGoogle ScholarPubMed
Pinto, MGV, Franz, CM, Schillinger, U and Holzapfel, WH (2006). Lactobacillus spp. with in vitro probiotic properties from human faeces and traditional fermented products. International Journal of Food Microbiology 109: 205214.CrossRefGoogle Scholar
Qiao, L, Han, SI, Fang, Y, Park, JS, Gupta, S, Gilfor, D, Amorino, G, Valerie, K, Sealy, L and Engelhardt, JF (2003). Bile acid regulation of C/EBPβ, CREB, and c-Jun function, via the extracellular signal-regulated kinase and c-Jun NH2-terminal kinase pathways, modulates the apoptotic response of hepatocytes. Molecular and Cellular Biology 23: 30523066.CrossRefGoogle ScholarPubMed
Reinhart, G, Mahan, D and Cera, K (1988). Effect of bile salt supplementation on tallow digestion and serum vitamin E concentration in weanling pigs. Nutrition Reports International 38: 563570.Google Scholar
Ridlon, JM, Kang, D-J and Hylemon, PB (2006). Bile salt biotransformations by human intestinal bacteria. Journal of Lipid Research 47: 241259.CrossRefGoogle ScholarPubMed
Roberts, MS, Magnusson, BM, Burczynski, FJ and Weiss, M (2002). Enterohepatic circulation. Clinical Pharmacokinetics 41: 751790.CrossRefGoogle ScholarPubMed
Rose, P, Moore, PK, Ming, SH, Nam, OC, Armstrong, JS and Whiteman, M (2005). Hydrogen sulfide protects colon cancer cells from chemopreventative agent beta-phenylethyl isothiocyanate induced apoptosis. World Journal of Gastroenterology 11: 3990.CrossRefGoogle ScholarPubMed
Rossocha, M, Schultz-Heienbrok, R, von Moeller, H, Coleman, JP and Saenger, W (2005). Conjugated bile acid hydrolase is a tetrameric N-terminal thiol hydrolase with specific recognition of its cholyl but not of its tauryl product. Biochemistry 44: 57395748.CrossRefGoogle Scholar
Russell, DW (2009). Fifty years of advances in bile acid synthesis and metabolism. Journal of Lipid Research 50: S120S125.CrossRefGoogle ScholarPubMed
Schaap, FG, Trauner, M and Jansen, PL (2014). Bile acid receptors as targets for drug development. Nature Reviews Gastroenterology & Hepatology 11: 5567.CrossRefGoogle ScholarPubMed
Sharifi, S, Dibamehr, A, Lotfollahian, H and Baurhoo, B (2012). Effects of flavomycin and probiotic supplementation to diets containing different sources of fat on growth performance, intestinal morphology, apparent metabolizable energy, and fat digestibility in broiler chickens. Poultry Science 91: 918927.CrossRefGoogle ScholarPubMed
Sieladie, DV, Zambou, NF, Kaktcham, PM, Cresci, A and Fonteh, F (2011). Probiotic properties of lactobacilli strains isolated from raw cow milk in the western highlands of Cameroon. Innovative Romanian Food Biotechnology 9: 1228.Google Scholar
Smet, I, Hoorde, L, Woestyne, M, Christiaens, H and Verstraete, W (1995). Significance of bile salt hydrolytic activities of lactobacilli. Journal of Applied Bacteriology 79: 292301.CrossRefGoogle ScholarPubMed
Smith, K, Zeng, X and Lin, J (2014). Discovery of bile salt hydrolase inhibitors using an efficient high-throughput screening system. PloS ONE 9: e85344.CrossRefGoogle ScholarPubMed
Sridevi, N, Srivastava, S, Khan, BM and Prabhune, AA (2009). Characterization of the smallest dimeric bile salt hydrolase from a thermophile Brevibacillus sp. Extremophiles 13: 363370.CrossRefGoogle ScholarPubMed
Stellwag, E and Hylemon, P (1976). Purification and characterization of bile salt hydrolase from Bacteroides fragilis subsp. fragilis. Biochimica et Biophysica Acta (BBA)-Enzymology 452: 165176.CrossRefGoogle ScholarPubMed
Svensson, P-A, Olsson, M, Andersson-Assarsson, JC, Taube, M, Pereira, MJ, Froguel, P and Jacobson, P (2013). The TGR5 gene is expressed in human subcutaneous adipose tissue and is associated with obesity, weight loss and resting metabolic rate. Biochemical and Biophysical Research Communications 433: 563566.CrossRefGoogle ScholarPubMed
Tanaka, H, Hashiba, H, Kok, J and Mierau, I (2000). Bile salt hydrolase of Bifidobacterium longum-biochemical and genetic characterization. Applied and Environmental Microbiology 66: 25022512.CrossRefGoogle ScholarPubMed
Tremaroli, V and Bäckhed, F (2012). Functional interactions between the gut microbiota and host metabolism. Nature 489: 242249.CrossRefGoogle ScholarPubMed
Van Eldere, J, Celis, P, De Pauw, G, Lesaffre, E and Eyssen, H (1996). Tauroconjugation of cholic acid stimulates 7 alpha-dehydroxylation by fecal bacteria. Applied and Environmental Microbiology 62: 656661.CrossRefGoogle ScholarPubMed
Van Faassen, A, Bol, J, van Dokkum, W, Pikaar, NA, Ockhuizen, T and Hermus, R (1987). Bile acids, neutral steroids, and bacteria in feces as affected by a mixed, a lacto-ovovegetarian, and a vegan diet. American Journal of Clinical Nutrition 46: 962967.CrossRefGoogle ScholarPubMed
Van Gaal, LF, Mertens, IL and Christophe, E (2006). Mechanisms linking obesity with cardiovascular disease. Nature 444: 875880.CrossRefGoogle ScholarPubMed
Venneman, NG and van Erpecum, KJ (2010). Pathogenesis of gallstones. Gastroenterology Clinics of North America 39: 171183.CrossRefGoogle ScholarPubMed
Vlahcevic, Z, Heuman, D and Hylemon, P (1996). Physiology and pathophysiology of enterohepatic circulation of bile acids. Hepatology: a Textbook of Liver Disease 1: 376417.Google Scholar
Walker, AW and Parkhill, J (2013). Fighting obesity with bacteria. Science 341: 10691070.CrossRefGoogle ScholarPubMed
Wang, Z, Zeng, X, Mo, Y, Smith, K, Guo, Y and Lin, J (2012). Identification and characterization of a bile salt hydrolase from Lactobacillus salivarius for development of novel alternatives to antibiotic growth promoters. Applied and Environmental Microbiology 78: 87958802.CrossRefGoogle ScholarPubMed
Watanabe, M, Houten, SM, Wang, L, Moschetta, A, Mangelsdorf, DJ, Heyman, RA, Moore, DD and Auwerx, J (2004). Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. Journal of Clinical Investigation 113: 14081418.CrossRefGoogle Scholar
Watanabe, M, Houten, SM, Mataki, C, Christoffolete, MA, Kim, BW, Sato, H, Messaddeq, N, Harney, JW, Ezaki, O and Kodama, T (2006). Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439: 484489.CrossRefGoogle ScholarPubMed
Xu, F, Guo, F, Hu, X-J and Lin, J (2016). Crystal structure of bile salt hydrolase from Lactobacillus salivarius . Acta Crystallographica Section F: Structural Biology Communications 72: 376381.Google ScholarPubMed
Zhang, Y, Lee, FY, Barrera, G, Lee, H, Vales, C, Gonzalez, FJ, Willson, TM and Edwards, PA (2006). Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proceedings of the National Academy of Sciences of the United States of America 103: 10061011.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. The BSH enzymes identified in bacteria from various sources