microorganisms

Article Identification of Bile Salt Hydrolase and Bile Salt Resistance in a Probiotic Bacterium gasseri JCM1131T

Hiroyuki Kusada 1,*, Kana Morinaga 1 and Hideyuki Tamaki 1,2,*

1 Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8566, Japan; [email protected] 2 Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan * Correspondence: [email protected] (H.K.); [email protected] (H.T.); Tel.: +81-29-861-6591 (H.K.); +81-29-861-6592 (H.T.); Fax: +81-29-861-6587 (H.K. & H.T.)

Abstract: Lactobacillus gasseri is one of the most likely probiotic candidates among many Lactobacillus species. Although bile salt resistance has been defined as an important criterion for selection of probiotic candidates since it allows probiotic to survive in the gut, both its capability and its related enzyme, bile salt hydrolase (BSH), in L. gasseri is still largely unknown. Here, we report that the well-known probiotic bacterium L. gasseri JCM1131T possesses BSH activity and bile salt resistance capability. Indeed, this strain apparently showed BSH activity on the plate assay and highly tolerated the primary bile salts and even taurine-conjugated secondary bile salt. We further isolated a putative BSH enzyme (LagBSH) from strain JCM1131T and characterized the enzymatic function. The purified LagBSH protein exhibited quite high deconjugation activity for taurocholic acid and taurochenodeoxycholic acid. The lagBSH gene was constitutively expressed in strain JCM1131T,  suggesting that LagBSH likely contributes to bile salt resistance of the strain and may be associated  with survival capability of strain JCM1131T within the human intestine by bile detoxification. Thus, Citation: Kusada, H.; Morinaga, K.; this study first demonstrated the bile salt resistance and its responsible enzyme (BSH) activity in strain Tamaki, H. Identification of Bile Salt JCM1131T, which further supports the importance of the typical lactic acid bacterium as probiotics. Hydrolase and Bile Salt Resistance in a Probiotic Bacterium Lactobacillus Keywords: bile salt hydrolase; Lactobacillus gasseri; Ntn-hydrolase family protein; probiotics gasseri JCM1131T. Microorganisms 2021, 9, 1011. https://doi.org/ 10.3390/microorganisms9051011 1. Introduction Academic Editor: Julio Villena Lactobacillus species have been considered as one of the major targets of probiotic Received: 13 April 2021 research. Several Lactobacillus species provide positive impacts on human health; symp- Accepted: 4 May 2021 tomatic improvements by probiotics have been reported in cases of various hard-to-heal Published: 8 May 2021 diseases, e.g., allergy [1], diarrhea [2], Helicobacter pylori infection [3], and irritable bowel syndrome [4]. These probiotic effects are generally strain-specific and differ depending Publisher’s Note: MDPI stays neutral on each strain even among Lactobacillus strains of same species [5–9]. Among the several with regard to jurisdictional claims in criteria for selecting candidate probiotic strains of Lactobacillus spp., bile salt resistance is published maps and institutional affil- one of the most important selective criteria, since bile salts are well known as strong surfac- iations. tants and bile exposure in gastrointestinal tract is intensely toxic for probiotic Lactobacillus species to survive and retain activity in human intestine [10,11]. Bile salt resistance is mainly provided by bile salt hydrolase (BSH, EC3.5.1.24), an enzyme that deconjugates glycine and/or taurine-conjugated bile salts [12], though other Copyright: © 2021 by the authors. resistance mechanisms (i.e., efflux pumps, stress response proteins, and cell wall mod- Licensee MDPI, Basel, Switzerland. ifications) have been reported [13]. Genes encoding BSH have been found in various This article is an open access article Lactobacillus species [14], and the number of bsh gene orthologs vary in accordance with distributed under the terms and species and strains [11,14–17]. BSH enzymes perform a crucial role in bile detoxification conditions of the Creative Commons and thereby improve the colonization and survival of host probiotic bacteria in the human Attribution (CC BY) license (https:// gastrointestinal tract [18]. In addition, BSH enzymes are known to involve in the reduction creativecommons.org/licenses/by/ of blood cholesterol levels, the regulation of lipid absorption, glucose metabolism, and 4.0/).

Microorganisms 2021, 9, 1011. https://doi.org/10.3390/microorganisms9051011 https://www.mdpi.com/journal/microorganisms Microorganisms 2021, 9, 1011 2 of 12

energy homeostasis in humans [19], which means that the bile salt deconjugation ability through the enzyme BSH has been widely recognized as a probiotic biomarker [20]. Lactobacillus gasseri type strain JCM1131T is commensal, produces lactic acid, and is widely known as a typical probiotic bacterium. In fact, several probiotic properties of this strain have been reported [21–23]. However, it has remained largely unclear whether strain JCM1131T has BSH activity and bile salt resistance capability. In the present study, we demonstrated the BSH activity and bile salt resistance ability in L. gasseri strain JCM1131T. We found the putative bsh gene in the genome and determined that its recombinant protein could functionally act as BSH mediating the bile salt resistance in the strain through molecular cloning, biochemical characterization, and transcriptional analyses.

2. Materials and Methods 2.1. Bacterial Strains Used in This Study A probiotic lactic acid bacterium, Lactobacillus gasseri JCM1131T (=DSM20243T=ATCC 33323T), was obtained from the Japan Collection of Microorganisms (RIKEN BRC, Tsukuba, Japan). This strain was cultivated using Gifu anaerobic medium (GAM, Nissui Phar- maceutical Co., Ltd., Tokyo, Japan) and de Man–Rogosa–Sharpe medium (MRS, Difco ◦ Laboratories, Detroit, MI, USA) with headspace gas of N2/CO2 (80:20, v/v) at 37 C under anaerobic conditions. Escherichia coli strain BL21 (DE3) ChampionTM21 (SMOBIO Tech- nologies, Hsinchu City, Taiwan) was used for heterologous expression experiments. E. coli was cultured in LB broth supplemented with 50 µg/mL kanamycin (FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan) at 37 ◦C with shaking.

2.2. Cloning and Heterologous Expression of a Putative Bile Salt Hydrolase Gene Based on the sequence analyses and homology searches using NCBI BLAST pro- gram (https://blast.ncbi.nlm.nih.gov/Blast.cgi) (accessed on 8 June 2020), UniProt BLAST tool (https://www.uniprot.org/blast/) (accessed on 8 June 2020), InterProScan (http: //www.ebi.ac.uk/interpro/search/sequence-search) (accessed on 8 June 2020), and Pfam (http://pfam.xfam.org/) (accessed on 8 June 2020), we screened a gene encoding putative BSH from the complete genome sequence of strain JCM1131T (accession number CP000413). A putative bsh gene (named as lagBSH) was commercially synthesized with codon op- timization for heterologous expression in E. coli (GenScript, Piscataway, NJ, USA). The lagBSH gene was subcloned into the NdeI and EcoRI sites of pET28-b (Novagen, Madison, WI, USA) expression vector. The heterologous gene expression and protein purification experiments were per- formed according to our previous study with slight modifications [24]. In brief, the con- structed plasmid was transformed into E. coli BL21 (DE3) ChampionTM21 competent cells ◦ and E. coli strain was cultured on LB broth at 37 C until OD600 reached 0.4–0.6. Isopropyl- β-D-thiogalactopyranoside (IPTG, Nacalai Tesque, Kyoto, Japan) was added at the final concentration of 100 µM to the culture medium. After adding of IPTG, the E. coli cells were incubated at 20 ◦C for overnight with shaking. The cells were harvested by centrifugation at 5800× g for 10 min, suspended in buffer (20 mM Tris, 150 mM NaCl, 5% glycerol, 5 mM imidazole, pH 7.5), and disrupted for 5 min by sonication using an ultrasonic disintegrator (Sonicator Branson Sonifier 250 (Branson, Danbury, CT, USA); output control: 5, duty cycle: 50) in an ice-water bath. The cell debris were removed by centrifugation and the result- ing supernatant was mixed with Ni-NTA Agarose HP (FUJIFILM Wako Pure Chemical Corporation). The His6-tagged recombinant protein was washed and eluted according to the previous study [24]. The eluted fraction was further dialyzed with buffer (20 mM Tris, 150 mM NaCl, 5% glycerol) using semipermeable membrane (Spectra/Por 3 membrane MWCO: 3500, Repligen, Waltham, MA, USA) and concentrated using Amicon Ultra cen- trifugal filter devices (30,000 MWCO, Millipore, Billerica, MA, USA). The purified protein was treated with sample buffer (Bio-Rad, Hercules, CA, USA), heat-denatured at 95 ◦C for 5 min, and analyzed on sodium dodecyl sulfate polyacrylamide gel electrophoresis Microorganisms 2021, 9, 1011 3 of 12

(SDS-PAGE) using 12% Mini-PROTEAN TGX precast polyacrylamide gel (Bio-Rad) [24]. The gel was stained with QC Colloidal Coomassie Stain (Bio-Rad) through gentle agitation.

2.3. Bile Salt Hydrolase Activity The bile salt hydrolyzing activity of purified LagBSH was determined as described previously [25]. In brief, purified protein was mixed with 0.24 mg/100 µL of conjugated bile salts (glycocholic acid (GCA, Sigma-Aldrich, St. Louis, MO, USA), glycodeoxycholic acid (GDCA, Sigma-Aldrich), taurocholic acid (TCA, Nacalai Tesque), taurochenodeoxycholic acid (TCDCA, Sigma-Aldrich), and taurodeoxycholic acid (TDCA, Nacalai Tesque)) and incubated at 37 ◦C. The reaction was stopped by adding 15% trichloroacetic acid (FUJIFILM Wako Pure Chemical Corporation) and the resulting solution was centrifuged at 10,000× g for 15 min at 20 ◦C. The supernatant was then mixed with 0.3 M borate buffer with 1% SDS (pH 9.5), and 0.3% 2,4,6-trinitrobenzenesulfonic acid solution (Tokyo Kasei Kogyo Co., Ltd., Tokyo, Japan). The resulting mixture was incubated for 30 min at room temperature under dark condition, and then 0.6 mM HCl was added to stop the reaction. The absorbance at 416 nm was measured using a SPARK 10M multimode microplate reader (TECAN, Männedorf, Switzerland). The assays were performed in eight replicates. Student’s t-test was used to assess the presence of statistically significant differences (α = 0.05) using GraphPad Prism version 8.0 software program (GraphPad Software, San Diego, CA, USA). As a negative control, a bile salt solution was reacted with buffer instead of purified protein.

2.4. Biochemical Characterization The optimum pH and temperatures of LagBSH were determined according to the methods described [20] with slight modifications as follows. The purified LagBSH protein was mixed with taurocholic acid (TCA) at selected temperatures (10–90 ◦C, in intervals of 10 ◦C) and pH (pH 3.0–pH 10.0, in intervals of pH 1.0) ranges. To determine the effects of pH on enzyme activity of LagBSH, we used various Good’s buffer solution based on the pH range (acetate buffer [CH3COONa·3H2O] pH 3.0–4.0; MES buffer [C6H13NO4S·H2O] pH 5.0–6.0; HEPES buffer [C8H18N2O4S] pH 7.0–8.0; CAPS [C9H19NO3S] pH9.0–10.0). Af- ter incubation for 6 h, the released taurine was detected as described above. All experiments were performed in eight replicates.

2.5. Bile Salt Tolerance Test The bile salt tolerance ability of Lactobacillus gasseri JCM1131T was estimated and calculated from its survival rates according to the previous study [26]. In brief, a full- grown culture of strain JCM1131T was mixed with GCA, GDCA, TCA, and TDCA at final concentrations of 0.05%. Cells were anaerobically incubated at 37 ◦C for 6 h, and the optimal densities (OD600) were measured every hour using an Ultrospec 500 Pro visible spectrophotometer (GE Healthcare Life Sciences, Buckinghamshire, UK). As a negative control, strain JCM1131T was incubated in GAM medium without bile salt. The assays were performed in triplicates. Minimum inhibitory concentrations (MICs) were determined as the lowest concen- tration of bile salts preventing visible growth of L. gasseri JCM1131T on MRS agar. Strain JCM1131T was cultivated in MRS broth and inoculated on MRS agar plate with a selected bile salt. Plates were anaerobically incubated at 37 ◦C for 5 days. The tested bile salts were GCA, GDCA, TCA, and TDCA at final concentrations of 0.01%, 0.05%, 0.1%, 0.25%, and 0.5%. All experiments were carried out in triplicates.

2.6. Structural Modeling Three-dimensional conformations of LagBSH were predicted using Swiss-Model workspace (https://swissmodel.expasy.org/) (accessed on 8 June 2020). [27]. The superpo- sition analyses were performed and visualized using UCSF Chimera software [28]. Crystal structure of a known BSH enzyme, CpBSH from Clostridium perfringens 13 [29], was pro- Microorganisms 2021, 9, 1011 4 of 12

Microorganisms 2021, 9, x FOR PEER videdREVIEW by Protein Data Bank (http://www.rcsb.org/pdb/home/home.do)4 (accessedof 12 on 8 June 2020).

2.7. Transcriptional Analysis structure of a known BSH enzyme, CpBSH from Clostridium perfringens 13 [29], was pro- videdReverse by Protein transcription Data Bank (http:/ polymerase/www.rcsb.org/pdb/home/home.do). chain reaction (RT-PCR) analyses of the lagBSH gene were performed as follows. Strain JCM1131T was cultured on MRS broth with or without2.7. Transcriptional TCA and Analysis TDCA at final concentration of 0.05%. The total RNA samples were isolatedReverse using transcription the RNeasy polymerase Mini Kit (Qiagen,chain reaction Germantown, (RT-PCR) analyses MD, USA), of the and lagBSH the resulting RNAgene were samples performed were treatedas follows. with Strain TURBO JCM1131™ DNaseT was cultured (Thermo on Fisher MRS broth Scientific, with or Waltham, MA,without USA) TCA to removeand TDCA contaminated at final concentration genomic DNA.of 0.05%. The The presence total RNA of chromosomal samples were genomic DNAisolated was using confirmed the RNeasy by Mi PCRni Kit analysis (Qiagen, with Germantown, the 16S rRNA MD, USA), gene and universal the resulting PCR primers 530FRNA andsamples 907R were using treated each with RNA TURBO™ samples DNase as template. (Thermo Fisher Reverse Scientific, transcription Waltham, reactions wereMA, performedUSA) to remove using contaminated SuperScript IVgenomic Reverse DNA. Transcriptase The presence (Thermo of chromosomal Fisher Scientific) ge- in a nomic DNA was confirmed by PCR analysis with the 16S rRNA gene universal PCR pri- 25 µL reaction volume according to the manufacturer’s instruction. The synthesized cDNA mers 530F and 907R using each RNA samples as template. Reverse transcription reactions samples were used as the PCR template with the following two PCR primer sets: lagBSH- were performed using SuperScript IV Reverse Transcriptase (Thermo Fisher Scientific) in Aseta 25 (5’-TCACACCACGCAACTATCCTC-3’μL reaction volume according to the manufacturer’s and 5’-GTTGCCAAGGTTAGTAAGATGCC- instruction. The synthesized 3’,cDNA amplicon samples size: were 467 used bp) as andthe PCRlagBSH template-Bset with (5’-TTAGCTTCTTACGAAATTATGC-3’ the following two PCR primer sets: and 5’-GAATGCTATCACCTGGTAAAC-3’,lagBSH-Aset (5’-TCACACCACGCAACTATCCTC-3’ amplicon and size: 5’-GTTGCCAAGGTTAGTAA- 376 bp). The PCR products were analyzedGATGCC-3’, using amplicon agarose gelsize: electrophoresis 467 bp) and in 2.0%lagBSH agarose-Bset and(5’-TTAGCTTCTTAC- were stained with Gelred (FujifilmGAAATTATGC-3’ Wako Pure and Chemical 5’-GAATGCTATCACCTGG Corporation). TAAAC-3’, amplicon size: 376 bp). The PCR products were analyzed using agarose gel electrophoresis in 2.0% agarose and 3.were Results stained and with Discussion Gelred (Fujifilm Wako Pure Chemical Corporation). 3.1. Identification of BSH Activity and Bile Salt Resistance of Lactobacillus gasseri JCM1131T 3. Results and Discussion In the present study, we first investigated whether L. gasseri JCM1131T shows BSH 3.1. Identification of BSH Activity and Bile Salt Resistance of Lactobacillus gasseri JCM1131T activity using the standard plate assay method. We observed that the visible halo surround- In the present study, we first investigated whether L. gasseri JCM1131T shows BSH ing colonies and the white precipitates with colonies when strain JCM1131T was cultured activity using the standard plate assay method. We observed that the visible halo sur- on an MRS agar plate supplemented with taurodeoxycholic acid (TDCA), one of the major rounding colonies and the white precipitates with colonies when strain JCM1131T was conjugatedcultured on an bile MRS salts agar in plate human supplemented gastrointestinal with taurodeoxycholic tract (Figure1). acid These (TDCA), characteristics one of (i.e., halothe major and whiteconjugated precipitates) bile salts in are human the well-known gastrointestinal indicators tract (Figure of BSH 1). These activity character- [19,30], clearly T suggestingistics (i.e., halo that and strain white JCM1131 precipitates)represents are the BSHwell-known activity, indicators though of the BSH previous activity study re- T T ported[19,30], thatclearlyL. gasserisuggestingATCC33323 that strain JCM1131(=JCM1131T represents) showed BSH no activity, significant though BSH the activitypre- [31]. Importantly,vious study reported Allain that et al. L. gasseri demonstrated ATCC33323 thatT (=JCM1131L. gasseriT)strain showed CNCM no significant I-4884 BSH with strong BSHactivity activity [31]. Importantly, exhibited significant Allain et al. antiparasitic demonstrated ability that thatL. gasseri antagonizes strain CNCM growth I-4884 of the most commonwith strong waterborne BSH activity parasite exhibited (Giardia significant)[32] and antiparasitic further revealed ability that theantagonizes antiparasitic ef- fectsgrowth of ofLactobacillus the most commonspp. were waterborne well correlated parasite (Giardia with the) [32] expression and further of revealed BSH activities that [32]. Basedthe antiparasitic on this fact, effects we expectof Lactobacillus that L. spp. gasseri wereJCM1131 well correlatedT with BSH with activitythe expression may also of exhibit T antiparasiticBSH activities activity[32]. Based as on well this as fact, strain we expect CNCM that I-4884, L. gasseri though JCM1131 future with study BSH needsactivity to clarify may also exhibit antiparasitic activity as well as strain CNCM I-4884, though future study this point. needs to clarify this point.

T FigureFigure 1. BileBile salt salt hydrolase hydrolase activity activity in Lactobacillus in Lactobacillus gasseri gasseri JCM1131JCM1131. Full-grownT. Full-grown culture of culture L. gasseri of L. gasseri JCM1131T was streaked on an MRS agar plate (A) or an MRS agar plate supplemented with 0.25% T A JCM1131taurodeoxycholicwas streaked acid (B). onThe an plates MRS were agar anaerobically plate ( ) or incubated an MRS agarat 37 °C plate for supplemented5 days. The visible with 0.25% taurodeoxycholic acid (B). The plates were anaerobically incubated at 37 ◦C for 5 days. The visible halo surrounding colonies and the white precipitates with colonies are the well-known indicator of bacterial BSH activity [19,30].

Microorganisms 2021, 9, x FOR PEER REVIEW 5 of 12

halo surrounding colonies and the white precipitates with colonies are the well-known indicator of bacterial BSH activity [19,30]. Microorganisms 2021, 9, 1011 5 of 12 We further determined the survivability of L. gasseri JCM1131T against four different conjugated bile salts (GCA, GDCA, TCA, and TDCA) at final concentration of 0.05%. As shown in Figure 2, strainWe further JCM1131 determinedT showed the survivabilityhigh survivability of L. gasseri towardJCM1131 primaryT against bile four differentsalts (TCA and GCA) andconjugated the survival bile salts rates (GCA, of GDCA,strain TCA,JCM1131 and TDCA)T against at final TCA concentration and GCA reached of 0.05%. As T above 90% after exposedshown in Figureto the2 ,bile strain salts JCM1131 for 6 h.showed Additionally, high survivability this strain toward also primary exhibited bile salts (TCA and GCA) and the survival rates of strain JCM1131T against TCA and GCA reached moderate and lowabove survivability 90% after exposedtoward to TDCA the bile and salts GDCA for 6 h. Additionally,(secondary thisbile strainsalts), also and exhibited the survival rates againstmoderate TDCA and lowand survivability GDCA were toward above TDCA 70% and and GDCA below (secondary 60%, bilerespectively salts), and the (Figure 2). survival rates against TDCA and GDCA were above 70% and below 60%, respectively (Figure2).

FigureFigure 2. Bile 2. salt Bile tolerance salt tolerance activity in activityLactobacillus in gasseriLactobacillusJCM1131 gasseriT. Full-grown JCM1131 cultureT. Full-grown of strain JCM1131 cultureT was of mixed strain withJCM1131 GCA, GDCA,T was TCA, mixed and TDCAwith GCA, at final GDCA, concentrations TCA,of and 0.05% TDCA and incubated at final concentrations anaerobically at 37 of◦ C.0.05% The optimal and in- densitycubated (OD600 anaerobically) was measured everyat 37 hour °C. andThe survival optimal rates density were calculated (OD600) aswas described measured previously every [26 hour]. The and survival survival rate of controlrates (withoutwere calculated bile salt) was as defined described as 100%. previously Results indicated [26]. The mean survival± SD obtained rate of in control triplicate (without experiments. bile salt) was defined as 100%. Results indicated mean ± SD obtained in triplicate experiments. We further investigated the bile salt tolerance capacity of L. gasseri JCM1131T by determining the minimum inhibitory concentrations (MICs). As shown in Table1, this We further investigatedstrain showed the low bile MIC salt value tolerance (0.05%) to capacity GDCA, indicating of L. gasseri that GDCAJCM1131 is toxicT by to de- strain termining the minimumJCM1131 inhibitoryT. De Smet et concentrations al. suggested that (MICs). the high As toxicity shown of GDCA in Table would 1, bethis caused strain by its showed low MICweak value acid property(0.05%) (TDCA to GDCA, is strong indi acidcating property) that [33]. GDCA They further is hypothesizedtoxic to strain that the T protonated form of bile salts exhibited toxicity as it imported protons in the cell [33]. This JCM1131 . De Smethypothesis et al. suggested seems to be that reasonable the high since toxicity weak acids of GDCA are pretty would much easierbe caused to protonate by its weak acid propertythan strong (TDCA acids. is However, strong acid strain property) JCM1131T displayed [33]. They a higher further resistance hypothesized ability (MICs that the protonatedwere form >0.5%) of bile to a secondarysalts exhibite bile saltd toxicity (TDCA) as wellit imported as the primary protons bile saltsin the (TCA cell and [33]. This hypothesisGCA) seems (Table1 to), despite be reasonable the fact that since secondary weak bile acids salts haveare pretty been known much to beeasier more to toxic protonate than strongthan primaryacids. However, bile salts [34 strain]. Since JCM1131 the averageT displayed bile concentration a higher in resistance human intestine abil- has ity (MICs were >0.5%) to a secondary bile salt (TDCA) as well as the primary bile salts (TCA and GCA) (Table 1), despite the fact that secondary bile salts have been known to be more toxic than primary bile salts [34]. Since the average bile concentration in human intestine has been estimated to be 0.3% w/v [35], our findings suggest that strain JCM1131T

Microorganisms 2021, 9, 1011 6 of 12

been estimated to be 0.3% w/v [35], our findings suggest that strain JCM1131T could have bile salt tolerance ability toward TCA, GCA, and TDCA. Previous studies reported that other strains of L. gasseri (BGHO89, 4M13, and FR4) showed high bile salt resistance ability (toward more than 0.3% bile salts) [36–38], suggesting that gut-derived L. gasseri strains would generally have bile salt resistance ability to survive and colonize the mammalian digestive tracts. Interestingly, these L. gasseri strains (BGHO89, 4M13, and FR4) with high bile salt resistance capacity further exhibited some probiotic functions including acid tolerance, bacteriocin production, antioxidation, and cholesterol-lowering activity [36–38]. Thus, although it has been reported that some other strains of L. gasseri show bile salt tolerance so far [21,36,37], the correlations between their bile salt tolerance ability and BSH activity have not been well demonstrated. In the present study, we first revealed both bile salt tolerance capability and its related key enzymatic function (BSH activity) in L. gasseri JCM1131T, and these findings provide additional insights into the probiotic function in a well-known representative of the probiotic lactic acid bacterium.

Table 1. Minimum inhibitory concentrations of bile salts against strain JCM1131T a.

Minimum Inhibitory Concentrations (%) Strain Substrates TCA TDCA GCA GDCA JCM1131T >0.5 >0.5 >0.5 0.05 a TCA, taurocholic acid; TDCA, taurodeoxycholic acid; GCA, glycocholic acid; GDCA, glycodeoxycholic acid. The tested concentrations of bile salts were 0.01, 0.05, 0.1, 0.25, and 0.5%.

3.2. Sequence and Phylogenetic Analyses of a Putative BSH Gene Since both BSH activity and bile salt resistance capability of L. gasseri JCM1131T were revealed, we then performed cloning and heterologous expression of the gene candidates associated with BSH activity. We herein found a putative bile salt hydrolase gene (desig- nated as lagBSH) in L. gasseri JCM1131T genome (CP000413) based on the sequence analyses and homology searches (Figure3A). The putative lagBSH gene comprises 951 bp. The deduced amino acid sequence of LagBSH (316 amino acids) was related to the cholylglycine hydrolase family of the Ntn-hydrolase superfamily proteins based on the domain and sequence comparison. The multiple amino acid sequence alignments revealed that LagBSH protein shared five residues (Cys, Arg, Asp, Asn, and Arg) associated with active site with previously identified BSHs from Lactobacillus species (Figure3B). Three-dimensional superposition analyses further revealed that the overall structure of LagBSH is composed of well-known αββα-sandwich folds of cholylglycine hydrolase proteins (Supplementary Figure S1A), which are similar to the structure of CpBSH, BSH from Clostridium perfringens 13 [29]. The putative LagBSH further conserved the catalytic active site structure identified with CpBSH (Supplementary Figure S1B). It has been reported in previous studies that N-terminal cysteine residue (Cys-2) plays a critical role in the BSH activity as catalytic nucleophile [29], and thus the putative protein would function as the BSH enzyme. Amino acid sequence comparison analyses using the standard BLASTP protein– protein BLAST search revealed that LagBSH exhibited high similarity (~93.99% amino acid sequence homology) to known BSHs, especially BSHs from L. johnsonii strain 100-100 (93.99%), strain NCC533 (93.67%), and strain PF01 (93.35%) (Supplementary Table S1). LagBSH exhibited significantly lower similarity to LgBSH form L. gasseri FR4 (39.94%) [20], despite the fact that both BSH enzymes are commonly derived from L. gasseri. The phy- logenetic analysis demonstrated that the cholylglycine hydrolase family proteins were subdivided into several groups (Figure4), and we found that LagBSH was classified into the L. johnsonii BSH subgroup (Figure4). LgBSH was categorized into the L. aci- dophilus/johnsonii BSH subgroup, indicating that LagBSH are phylogenetically distinct from LgBSH. These sequence, structural, and phylogenetic analyses further suggested that LagBSH would have BSH activity as well as known BSHs from other Lactobacillus species. Microorganisms 2021, 9, x FOR PEER REVIEW 7 of 12 Microorganisms 2021, 9, 1011 7 of 12

FigureFigure 3. ( A3.) ( PhysicalA) Physical map of themap predicted of thebsh predictedgene on the bsh genome gene sequence on the ofgenomeLactobacillus sequence gasseri of Lactobacillus gasseri T JCM1131JCM1131(accessionT (accession number number CP000413). CP000413). The scale bar The indicates scale abar 1 kb indi lengthcates of nucleotide.a 1 kb length A of nucleotide. A pu- putativetative bsh gene gene (lagBSH (lagBSH) and) itsand surrounding its surrounding ORFs are represented ORFs are by represen filled andted open by symbols, filled and open symbols, re- respectively. Brief annotation and protein ID were provided. MegG, demethylmenaquinone methyl- transferase;spectively. MP, Brief membrane annotation protein. (B and) Multiple protein alignment ID we of aminore provided. acid sequences MegG, of BSHs. demethylmenaquinone Amino methyl- acidtransferase; sequence of LagBSHMP, membrane was aligned andprotein. compared (B) with Multiple known BSHs alignment from Lactobacillus of aminospecies. acid sequences of BSHs. TheAmino black andacid gray sequence shading indicates of LagBSH identical was and alig similarned amino and acidcompared residues, respectively.with known The BSHs from Lactobacillus conservedspecies. residuesThe black (Cys, Arg,and Asp, gray Asn, shad and Arg)ing relevantindicates to the identical predicted activeand similar site are indicated amino acid residues, respec- bytively. blackasterisks. The conserved Abbreviations: residues LaBSH (AAV42923)(Cys, Arg, from Asp,Lactobacillus Asn, and acidophilus Arg) relevantNCFM; LgBSH to the predicted active site Lactobacillus gasseri Lactobacillus johnsonii (WP_020806888)are indicated from by black asterisks.FR4; Abbreviations: LjBSH (AAC34381) fromLaBSH (AAV42923)100-100; from Lactobacillus acidophilus LsBSH (JX120368) from Lactobacillus salivarius B-30514. NCFM; LgBSH (WP_020806888) from Lactobacillus gasseri FR4; LjBSH (AAC34381) from Lactobacillus johnsonii 100-100; LsBSH (JX120368) from Lactobacillus salivarius B-30514.

Microorganisms 20212021,, 99,, 1011x FOR PEER REVIEW 88 of of 12

Figure 4. PhylogeneticPhylogenetic analysis analysis of of LagBSH LagBSH with with cholylglycine cholylglycine hy hydrolasedrolase family family proteins. proteins. The Thephylogenetic phylogenetic tree was tree con- was constructedstructed with with MEGA MEGA X software X software using using the the neighb neighbor-joiningor-joining method method (1000 (1000 bootstrap bootstrap replications) replications) [39] [39].. Bootstrap Bootstrap values greater thanthan 50%50% areare shownshown by by circle circle symbols symbols whose whose size size correlates correlates with with the the bootstrap bootstrap values. values. CpBSH, CpBSH, BSH BSH from fromClostridium Clostrid- ium perfringens 13, was used as an outgroup. perfringens 13, was used as an outgroup.

3.3. Heterologous Expression of the Putative BSH Gene in E. coli To obtain the recombinant protein, lagBSHlagBSH gene was commercially synthesized with codon optimization optimization for for heterologous heterologous expression expression in inEscherichiaEscherichia coli coli andand subcloned subcloned into into the NdeIthe NdeI and andEcoRI EcoRI sites sitesof pET2 of pET28-b8-b expression expression vector. vector. The gene The was gene overexpressed was overexpressed in E. coli in E.BL21 coli (DE3)BL21 Champion (DE3) ChampionTM21 andTM recombinant21 and recombinant protein was protein purified was purifiedby Ni-affinity by Ni-affinity chroma- tography.chromatography. Based on Based sodium on sodium dodecyl dodecyl sulfate sulfatepolyacrylamide polyacrylamide gel electrophoresis gel electrophoresis (SDS- (SDS-PAGE)PAGE) analysis, analysis, the molecular the molecular weight weight of purified of purified His His6-LagBSH6-LagBSH protein protein was was approxi- approx- matelyimately 35.0 35.0 kDa kDa in in size size (Supplementary (Supplementary Figure Figure S2), S2), which which is nearly is nearly identical identical with with the the- the oreticaltheoretical molecular molecular weight weight based based on onits amino its amino acid acid sequence. sequence.

3.4. Bile Salt Hydrolase Assay The bile saltsalt hydrolyzinghydrolyzing activityactivity of of the the purified purified LagBSH LagBSH was was determined determined by by detecting detect- ingthe releasedthe released glycine glycine or taurine or taurine from from conjugated conjugated bile bile salts salts as described as described previously previously [25]. [25]. We Weselected selected five five major major mammalian mammalian conjugated conjugated bile bile salts salts (TCA, (TCA, TDCA, TDCA, TCDCA, TCDCA, GCA,GCA, andand GDCA) as substrates. The recombinant LagBSH clearly exhibited significant significant BSH activity toward all substrates tested. In particular, as shown in Figure5A, LagBSH showed its high toward all substrates tested. In particular, as shown in Figure 5A, LagBSH showed its high activity toward TCA and TCDCA. The BSH activities toward the other three substrates activity toward TCA and TCDCA. The BSH activities toward the other three substrates (TDCA, GCA, and GDCA) are relatively low, suggesting that LagBSH is a functional BSH (TDCA, GCA, and GDCA) are relatively low, suggesting that LagBSH is a functional BSH enzyme, particularly showing high specificity for taurine-conjugated bile salts (TCA and enzyme, particularly showing high specificity for taurine-conjugated bile salts (TCA and TCDCA). Such substrate specificity of LagBSH was consistent with the previous studies. TCDCA). Such substrate specificity of LagBSH was consistent with the previous studies. In fact, the previously identified BSH from L. johnsonii PF01 sharing high homology with In fact, the previously identified BSH from L. johnsonii PF01 sharing high homology with LagBSH (93.35% homology) exhibited deconjugation activity against taurine-conjugated LagBSH (93.35% homology) exhibited deconjugation activity against taurine-conjugated bile salts, but not glycine-conjugated bile salts [40], though most BSHs from lactic acid bile salts, but not glycine-conjugated bile salts [40], though most BSHs from are more likely to deconjugate glycine-conjugated bile salts rather than taurine- bacteria are more likely to deconjugate glycine-conjugated bile salts rather than taurine- conjugated bile salts [40]. LgBSH from L. gasseri FR4 showed higher BSH activity toward conjugatedglycine-conjugated bile salts bile [40]. salts LgBSH than from taurine-conjugated L. gasseri FR4 showed ones [ 20higher], indicating BSH activity that the toward sub- glycine-conjugatedstrate specificity were bile also salts quite than different taurine-co betweennjugated LagBSH ones and [20], LgBSH, indicating even that though the bothsub- strateenzymes specificity are derived were fromalso quite the samedifferent species, betweenL. gasseri LagBSH. These and enzymaticLgBSH, even characteristics though both enzymesagree well are with derived the previous from the report same that species, the substrate L. gasseri specificity. These enzymatic of BSH enzymes characteristics may be agreestrain-specific well with [41 the]. Furtherprevious structural report that and the site-directed substrate specificity mutagenesis of BSH analyses enzymes of LagBSH may be strain-specific [41]. Further structural and site-directed mutagenesis analyses of LagBSH

Microorganisms 2021, 9, x FOR PEER REVIEW 9 of 12

would perhaps lead to a better understanding of its substrate preference. In total, LagBSH has apparent BSH activity, and this functional enzyme would confer bile detoxification on the host microorganism L. gasseri JCM1131T.

3.5. Biochemical Characterization of LagBSH The optimum temperature and pH of LagBSH were determined. The purified LagBSH protein was mixed with taurocholic acid (TCA) at selected temperature (10–90 °C, in intervals of 10 °C) and pH (pH 3.0–pH 10.0, in intervals of pH 1.0) ranges. The max- imum BSH activity was observed at 37 °C (Figure 5B). We confirmed that LagBSH exhib- ited high BSH activity in wide temperature range (at 10–50 °C) and it retained above 80% of its original activity, whereas the enzyme activity significantly declined with higher tem- perature (>60 °C) (Figure 5B). In addition, the maximum BSH activity of LagBSH was ob- served at pH 6.0 (Figure 5C). LagBSH exhibited stable activity and retained approximately above 80% of their original activity at broad pH range (pH 3.0–8.0), whereas significant decreases in enzyme activities were observed at more than or equal to pH 9.0 (Figure 5C). The optimum temperature and pH of LagBSH (37 °C and pH 6.0) are highly consistent Microorganisms 2021, 9, 1011 9 of 12 with conditions of the human small intestine (around 37 °C and pH 5.0–8.0) and the growth condition of strain JCM1131T in MRS broth (pH 6.0–6.5, 37 °C) according to the website of the Japan Collection of Microorganisms (https://jcm.brc.riken.jp/en/) (accessed would perhaps lead to a better understanding of its substrate preference. In total, LagBSH hason apparent8 June 2020). BSH activity, These and biochemical this functional features enzyme wouldof LagBSH confer bile further detoxification support on our hypothesis thethat host this microorganism enzyme mayL. gassericontributeJCM1131 toT bile. detoxification of L. gasseri JCM1131T.

FigureFigure 5. 5.Bile Bile salt salt hydrolase hydrolase activity activity and biochemical and bioc characterizationhemical characterization of LagBSH. (A )of BSH LagBSH. activities (A) BSH activities werewere measured measured toward toward five human five bile human salts: glycocholicacid bile salts: (GCA),glycocholicacid glycodeoxycholic (GCA), acid (GDCA),glycodeoxycholic acid taurocholic(GDCA), acidtaurocholic (TCA), taurodeoxycholic acid (TCA), taurodeoxycholic acid (TDCA), and taurochenodeoxycholic acid (TDCA), and acid taurochenodeoxycholic (TCDCA). acid Values(TCDCA). are indicated Values as are means indicated for eight as technical means experiments for eight technical (n = 8). Error experiments bars represent (n standard= 8). Error bars represent deviation (SD). (B) Effect of temperature (10–90 ◦C) and (C) pH (pH 3.0–pH 10.0) on BSH activity standard deviation (SD). (B) Effect of temperature (10–90 °C) and (C) pH (pH 3.0–pH 10.0) on BSH toward TCA of LagBSH. Each value is expressed as means for eight technical replicates (n = 8). activity toward TCA of LagBSH. Each value is expressed as means for eight technical replicates (n Maximum activity was taken as 100%. = 8). Maximum activity was taken as 100%. 3.5. Biochemical Characterization of LagBSH The optimum temperature and pH of LagBSH were determined. The purified LagBSH protein was mixed with taurocholic acid (TCA) at selected temperature (10–90 ◦C, in inter- vals of 10 ◦C) and pH (pH 3.0–pH 10.0, in intervals of pH 1.0) ranges. The maximum BSH activity was observed at 37 ◦C (Figure5B). We confirmed that LagBSH exhibited high BSH activity in wide temperature range (at 10–50 ◦C) and it retained above 80% of its original ac- tivity, whereas the enzyme activity significantly declined with higher temperature (>60 ◦C) (Figure5B). In addition, the maximum BSH activity of LagBSH was observed at pH 6.0 (Figure5C). LagBSH exhibited stable activity and retained approximately above 80% of their original activity at broad pH range (pH 3.0–8.0), whereas significant decreases in enzyme activities were observed at more than or equal to pH 9.0 (Figure5C). The optimum temperature and pH of LagBSH (37 ◦C and pH 6.0) are highly consistent with conditions of the human small intestine (around 37 ◦C and pH 5.0–8.0) and the growth condition of Microorganisms 2021, 9, 1011 10 of 12

Microorganisms 2021, 9, x FOR PEER REVIEW strain JCM1131T in MRS broth (pH 6.0–6.5, 37 ◦C) according to the website10 of of12 the Japan

Collection of Microorganisms (https://jcm.brc.riken.jp/en/) (accessed on 8 June 2020). These biochemical features of LagBSH further support our hypothesis that this enzyme may contribute to bile detoxification of L. gasseri JCM1131T. 3.6. Transcriptional Analysis of lagBSH Gene To determine3.6. the Transcriptional regulation Analysisof gene oftranscription lagBSH Gene of the lagBSH gene, reverse tran- scription polymeraseTo chain determine reaction the (RT-PCR) regulation analyses of gene transcription were conducted. of the lagBSH We foundgene, that reverse tran- the lagBSH gene scriptionwas constituently polymerase expressed chain reaction in L. (RT-PCR) gasseri JCM1131 analysesT were(Figure conducted. 6, lane 1). We In found that T addition, the lagBSHthe lagBSH gene transcriptiongene was constituently was also expressed observed in inL. this gasseri strainJCM1131 exposed(Figure to TCA6, lane 1). In addition, the lagBSH gene transcription was also observed in this strain exposed to TCA (Figure 6, lane 2) and TDCA (Figure 6, lane 3), suggesting that the exposure to TCA and (Figure6, lane 2) and TDCA (Figure6, lane 3), suggesting that the exposure to TCA and T TDCA may haveTDCA little mayeffect have on littlethe effectlagBSH on thegenelagBSH transcriptiongene transcription in strain in JCM1131 strain JCM1131. SinceT. Since bile bile salt concentrationssalt concentrations reach the reach millimolar the millimolar level in level the in human the human small small intestine intestine and itit should be should be toxic totoxic the tointestinal the intestinal bacteria bacteria [12], [ strain12], strain JCM1131 JCM1131T seemsT seems to toconstantly constantly produce produce LagBSH LagBSH enzymeenzyme to tolerate to tolerate high concentration high concentration of bile of bilesalts salts and and survive survive in inthe the gut. gut.

Figure 6.FigureRT-PCR 6. analysesRT-PCR ofanalyseslagBSH genesof lagBSH in L.gasseri genesJCM1131 in L. gasseriT. Sterilized JCM1131 waterT. Sterilized (lane N) and water genomic (lane DNA N) and of strain JCM1131genomicT (lane P) DNA were of used strain as negative JCM1131 andT (lane positive P) control,were used respectively. as negative The productsand positive of reverse control, transcription respectively. from total RNA ofThe nonsupplemented products of reverse (lane 1), transcription TCA-supplemented from to (lanetal RNA 2), and of TDCA-supplemented nonsupplemented (lane(lane 3) 1), strain TCA-supple- JCM1131T cells were usedmented as the (lane template 2), and for PCR,TDCA-suppl respectively.emented The 16S (lane rRNA 3) genestrain (378 JCM1131 bp) wasT usedcells aswere internal used standard as the template control. Lane M, molecularfor PCR, size respectively. markers (100 bpThe DNA 16S ladder,rRNA Promega,gene (378 Madison, bp) was WI, used USA). as internal standard control. Lane M, molecular size markers (100 bp DNA ladder, Promega, Madison, WI, USA). 4. Conclusions 4. Conclusions In this study, we identified that Lactobacillus gasseri JCM1131T displayed bile salt resis- tance capacity toward primary bile salts and taurine-conjugated secondary bile salt. The In this study, we identified that Lactobacillus gasseri JCM1131T displayed bile salt re- present study further demonstrated that strain JCM1131T exhibited apparent BSH activity, sistance capacityalthough toward thisprimary strain bile has beensalts considered and taurine-conjugated to be a non-BSH-producer secondary so bile far. Moreover,salt. we The present studyclarified further the demonstrated correlations between that strain bile JCM1131 salt resistanceT exhibited and BSH apparent activity in BSHL. gasseri ac- , which tivity, although thishas beenstrain rarely has investigatedbeen consider anded poorly to be understood; a non-BSH-producer indeed, only twoso far. strains Moreo- (L. gasseri FR4 ver, we clarified theand correlationsL. gasseri CNCM between I-4884 bile isolated salt resistance from chicken and and BSH carious activity tooth, in respectivelyL. gasseri, [20,32]) which has been rarelyhave been investigated reported to and show poorly both bile understood; salt resistance indeed, ability only and BSH two activity strains by (L. producing gasseri FR4 and L.their gasseri BSH CNCM enzymes I-4884 (LgBSH isolated isolated from from chicken strain FR4 and [20 ])carious among tooth,L. gasseri respec-isolates. In the T tively [20,32]) havepresent been study, reported we also to show found bo thatth BSHbile enzymesalt resistance from L. ability gasseri JCM1131 and BSH (LagBSH)activity was sig- nificantly different from LgBSH in terms of their amino acid sequence homology, substrate by producing their BSH enzymes (LgBSH isolated from strain FR4 [20]) among L. gasseri specificity, and phylogenetic position. Since strain JCM1131T is a human-derived lactic acid T isolates. In the presentbacterium study, that we exhibits also oxalate-degradationfound that BSH enzyme activity from [23] andL. gasseri increases JCM1131 in interleukin-10 (LagBSH) was significantlyproduction [22different], this study from could LgBSH further in terms expand of andtheir deepen amino the acid understanding sequence of this homology, substratebeneficial specificity, probiotic and bacterium. phylogenetic position. Since strain JCM1131T is a hu- man-derived lactic acidIn addition,bacterium we that performed exhibits the oxalate-degradation enzymatic, transcriptional, activity and [23] phylogenetic and in- char- creases in interleukin-10acterization production of LagBSH [22], isolated this study from straincould JCM1131further expandT. LagBSH and could deepen function the as BSH understanding ofenzyme this beneficial able to hydrolyze probiotic conjugatedbacterium. bile salts especially against taurocholic acid and In addition,taurochenodeoxycholic we performed the enzymatic, acid. We further transcriptional, demonstrated and that phylogenetic the lagBSH gene charac- was constantly transcribed in L. gasseri JCM1131T. Therefore, this functional enzyme would confer a terization of LagBSH isolated from strain JCM1131T. LagBSH could function as BSH en- survival advantage on strain JCM1131T within the human intestine by bile detoxification. zyme able to hydrolyze conjugated bile salts especially against taurocholic acid and tau- rochenodeoxycholic acid. We further demonstrated that the lagBSH gene was constantly transcribed in L. gasseri JCM1131T. Therefore, this functional enzyme would confer a sur- vival advantage on strain JCM1131T within the human intestine by bile detoxification. Be- cause BSH activity exert further positive effects on human health such as weight loss and cholesterol lowering [19], future studies need to examine the probiotic effects related to the BSH activity of L. gasseri JCM1131T by in vivo animal model study. Altogether, our findings provide additional insights into the probiotic function in a well-known repre- sentative of probiotic lactic acid bacterium L. gasseri JCM1131T.

Microorganisms 2021, 9, 1011 11 of 12

Because BSH activity exert further positive effects on human health such as weight loss and cholesterol lowering [19], future studies need to examine the probiotic effects related to the BSH activity of L. gasseri JCM1131T by in vivo animal model study. Altogether, our findings provide additional insights into the probiotic function in a well-known representative of probiotic lactic acid bacterium L. gasseri JCM1131T.

Supplementary Materials: The following are available online at https://www.mdpi.com/article/10 .3390/microorganisms9051011/s1, Figure S1: Structural analyses of LagBSH, Figure S2: SDS-PAGE analysis of purified LagBSH, Table S1, The BSH sequence homology among Lactobacillus species. Author Contributions: Conceptualization, H.K. and H.T.; data curation, H.K.; formal analysis, H.K. and H.T.; investigation, H.K. and H.T.; funding acquisition, H.K. and H.T.; supervision, H.T.; project administration, H.T.; writing—original draft, H.K. and H.T.; writing—review and editing, H.K., K.M., and H.T. All authors have read and agreed to the published version of the manuscript. Funding: This work was partially supported by JSPS KAKENHI (Grant numbers JP19K16633, JP19H05683 and JP19H05679), JST ERATO (Grant Number JPMJER1502), and AMED PRIME (Grant number JP18gm6010019). Institutional Review Board Statement: Not applicable. Informed Consent Statement: Not applicable. Data Availability Statement: Not applicable. Conflicts of Interest: The authors declare no conflict of interest.

References 1. Pohjavuori, E.; Viljanen, M.; Korpela, R.; Kuitunen, M.; Tiittanen, M.; Vaarala, O.; Savilahti, E. Lactobacillus GG effect in increasing IFN-gamma production in infants with cow’s milk allergy. J. Allergy Clin. Immunol. 2004, 114, 131–136. [CrossRef][PubMed] 2. Vanderhoof, J.A.; Whitney, D.B.; Antonson, D.L.; Hanner, T.L.; Lupo, J.V.; Young, R.J. Lactobacillus GG in the prevention of antibiotic-associated diarrhea in children. J. Pediatr. 1999, 135, 564–568. [CrossRef] 3. Sakamoto, I.; Igarashi, M.; Kimura, K.; Takagi, A.; Miwa, T.; Koga, Y. Suppressive effect of Lactobacillus gasseri OLL 2716 (LG21) on Helicobacter pylori infection in humans. J. Antimicrob. Chemother. 2001, 47, 709–710. [CrossRef] 4. Niedzielin, K.; Kordecki, H.; ena Birkenfeld, B. A controlled, double-blind, randomized study on the efficacy of Lactobacillus plantarum 299V in patients with irritable bowel syndrome. Eur. J. Gastroenterol. Hepatol. 2001, 13, 1143–1147. [CrossRef] 5. Ross, R.P.; Desmond, C.; Fitzgerald, G.F.; Stanton, C. Overcoming the technological hurdles in the development of probiotic foods. J. Appl. Microbiol. 2005, 98, 1410–1417. [CrossRef][PubMed] 6. McFarland, L.V.; Evans, C.T.; Goldstein, E.J.C. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review and Meta-Analysis. Front. Med. 2018, 5, 124. [CrossRef] 7. Ryan, K.A.; Daly, P.; Li, Y.; Hooton, C.; O’Toole, P.W. Strain-specific inhibition of Helicobacter pylori by Lactobacillus salivarius and other lactobacilli. J. Antimicrob. Chemother. 2008, 61, 831–834. [CrossRef] 8. Bhushan, B.; Sakhare, S.M.; Narayan, K.S.; Kumari, M.; Mishra, V.; Dicks, L.M.T. Characterization of Riboflavin-Producing Strains of Lactobacillus plantarum as Potential Probiotic Candidate through in vitro Assessment and Principal Component Analysis. Probiotics Antimicrob. Proteins 2020, 13, 453–467. [CrossRef][PubMed] 9. Wine, E.; Gareau, M.G.; Johnson-Henry, K.; Sherman, P.M. Strain-specific probiotic (Lactobacillus helveticus) inhibition of Campy- lobacter jejuni invasion of human intestinal epithelial cells. FEMS Microbiol. Lett. 2009, 300, 146–152. [CrossRef] 10. Selle, K.; Klaenhammer, T.R. Genomic and phenotypic evidence for probiotic influences of Lactobacillus gasseri on human health. FEMS Microbiol. Rev. 2013, 37, 915–935. [CrossRef][PubMed] 11. Urdaneta, V.; Casadesús, J. Interactions between bacteria and bile salts in the gastrointestinal and hepatobiliary tracts. Front. Med. 2017, 4, 163. [CrossRef] 12. Foley, M.H.; O’Flaherty, S.; Barrangou, R.; Theriot, C.M. Bile salt hydrolases: Gatekeepers of bile acid metabolism and host- microbiome crosstalk in the gastrointestinal tract. PLoS Pathog. 2019, 15, e1007581. [CrossRef] 13. Bustos, A.Y.; de Valdez, G.F.; Fadda, S.; Taranto, M.P. New insights into bacterial bile resistance mechanisms: The role of bile salt hydrolase and its impact on human health. Food Res. Int. 2018, 112, 250–262. [CrossRef][PubMed] 14. O’Flaherty, S.; Crawley, A.B.; Theriot, C.M.; Barrangou, R. The Lactobacillus Bile Salt Hydrolase Repertoire Reveals Niche-Specific Adaptation. mSphere 2018, 3, e00140-18. [CrossRef][PubMed] 15. Ren, J.; Sun, K.; Wu, Z.; Yao, J.; Guo, B. All 4 Bile salt hydrolase proteins are responsible for the hydrolysis activity in Lactobacillus plantarum ST-III. J. Food Sci. 2011, 76, M622–M628. [CrossRef] 16. Öztürk, M.; Aydin, Y.; Kiliçsaymaz, Z.; Önal, C.; Ba, N. Molecular cloning, characterization, and comparison of four bile salt hydrolase-related enzymes from Lactobacillus plantarum GD2 of human origin. Food Biotechnol. 2018, 32, 191–205. [CrossRef] Microorganisms 2021, 9, 1011 12 of 12

17. Lambert, J.M.; Bongers, R.S.; de Vos, W.M.; Kleerebezem, M. Functional analysis of four bile salt hydrolase and penicillin acylase family members in Lactobacillus plantarum WCFS1. Appl. Environ. Microbiol. 2008, 74, 4719–4726. [CrossRef] 18. Ruiz, L.; Margolles, A.; Sanchez, B. Bile resistance mechanisms in Lactobacillus and Bifidobacterium. Front. Microbiol. 2013, 4, 396. [CrossRef][PubMed] 19. Begley, M.; Hill, C.; Gahan, C.G. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 2006, 72, 1729–1738. [CrossRef] 20. Rani, R.P.; Anandharaj, M.; Ravindran, A.D. Characterization of Bile Salt Hydrolase from Lactobacillus gasseri FR4 and Demonstra- tion of Its Substrate Specificity and Inhibitory Mechanism Using Molecular Docking Analysis. Front. Microbiol. 2017, 8, 1004. [CrossRef] 21. Azcarate-Peril, M.A.; Altermann, E.; Goh, Y.J.; Tallon, R.; Sanozky-Dawes, R.B.; Pfeiler, E.A.; O’Flaherty, S.; Buck, B.L.; Dobson, A.; Duong, T. Analysis of the genome sequence of Lactobacillus gasseri ATCC 33323 reveals the molecular basis of an autochthonous intestinal organism. Appl. Environ. Microbiol. 2008, 74, 4610–4625. [CrossRef] 22. Stoeker, L.; Nordone, S.; Gunderson, S.; Zhang, L.; Kajikawa, A.; LaVoy, A.; Miller, M.; Klaenhammer, T.R.; Dean, G.A. Assessment of Lactobacillus gasseri as a candidate oral vaccine vector. Clin. Vaccine Immunol. 2011, 18, 1834–1844. [CrossRef][PubMed] 23. Lewanika, T.R.; Reid, S.J.; Abratt, V.R.; Macfarlane, G.T.; Macfarlane, S. Lactobacillus gasseri Gasser AM63(T) degrades oxalate in a multistage continuous culture simulator of the human colonic microbiota. FEMS Microbiol. Ecol. 2007, 61, 110–120. [CrossRef] 24. Kusada, H.; Tamaki, H.; Kamagata, Y.; Hanada, S.; Kimura, N. A novel quorum-quenching N-acylhomoserine lactone acylase from Acidovorax sp. strain MR-S7 mediates antibiotic resistance. Appl. Environ. Microbiol. 2017, 83, e00080-17. [CrossRef] 25. Allain, T.; Chaouch, S.; Thomas, M.; Vallee, I.; Buret, A.G.; Langella, P.; Grellier, P.; Polack, B.; Bermudez-Humaran, L.G.; Florent, I. Bile-Salt-Hydrolases from the Probiotic Strain Lactobacillus johnsonii La1 Mediate Anti-giardial Activity in Vitro and in Vivo. Front. Microbiol. 2018, 8, 2707. [CrossRef] 26. Gu, X.C.; Luo, X.G.; Wang, C.X.; Ma, D.Y.; Wang, Y.; He, Y.Y.; Li, W.; Zhou, H.; Zhang, T.C. Cloning and analysis of bile salt hydrolase genes from Lactobacillus plantarum CGMCC No. 8198. Biotechnol. Lett. 2014, 36, 975–983. [CrossRef] 27. Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F.T.; de Beer, T.A.P.; Rempfer, C.; Bordoli, L.; et al. SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 2018, 46, W296–W303. [CrossRef] 28. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualiza- tion system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [CrossRef][PubMed] 29. Rossocha, M.; Schultz-Heienbrok, R.; von Moeller, H.; Coleman, J.P.; Saenger, W. Conjugated bile acid hydrolase is a tetrameric N-terminal thiol hydrolase with specific recognition of its cholyl but not of its tauryl product. Biochemistry 2005, 44, 5739–5748. [CrossRef] 30. Sue, D.; Boor, K.J.; Wiedmann, M. σB-dependent expression patterns of compatible solute transporter genes opuCA and lmo1421 and the conjugated bile salt hydrolase gene bsh in Listeria monocytogenes. Microbiology 2003, 149, 3247–3256. [CrossRef][PubMed] 31. Christiaens, H.; Leer, R.J.; Pouwels, P.H.; Verstraete, W. Cloning and expression of a conjugated bile acid hydrolase gene from Lactobacillus plantarum by using a direct plate assay. Appl. Environ. Microbiol. 1992, 58, 3792–3798. [CrossRef] 32. Allain, T.; Chaouch, S.; Thomas, M.; Travers, M.A.; Valle, I.; Langella, P.; Grellier, P.; Polack, B.; Florent, I.; Bermudez-Humaran, L.G. Bile Salt Hydrolase Activities: A Novel Target to Screen Anti-Giardia Lactobacilli? Front. Microbiol. 2018, 9, 89. [CrossRef] 33. De Smet, I.; Van Hoorde, L.; Woestyne, M.V.; Christiaens, H.; Verstraete, W. Significance of bile salt hydrolytic activities of lactobacilli. J. Appl. Bacteriol. 1995, 79, 292–301. [CrossRef][PubMed] 34. Šuškovi´c,J.; Kos, B.; Matoši´c,S.; Besendorfer, V. The effect of bile salts on survival and morphology of a potential probiotic strain Lactobacillus acidophilus M92. World J. Microbiol. Biotechnol. 2000, 16, 673–678. [CrossRef] 35. Prasad, J.; Gill, H.; Smart, J.; Gopal, P.K. Selection and characterisation of Lactobacillus and Bifidobacterium strains for use as probiotics. Int. Dairy J. 1998, 8, 993–1002. [CrossRef] 36. Strahinic, I.; Busarcevic, M.; Pavlica, D.; Milasin, J.; Golic, N.; Topisirovic, L. Molecular and biochemical characterizations of human oral lactobacilli as putative probiotic candidates. Oral Microbiol. Immunol. 2007, 22, 111–117. [CrossRef][PubMed] 37. Oh, N.S.; Joung, J.Y.; Lee, J.Y.; Kim, Y. Probiotic and anti-inflammatory potential of Lactobacillus rhamnosus 4B15 and Lactobacillus gasseri 4M13 isolated from infant feces. PLoS ONE 2018, 13, e0192021. [CrossRef][PubMed] 38. Rani, R.P.; Anandharaj, M.; Hema, S.; Deepika, R.; Ravindran, A.D. Purification of Antilisterial Peptide (Subtilosin A) from Novel Bacillus tequilensis FR9 and Demonstrate Their Pathogen Invasion Protection Ability Using Human Carcinoma Cell Line. Front. Microbiol. 2016, 7, 1910. [CrossRef] 39. Kumar, S.; Stecher, G.; Li, M.; Knyaz, C.; Tamura, K. MEGA X: Molecular Evolutionary Genetics Analysis across Computing Platforms. Mol. Biol. Evol. 2018, 35, 1547–1549. [CrossRef][PubMed] 40. Oh, H.-K.; Lee, J.Y.; Lim, S.J.; Kim, M.J.; Kim, G.-B.; Kim, J.-H.; Hong, S.-K.; Kang, D.-K. Molecular cloning and characterization of a bile salt hydrolase from Lactobacillus acidophilus PF01. J. Microbiol. Biotechnol. 2008, 18, 449–456. [PubMed] 41. Dong, Z.; Lee, B.H. Bile salt hydrolases: Structure and function, substrate preference, and inhibitor development. Protein Sci. 2018, 27, 1742–1754. [CrossRef][PubMed]