Brief Article Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Feb 7, 2012; 18(5): 479-488
Published online Feb 7, 2012. doi: 10.3748/wjg.v18.i5.479
Safety assessment of Bifidobacterium longum JDM301 based on complete genome sequences
Yan-Xia Wei, Zhuo-Yang Zhang, Chang Liu, Xiao-Kui Guo, Department of Medical Microbiology and Parasitology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
Pradeep K Malakar, Institute of Food Research, Norwich Research Park, NR4 7UA, Norwich, United Kingdom
Author contributions: Guo XK and Wei YX designed the study; Wei YX and Liu C analyzed the data; Wei YX and Zhang ZY carried out the experiments; Wei YX wrote the paper; and Malakar PK edited the paper.
Supported by The National Key Program for Infectious Diseases of China, No. 2008ZX10004 and 2009ZX10004; the Program of Shanghai Subject Chief Scientist, No. 09XD1402700; the Program of Shanghai Research and Development, No. 10JC1408200; and a China Partnering Award from the Biotechnology and Biological Sciences Research Council, United Kingdom
Correspondence to: Xiao-Kui Guo, Professor, Department of Medical Microbiology and Parasitology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. microbiology@sjtu.edu.cn
Telephone: +86-21-64453285 Fax: +86-21-64453285
Received: May 25, 2011
Revised: July 31, 2011
Accepted: August 7, 2011
Published online: February 7, 2012

Abstract

AIM: To assess the safety of Bifidobacterium longum (B. longum) JDM301 based on complete genome sequences.

METHODS: The complete genome sequences of JDM301 were determined using the GS 20 system. Putative virulence factors, putative antibiotic resistance genes and genes encoding enzymes responsible for harmful metabolites were identified by blast with virulence factors database, antibiotic resistance genes database and genes associated with harmful metabolites in previous reports. Minimum inhibitory concentration of 16 common antimicrobial agents was evaluated by E-test.

RESULTS: JDM301 was shown to contain 36 genes associated with antibiotic resistance, 5 enzymes related to harmful metabolites and 162 nonspecific virulence factors mainly associated with transcriptional regulation, adhesion, sugar and amino acid transport. B. longum JDM301 was intrinsically resistant to ciprofloxacin, amikacin, gentamicin and streptomycin and susceptible to vancomycin, amoxicillin, cephalothin, chloramphenicol, erythromycin, ampicillin, cefotaxime, rifampicin, imipenem and trimethoprim-sulphamethoxazol. JDM301 was moderately resistant to bacitracin, while an earlier study showed that bifidobacteria were susceptible to this antibiotic. A tetracycline resistance gene with the risk of transfer was found in JDM301, which needs to be experimentally validated.

CONCLUSION: The safety assessment of JDM301 using information derived from complete bacterial genome will contribute to a wider and deeper insight into the safety of probiotic bacteria.

Key Words: Bifidobacterium longum, Safety assessment, Genome, Antibiotic resistance, Harmful metabolite, Virulence factor



INTRODUCTION

Bifidobacteria spp are high-GC content, Gram-positive bacteria which belong to the Actinobacteria branch and these species naturally colonize the gastrointestinal tract (GIT) of mammals, birds and insects[1]. Scientists have determined the major probiotic properties of Bifidobacteria spp isolated from the human intestine and these properties include the strengthening of the intestinal barrier, modulation of the immune response and antagonism of pathogens[2].

Bifidobacterium spp has been reported to possess various glycosyl hydrolases (GH) and these hydrolases metabolize plant- or milk-derived oligosaccharides including nondigestible ones such as galacto-oligosaccharides (GOS) and fructo-oligosaccharides (FOS)[3,4]. The capability to utilize nondigestible oligosaccharides confers a competitive advantage to Bifidobacterium spp in the human gut.

Bifidobacterium longum (B. longum) and various other bifidobacteria strains are often added to probiotic products in combination with other lactic acid bacteria (LAB). Through their long and safe history of application, LAB have acquired the status of “Generally Regarded As Safe” (GRAS), but the safety of Bifidobacteria and other LAB strains selected for probiotics still need to be carefully evaluated. The key safety aspects for use of bifidobacteria and other LAB strains in probiotics include antibiotic resistance, production of harmful metabolites and the potential for virulence. Antibiotic resistance in potential probiotic strains is not considered a risk factor unless resistance is transferred to pathogens or it renders the probiotic untreatable in very rare cases of infection[5]. Biogenic amines, D-lactic acid, azoreductases and nitroreductases produced by bifidobacteria and other LAB strains are potential health hazards[6,7] and the safety of some of these compounds have been evaluated[8]. Virulence genes may be present in commensal bacteria and absence of virulence in these bacteria needs to be proved on a case by case basis.

Probiotic agents are widely used in the food and drug industry and as more commercial probiotic products are being introduced in the market, it is timely to reassess the safety of these probiotic products using the latest technology. Information from the complete genome sequences of Bifidobacteria will provide additional insight into the genetic basis for their safety. We sequenced the complete genome sequences of B. longum JDM301 (GenBank accession number CP002010), a commercial strain used widely in China with several probiotic functions, for this purpose[9].

The aim of the present work was to assess the safety of B. longum JDM301 based on complete genome sequences. The criteria used were the potential to transfer antibiotic resistance to pathogens, the potential for production of harmful metabolites and the potential for virulence.

MATERIALS AND METHODS
Bacterial strains and growth conditions

JDM301 was isolated from commercial probiotic product and identified using a sequence analysis of its 16S rRNA gene. De Man-Rogosa-Sharpe (MRS) broth (Difco) supplemented with 0.05% L-cysteine·HCl (Sigma) was used for cultivating JDM301. Cultures were incubated at 37  °C under anaerobic conditions.

Genome sequencing and assembly

We determined the complete genome sequence of JDM301 at the Chinese National Human Genome Center in Shanghai using the GS 20 system (454 Life Science Corporation, Branford, Connecticut). A total of 192  888 reads with an average length of 210 bps were assembled into 112 contigs by the 454 assembly tool. The order of most large contigs, which were larger than 500 bp, was determined through the basic local alignment search tool (BLAST) analysis with the reference strain B. longum ATCC15697 (GenBank accession number CP001095) and the others were arranged by multiplex polymerase chain reaction (PCR). Gap closure was carried out by sequencing gap-spanning PCR products or clones using ABI 3730 xl DNA sequencers. Primer design and sequence assembly were performed by the Phred/Phrap/Consed software package[9]. The locations of low-quality sequences in genome were verified by directly resequencing the PCR products spanning the low-quality sequences using the ABI 3730 xl DNA sequencers.

Statistical analysis

The genome sequences of Bifidobacteria except JDM301 were retrieved from GenBank at the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/)[10]. Potential open reading frames (ORF) were identified using Glimmer[11] and ZCURVE[12] 1.0 using default settings. Clusters of orthologous group (COG) functional categories were used for functional classification of all genes in the genome sequences of JDM301 and the COGs. A BLAST analysis of the translations with GenBank’s nonredundant database was performed, which was followed by manual curation. The best matches were chosen for preliminary product assignments. Insertion sequences (IS) elements, prophage sequences and clustered regularly interspaced short palindromic repeats (CRISPR) were identified by IS finder (http://www-is.biotoul.fr/is.html), Prophage Finder[13] and CRISPRFinder (http://crispr.u-psud.fr/crispr/)[14] respectively. Putative orthologues were determined by Omics Explorer (http://omics.biosino.org:14000/kweb/about.jsp) using default values. Ribosomal RNA genes were detected on the basis of BLASTN searches and transfer RNA genes were identified using tRNAscan-SE[15]. The atlas of genome was drawn using GenomeViz1.1[16]. Putative virulence factors and putative antibiotic resistance genes were identified by blast with virulence factors database (VFDB) (http://www.mgc.ac.cn/VFs/main.htm)[17] and antibiotic resistance genes database (ARDB) (http://ardb.cbcb.umd.edu/)[18] respectively.

Antibiotic susceptibility

Minimum inhibitory concentration (MIC) of 16 common antimicrobial agents was evaluated by E-test (AB Biodisk, Solna, Sweden) including amoxicillin (0.016-256 mg/L), amikacin (0.016-256 mg/L), ampicillin (0.016-256 mg/L), bacitracin (0.016-256 mg/L), cephalothin (0.016-256 mg/L), ciprofloxacin (0.002-32 mg/L), cefotaxime (0.016-256 mg/L), chloramphenicol (0.016-256 mg/L), erythromycin (0.016-256 mg/L), gentamicin (0.016-256 mg/L), imipenem (0.002-32 mg/L), rifampicin (0.016-256 mg/L), streptomycin (0.016-256 mg/L), tetracycline (0.016-256 mg/L), trimethoprim-sulphamethoxazol (0.002-32 mg/L), and vancomycin (0.016-256 mg/L). Tests were done with MRS agar supplemented with 0.05% L-cysteine·HCl (Sigma) and were conducted in triplicate for each antibiotics. Cultures sub-inoculated into the MRS agar supplemented with 0.05% L-cysteine·HCl were incubated anaerobically at 37  °C for 24 h.

RESULTS
Comparative genomic analysis of Bifidobacteria

The predicted proteins of B. longum JDM301 were functionally categorized. The functional distribution of genes assigned to clusters of orthologous groups of proteins was relatively similar to the other Bifidobacteria, e.g., B. longum and B. adolescentis in the GIT and B. dentium in the oral cavity[3,4,19]. The top four functional categories in B. longum JDM301, namely, carbohydrate transport and metabolism, amino acid transport and metabolism, were identical with other Bifidobacteria[20].

Putative orthologues among B. longum strains were determined in a comparative study (Figure 1). Overall, 1265 proteins were conserved in all four B. longum strains (B. longum JDM301, B. longum NCC2705, B. longum DJO10A and B. longum ATCC15697). These proteins represent the “core” genome of B. longum, whereas 219 proteins are unique to B. longum JDM301. The most common functional distributions of the core proteins were these involved in housekeeping functions including amino acid transport and metabolism, translation, ribosomal structure and biogenesis, carbohydrate transport and metabolism and DNA replication, recombination and repair. Twenty-one percent of the core proteins were dedicated to carbohydrate and amino acid transport and metabolism, indicating the important roles of these proteins in Bifidobacteria.

Figure 1
Figure 1 Functional distribution of Bifidobacterium longum core proteins. A total of 1265 proteins were conserved in all four Bifidobacterium longum (B. longum) strains (B. longum JDM301, B. longum NCC2705, B. longum DJO10A and B. longum ATCC15697), representing the “core” genome of B. longum.
Stability of the genome of B. longum JDM301

Horizontal gene transfer (HGT) events are responsible for introduction of alien genes, which may reinforce the adaptation of bacteria in their specific niches. Genes on plasmids, bacteriophages, genomic islands and IS are sensitive to HGT[21]. Twelve phage-related fragments were identified in the genome of B. longum JDM301[9], but no complete prophages were found. The JDM301 chromosome also possesses 15 complete or disrupted IS elements[9]. The number of IS element in JDM301 is relatively smaller than the other sequenced B. longum spp[3,4]. Another set of genes disseminated by HGT in Bifidobacteria is the CRISPR-related system. No CRISPR was discovered in the genome.

One complete type II restriction-modification (R-M) system and one type III R-M system were present in the genome of JDM301. A complete and incomplete type I R-M system was also identified in this genome. Two complete type II R-M systems and one type I R-M system were present in the genome of B. longum NCC2705, while one complete type II R-M system and type I R-M system were found in B. longum DJO10A.

Antibiotic resistance determinants

The antibiotic resistance genes in JDM301 were identified using ARDB (E < 1e-2, coverage > 70%)[18]. Homologs of the antibiotic resistance determinants for vancomycin, methicillin, tetracycline, chloramphenicol and trimethoprim were found in the genome of JDM301 (Table 1) and 6 putative resistance genes for vancomycin. B. longum JDM301 also possessed 5 putative bacitracin efflux pumps, 5 homologs of macrolide efflux proteins. Additionally, 7 putative multidrug resistance efflux pumps belonging to an ATP-binding cassette (ABC)-type transport system, a major facilitator superfamily transporter and resistance-nodulation-cell division (RND) family were found in the genome. The genome of B. longum JDM301 also contains 4 tetracycline resistance genes encoding for TetV, TetW, TetPB and TetQ. The gene for TetW shows a strong difference in G + C content (53.0%) compared to the average value of B. longum JDM301 (59.8%) genome and it is flanked by genes encoding for integrases, indicating that this region may have been acquired by HGT.

Table 1 Putative antibiotic resistance genes identified in the genome of Bifidobacterium longum JDM301.
AntibioticsAntibiotic resistance genesProduct name
BacitracinBLJ_1636ABC transporter-related protein
BLJ_0984ABC transporter-related protein
BLJ_0923ABC transporter-related protein
BLJ_1055Undecaprenyl pyrophosphate phosphatase
BLJ_1119Bacitracin transport ATP-binding protein bcrA
VancomycinBLJ_0853VanU
BLJ_1764Dehydrogenase VanH
BLJ_1084Sensor protein vanSB
BLJ_0707VanSD5
BLJ_0343Histidine kinase VanSc3
BLJ_0287D-Ala: D-Lac ligase VanD
Multiple drugsBLJ_1090ATP-binding protein
BLJ_1650Lsa
BLJ_1437LmrB
BLJ_0618Multidrug export protein MepA
BLJ_0769Efflux transporter, RND family, MFP subunit
BLJ_0181Multidrug efflux protein QacB
BLJ_1062Multidrug export protein MepA
ChloramphenicolBLJ_1672Chloramphenicol resistance protein
BLJ_1322Chloramphenicol resistance protein
ThiostreptonBLJ_0885Thiostrepton-resistance methylase
PenicillinBLJ_1301Penicillin binding protein
KasugamycinBLJ_2030S-adenosylmethionine-6-N', N'-adenosyl
(rRNA) dimethyltransferase
TetracyclineBLJ_0814Tetracycline-resistance determinant tetV
BLJ_1245TetW
BLJ_0594Tetracycline resistance protein
BLJ_1401TetQ
CarbomycinBLJ_1625Carbomycin resistance protein
SulfonamideBLJ_1629Dihydropteroate synthase
Tetracenomycin CBLJ_1624Tetracenomycin C efflux protein
TrimethoprimBLJ_1657dihydrofolate reductase
MacrolideBLJ_0925Macrolide-efflux protein
BLJ_1936Macrolide-efflux protein
BLJ_0819Macrolide-efflux protein
BLJ_0042Macrolide-efflux protein
BLJ_1154Macrolide-efflux protein variant

The antibiotic susceptibility of B. longum JDM301 to 16 antibiotics was determined by an E-test to probe the in silico analyses of the complete genome sequence. The results of the E-test are summarized in Table 2. The breakpoints for determining susceptibility were determined using accepted protocols[22-25]. B. longum JDM301 showed a high resistance to ciprofloxacin, amikacin and gentamicin, moderate resistance to streptomycin and bacitracin and were sensitive to tetracycline, vancomycin, amoxicillin, cephalothin, chloramphenicol, erythromycin, ampicillin, cefotaxime, rifampicin, imipenem and an antimicrobial compound, trimethoprim-sulphamethoxazol.

Table 2 Minimum inhibitory concentration values of 16 antibiotics for Bifidobacterium longum JDM301.
AntibioticsMinimum inhibitory concentration (mg/L)
Ciprofloxacin> 32
Amikacin> 256
Gentamicin> 256
Bacitracin26.67
Streptomycin170.67
Vancomycin0.9
Amoxicillin0.064
Cephalothin1.33
Chloramphenicol0.25
Erythromycin0.04
Ampicillin0.058
Cefotaxime0.19
Rifampicin0.074
Tetracycline8
Imipenem0.19
Trimethoprim-sulphamethoxazol1.83
Putative enzymes for harmful metabolites

Genes encoding enzymes responsible for harmful metabolites, including beta-glucosidase (GS), arylsulphatase (AS), beta-glucuronidase (GN), nitroreductase (NR), azoreductase (AR), D-lactate dehydrogenase (DLD), amino acid decarboxylase (AD) and conjugated bile salt hydrolase (CBSH) were searched for in the genome of B. longum JDM301. Two GS genes (BLJ_1280, BLJ_1540) and one CBSH gene (BLJ_0948) were found in the chromosome of B. longum JDM301. Homologs of DLD (BLJ_1306, BLJ_1436) and NR (BLJ_1980) were also discovered in the genome. Enzymes involved in putatively harmful metabolites, AR, GN, AD and AS were not found in JDM301 genome.

Putative virulence factors

Published reports of rare infections involving Lactobacilli or Bifidobacteria are available and the potential virulence of Lactobacilli or Bifidobacteria used as probiotics should be assessed[5]. Putative virulence genes of B. longum JDM301 were determined by BLAST analysis of the VFDB[17]. A total of 141 homologs of virulence factors were identified in the genome of JDM301, including 28 sugar-binding transcriptional regulators, 20 genes associated with iron, amino acid and sugar transport, 5 transposases, and 2 glutamine synthetase related to plasminogen (Plg)-binding (Table 3).

Table 3 Putative virulence factors identified in the genome of Bifidobacterium longum JDM301.
QueryIdentitySubjectPredicted functions
BLJ_108924.9VFG09342,3-dihydro-2,3-dihydroxybenzoate dehydrogenase
BLJ_183526.36VFG09342,3-dihydro-2,3-dihydroxybenzoate dehydrogenase
BLJ_032329.3VFG09342,3-dihydro-2,3-dihydroxybenzoate dehydrogenase
BLJ_147622.11VFG23786 kDa early secretory antigenic target esxA
BLJ_099232.98VFG0869AatC ATB binding protein of ABC transporter
BLJ_108034.81VFG0869AatC ATB binding protein of ABC transporter
BLJ_196837.3VFG0869AatC ATB binding protein of ABC transporter
BLJ_077037.43VFG0869AatC ATB binding protein of ABC transporter
BLJ_002635.71VFG1404ahpC
BLJ_013628.73VFG2218ATPase VirB11 homolog
BLJ_088024.18VFG1042ATP-binding protein FecE
BLJ_078747.92VFG0077ATP-dependent Clp protease proteolytic subunit
BLJ_078653.8VFG0077ATP-dependent Clp protease proteolytic subunit
BLJ_094837.66VFG2162Bile salt hydrolase
BLJ_124322.97VFG2242Conjugal transfer protein trag
BLJ_055126.54VFG1108Conserved hypothetical protein
BLJ_195129.85VFG1269Cyclolysin secretion ATP-binding protein
BLJ_192532.31VFG1269Cyclolysin secretion ATP-binding protein
BLJ_186345.5VFG0079Endopeptidase Clp ATP-binding chain C
BLJ_146556.77VFG0079Endopeptidase Clp ATP-binding chain C
BLJ_071330.12VFG0925Ferric enterobactin transport ATP-binding protein fepC
BLJ_187225.51VFG2225GDP-mannose 4,6-dehydratase
BLJ_132432.49VFG1399glnA1
BLJ_062462.11VFG1399glnA1
BLJ_183429.47VFG0313Glucose/galactose transporter
BLJ_192630.02VFG1557HlyB protein
BLJ_147756.12VFG1855Hsp60, 60K heat shock protein HtpB
BLJ_006426.21VFG1397hspX
BLJ_144440.85VFG1563Hypothetical protein
BLJ_160627.78VFG1593Hypothetical protein
BLJ_164030.81VFG1593Hypothetical protein
BLJ_001122.16VFG1604Hypothetical protein
BLJ_151326.3VFG1604Hypothetical protein
BLJ_184627.67VFG1604Hypothetical protein
BLJ_033744.25VFG1630Hypothetical protein
BLJ_033644.38VFG1630Hypothetical protein
BLJ_150023.53VFG1963Hypothetical protein Cj1435c
BLJ_116924.64VFG1390Hypothetical protein Rv0981
BLJ_070836.8VFG1390Hypothetical protein Rv0981
BLJ_080228.83VFG1824Hypothetical protein Rv3133c
BLJ_135730.46VFG1824Hypothetical protein Rv3133c
BLJ_111332.41VFG1824Hypothetical protein Rv3133c
BLJ_083532.42VFG1824Hypothetical protein Rv3133c
BLJ_085927.93VFG1206Iron(III) ABC transporter, ATP-binding protein
BLJ_034828.13VFG1206Iron(III) ABC transporter, ATP-binding protein
BLJ_053029.29VFG1206Iron(III) ABC transporter, ATP-binding protein
BLJ_201635.81VFG1206Iron(III) ABC transporter, ATP-binding protein
BLJ_187536.19VFG1627IS100 transposase; transposase ORFA
BLJ_124937.55VFG1627IS100 transposase; transposase ORFA
BLJ_125239.22VFG1627IS100 transposase; transposase ORFA
BLJ_093042.29VFG1627IS100 transposase; transposase ORFA
BLJ_196630.68VFG1485L7045
BLJ_185059.7VFG1411leuD
BLJ_037939.24VFG0320Lipopolysaccharide core biosynthesis protein (kdtB)
BLJ_154922.02VFG1817mbtA
BLJ_120425.8VFG0574Mg<up>2+</up> transport protein
BLJ_201030.62VFG0574Mg<up>2+</up> transport protein
BLJ_127028.62VFG1116N-acetylglucosamine-6-phosphate deacetylase
BLJ_183221.89VFG1109N-acetylneuraminate lyase, putative
BLJ_049025.95VFG1109N-acetylneuraminate lyase, putative
BLJ_002126.83VFG0307Neutrophil activating protein (bacterioferritin)
BLJ_188924.14VFG2227O-antigen export system permease protein
BLJ_125126.05VFG1461ORF A protein
BLJ_021430.5VFG0594Pathogenicity island encoded protein: SPI3
BLJ_015933.25VFG0594Pathogenicity island encoded protein: SPI3
BLJ_147457.32VFG1386phoP
BLJ_170325.65VFG2220Phosphoglucomutase
BLJ_049728.35VFG2362Phosphomannomutase
BLJ_113725.1VFG1983ABC-type amino-acid transporter periplasmic solute-binding protein
BLJ_050825.93VFG1983ABC-type amino-acid transporter periplasmic solute-binding protein
BLJ_145329.27VFG1983ABC-type amino-acid transporter periplasmic solute-binding protein
BLJ_040838.22VFG2059ATP-binding component of ABC transporter
BLJ_048027.04VFG2061Phosphoprotein phosphatase
BLJ_080528.09VFG1384proC
BLJ_139631.06VFG1384proC
BLJ_058426.09VFG1387purC
BLJ_177222.28VFG0480Putative amino acid permease
BLJ_053825.17VFG0480Putative amino acid permease
BLJ_132924.42VFG1965Putative aminotransferase
BLJ_002530.45VFG2301Putative carbonic anhydrase
BLJ_092223.51VFG0031Putative glycosyl transferase
BLJ_167038.88VFG1668Putative lysil-tRNA synthetase LysU
BLJ_056325VFG1498Putative periplasmic solute binding protein
BLJ_117128.48VFG0483Putative regulatory protein, deoR family
BLJ_151729.25VFG0483Putative regulatory protein, deoR family
BLJ_034437.02VFG1702Putative response regulator
BLJ_004027.91VFG1746Putative two-component response regulator
BLJ_074029.13VFG1746Putative two-component response regulator
BLJ_110524.49VFG0168Pyochelin biosynthesis protein PchD
BLJ_040925.56VFG0168Pyochelin biosynthesis protein PchD
BLJ_072041.04VFG0479Pyruvate kinase I (formerly F), fructose stimulated
BLJ_116355.32VFG1826relA
BLJ_099525.84VFG1889Response regulator GacA
BLJ_167928.89VFG1889Response regulator GacA
BLJ_108340.89VFG0473Response regulator in two-component regulatory system with BasS
BLJ_127326.57VFG1115ROK family protein
BLJ_162026.62VFG1115ROK family protein
BLJ_162231.35VFG1115ROK family protein
BLJ_179627.31VFG0526Salmonella iron transporter: fur regulated
BLJ_066229.06VFG0526Salmonella iron transporter: fur regulated
BLJ_071225.4VFG0528Salmonella iron transporter: fur regulated
BLJ_117451.39VFG1405sigA
BLJ_125841.15VFG1412sigH
BLJ_134233.11VFG2161Signal peptidase II
BLJ_090621.73VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_192322.38VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_136022.88VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_142123.24VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_061123.31VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_183623.32VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_045923.33VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_127823.43VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_199823.51VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_152223.6VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_010923.63VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_041823.69VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_011823.7VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_052023.85VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_197624.27VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_009924.31VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_160524.34VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_013224.53VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_191224.58VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_091224.69VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_031824.71VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_151524.93VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_193325VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_171825VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_199725.36VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_040025.37VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_051527.08VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_132128.21VFG2197Sugar-binding transcriptional regulator, LacI family
BLJ_123229.83VFG1028Tn21 integrase IntI1
BLJ_116043.1VFG2168Transcriptional regulator, Cro/CI family
BLJ_074728.98VFG1122Transposase ORFAB, subunit B
BLJ_118043.64VFG1398trpD
BLJ_187139.62VFG1967UDP-galactopyranose mutase
BLJ_164439VFG2361UDP-glucose 4-epimerase
BLJ_168054.49VFG2361UDP-glucose 4-epimerase
BLJ_189152.63VFG0963UDP-glucose 6-dehydrogenase
BLJ_069746.15VFG1414whiB3

Although the ability to adhere to the intestinal wall has been one of the selection criteria for probiotics and also a characteristic of commensal bacteria in the intestine, adhesion is also considered to be a significant step in the initial pathogen infections[26]. Thus, predicted proteins for adhesion of JDM301 were also included in the analysis of virulence. A total of 21 predicted proteins for adhesion were identified in JDM301 (Table 4). A large number of predicted surface and extracellular proteins were identified in JDM301, which may be involved in the bacterium-host interaction as in other LAB[27]. A total of 217 proteins with probable Sec-type signal peptides were identified by the tool, Signal P[28]. The genome of JDM301 also harbors 18 copies of extracellular solute-binding protein (SBP, pfam01547) which is predicted to bind oligosaccharides (SBP family 1) as a component of the ABC transporter complex.

Table 4 Putative genes associated with adhesion identified in the genome of Bifidobacterium longum JDM301.
Locus_tagPfam numberProduct name
BLJ_1932pfam01547Family 1 extracellular solute-binding protein
BLJ_0112pfam01547Family 1 extracellular solute-binding protein
BLJ_1284pfam01547Family 1 extracellular solute-binding protein
BLJ_1420pfam01547Family 1 extracellular solute-binding protein
BLJ_0131pfam01547Family 1 extracellular solute-binding protein
BLJ_1604pfam01547Family 1 extracellular solute-binding protein
BLJ_1686pfam01547Family 1 extracellular solute-binding protein
BLJ_1964pfam01547Family 1 extracellular solute-binding protein
BLJ_1994pfam01547Family 1 extracellular solute-binding protein
BLJ_1996pfam01547Family 1 extracellular solute-binding protein
BLJ_2001pfam01547Family 1 extracellular solute-binding protein
BLJ_0288pfam01547Family 1 extracellular solute-binding protein
BLJ_0321pfam01547Family 1 extracellular solute-binding protein
BLJ_0345pfam01547phosphate ABC transporter periplasmic phosphate-binding protein
BLJ_0414pfam01547Family 1 extracellular solute-binding protein
BLJ_0522pfam01547Family 1 extracellular solute-binding protein
BLJ_0523pfam01547Family 1 extracellular solute-binding protein
BLJ_0524pfam01547Family 1 extracellular solute-binding protein
BLJ_0012pfam07174Hypothetical protein BLJ_0012
BLJ_1801pfam05738LPXTG-motif protein cell wall anchor domain-containing protein
BLJ_0140pfam07811TadE family protein
DISCUSSION

As more probiotic strains are used in the food and drug industry, more attentions should be paid to the safety of strains used as probiotics. Thus, the safety of LAB used as probiotics need to be reassessed using the latest technology. B. longum JDM301, is a commercial probiotic strain used in many probiotic products sold in China. Analysis of the genome of JDM301 reveals several potential risk factors needing further experimental validation, including a tetracycline resistance gene (tetW) with the risk of transfer, and the genes associated with harmful metabolites.

Bifidobacteria were considered free of phage infection until prophage-like elements were identified in the genomes of B. longum NCC2705, B. longum DJO10A and B. breve UCC2003[29]. Absence of complete prophages is important for the stability of genomes and for industrial applications of probiotic bacteria[21,30]. Absence of complete prophages and scarcity of IS element may play important roles in promoting genome stability of JDM301[31]. Another set of genes disseminated by HGT in Bifidobacteria is the CRISPR-related system (CASS), which is involved in defense against phages and plasmids[32]. No CRISPR was discovered in the genome. R-M systems are diverse and widespread in nature and they are considered as barriers to HGT, e.g., in transformation and phage infection[33]. The diversity of R-M systems in B. longum JDM301 may be significant to the stability of genome and its use in industry compared with the other two B. longum strains.

B. longum JDM301 was not resistant to tetracycline as the minimum inhibitory concentration (8.0 mg/L) was not higher than the breakpoint value (8.0 mg/L)[34]. However, the MIC for B. longum strains ranges from 0.5 to 2 mg/L in a report[35]. Thus, further experiments may be needed to determine the microbiological breakpoint. The tetW (BLJ_1245) gene encodes for a ribosomal protection protein and tetW genes were responsible for acquired tetracycline resistance in human B. longum strains[36]. The rest of the tetracycline resistance genes found in B. longum JDM301 were tetV (BLJ_0814), tetQ (BLJ_1401) and tetPB (BLJ_0594). The gene tetV encodes for a tetracycline efflux pump and the genes tetQ and tetPB encode for ribosomal protection proteins. Further experiments are needed to confirm whether the tetW gene in the chromosome of B. longum JDM301 is a transferable antibiotic resistance determinant and responsible for resistance to tetracycline in human B. longum strains.

The MIC of B. longum JDM301 to bacitracin was 26.7 mg/L, which indicated a moderate resistance. A previous report[25] indicated that B. longum strains were susceptible to bacitracin. A total of 7 putative bacitracin resistance genes were identified, including 6 genes encoding for ABC transporters and 1 for an uncharacterized bacitracin resistance protein. These genes may be responsible for the resistance to bacitracin.

The resistances to ciprofloxacin, amikacin, gentamicin and streptomycin and susceptibility of JDM301 to vancomycin, amoxicillin, cephalothin, chloramphenicol, erythromycin, ampicillin, cefotaxime, rifampicin, imipenem and an antimicrobial compound, trimethoprim-sulphamethoxazol were consistent with reported findings[22-25,36]. However, there are discrepancies between the phenotype and the genotype. B. longum JDM301 was sensitive to vancomycin and chloramphenicol but the genome contained vancomycin and chloramphenicol resistance genes. Further analysis will be needed to determine this discrepancy.

Several cases of D-lactic acidosis associated with consumption of LAB in patients with short bowel syndrome were reported[37,38], implying that bacteria used as probiotics should be screened for the ability to generate D-lactate. In this study, two homologs of DLD genes were identified in the genome of JDM301. Since there were no reported cases of D-lactic acidosis caused by bifidobacteria[37-39], the activities of these homologous DLDs in bifidobacteria may be low so that the amount of lactate produced is insufficient to cause D-lactic acidosis.

Although biogenic amines (BA) play an important physiological role in mammals, a high amount of BA in the diet may have a variety of toxic effects[40]. The main BA contained in food and beverage includes histamine, tyramine, putrescine, and cadaverine, some of which are associated with toxicological characteristics of food poisoning[41]. The decarboxylase activities of histidine, tyrosine and ornithine were reported in lactobacilli and the capabilities might be strain-dependent rather than species-dependent[42]. Therefore, BA production, especially thylamine and tyramine, must be carefully evaluated for individual strains.

Bacterial enzymes, such as GN, GS, NR, AR and AS, play important roles in the metabolism of carcinogens and other toxicants in the intestine. Homologs of GS are common in sequenced Bifidobacteria genomes where GS and GN facilitate the absorption of a variety of toxicants and may contribute to the development of colon cancer. The link between Bifidobacteria and the genotoxic enzyme activities of intestinal microflora has been reported[43,44], with Bifidobacteria inhibiting the activity of some genotoxic enzymes[45]. NR activity is common in oral bacteria and it plays an important role in bacterial nitrate reduction. Although NR activities have been reported in Bifidobacteria, the activity of this enzyme is lower than the NR activity of other gut bacteria[6].

CBSH mediates microbial bile tolerance and enhances microbial survival in the intestine[46]. Metagenomic analyses demonstrated that CBSH activity is enriched in the human gut microbiome, and has the potential to greatly influence host physiology[46]. In Bifidobacterium spp. and Lactobacillus spp., CBSH activity is also common and nearly all Bifidobacteria species and strains have bile salt hydrolase activities[47]. However, bile salt hydrolase activity releases free bile acids which are harmful to the human body and may act as mutagens[48,49]. Recommendations have been made for absence of bile salt transformation capacity in bacteria added to food[50]. However, it is noteworthy that the evidence for harmful effects is inconclusive so far and bile salt deconjugation activity may play a role in reducing human serum cholesterol[51]. Given the huge CBSH pool in intestinal microflora, the CBSH activities of the small number of additional bacteria consumed as probiotics can be ignored[48].

Putative genes for Plg-binding proteins, DnaK (BLJ_0123) and glutamine synthetase (BLJ_0624 and BLJ_1324) were found in the JDM301 genome, where these proteins play a role in the interaction with human epithelial cells. The protein DnaK has been shown to be present on the surface of pathogens, such as Neisseria meningitides[52]. The glutamine synthetases BLJ_0624 and BLJ_1324 had a 62.11% and 32.49% similarity to the glutamine synthetases in Mycobacterium tuberculosis H37 Rv. In the presence of Plg activators, Plg binding to the bacterial surface is converted to plasmin, which is a broad-spectrum serine protease involved in degradation of fibrin and noncollagenous proteins of extracellular matrices and activates latent procollagenases[53]. It is believed that the capability to intervene with the Plg/plasmin system of a host is a strategy for host colonization and bacterial metastasis shared by several pathogens and commensals of the human intestinal tract[53,54]. The plasminogen-dependent proteolytic activity of B. lactis BI07 and B. longum was shown to be dose-dependent[55,56].

A homolog (BLJ_0880, 24.18% identity) of a gene encoding a component in ferric dicitrate uptake system (Fec) of Shigella flexneri serotype 2a, FecE, was identified in the genome of JDM301. As an iron uptake system, Fec is critical for bacterial survival and plays an important role in bacterial virulence[57]. In addition, BLJ_1105 and BLJ_0409 proteins associated with iron acquisition in JDM301 were 24.49% and 25.56% similar to pyochelin biosynthesis protein in Pseudomonas aeruginosa, and BLJ_0712, BLJ_1796 and BLJ_0662 proteins were 25.4%, 27.31 and 29.06% similar to iron transporters of Salmonella enterica.

The human pathogen, Helicobacter pylori, produces a neutrophil activating protein (NAP) which activate human leukocytes and induces an inflammation, which facilitates the growth of the pathogen[58]. A homolog (BLJ_0021; 26.83% identity) of the gene encoding a NAP was identified in the genome of JDM301.

In JDM301, BLJ_0012 encodes a protein harboring fibronectin-binding motif (Pfam number 07174) that allows mycobacteria to bind to fibronectin in the extracellular matrix and may mediate the adhesion of JDM301 to its host[59]. A potential protein for Bifidobacteria adhesion to intestinal cells is the putative LPXTG-motif protein with collagen binding motifs (Cna_B, pfam05738) encoded by BLJ_1801, which shows a 34% identity to a predicted fimbrial subunit in the genome of B. dentium Bd1. This protein may be involved in the recognition of and adhesion to mucosal epithelial cell surfaces[19]. Its homologous proteins were also identified in the genome sequences of both B. longum NCC2705 and B. longum DJO10A genomes[3,60]. B. longum subsp. infantis 15697, B. longum NCC2705 and B. adolescentis contains 21, 10 and 11 copies of extracellular solute-binding protein, respectively[3,4]. Comparably, the SBP family 1 proteins are more abundant in JDM301 than the three other Bifidobacteria strains due to the genome size.

Finally, JDM301 encodes a number of proteases and peptidase that may contribute to virulence owing their ability to degrade host proteins for bacterial nutrition sources[61]. However, not all the genes associated with virulence have been known until now. Thus, despite the evaluation based on the whole genome sequences, it is recommended that the rat endocarditis and the immunocompromised mouse model should be used for in vivo assessment of safety for the low pathogenicity of LAB[48].

Recently, there has been more interest in using probiotic products to promote health and treat diseases. Probiotics have been investigated in clinical trials, such as treatment for diarrhea, D-lactic acidosis, necrotizing enterocolitis, inflammatory bowel disease and so on[39,62-64]. The mechanisms by which probiotics exert their effects are still obscure, which may include modification of gut pH, antagonism of pathogens, modulation of immunity as well as supplements of some nutrients[65]. However, safety issues of probiotics have been discussed in many reports[5,48]. There are reported cases of infections associated with probiotic strains[5]. Although the strain is safe based on phenotype, the information derived from complete bacterial genome sequences reveals some putative unfavorable genes, such as genes encoding for Plg-binding proteins, proteases and genes associated with production of D-lactate. In addition, patients are generally more susceptive to infection and harmful metabolites, such as D-lactate than healthy persons. Thus, the biosafety of probiotics, especially strains used in therapy, must be assessed more carefully and comprehensively.

In conclusion, this study compared the genome of JDM301 with other Bifidobacteria and assessed the genomic stability, the potential for antibiotic resistance, the potential for virulence and the potential production of harmful metabolites of this strain. The core genome of B. longum is composed of 1265 genes, and 219 genes are unique in JDM301. Our data showed putative virulence genes in the genomes of JDM301 as well as putative genes associated with production of harmful metabolites. In addition, a potentially transferable antibiotic resistance gene was detected in the chromosome of JDM301, which needs to be experimentally validated. This assessment provides information on potential risk factors, which should be further evaluated experimentally, e.g., in vivo assessment using animal models.

ACKNOWLEDGMENTS

The authors thank Dr. Hua-Jun Zheng from Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai for kindly providing us assistance in data analysis.

COMMENTS
Background

Bifidobacterium longum JDM301 is a commercial strain used widely in China with several probiotic functions. Recently, there has been more interest in using probiotic products to promote health and treat diseases. As model probiotic bacteria, Bifidobacteria are often added to probiotic products in combination with other lactic acid bacteria. The biosafety of probiotic bacteria is attracting more attentions with its enlarged applications. As more commercial probiotic products are being introduced in the market, it is necessary to reassess the safety of these probiotic products using the latest technology.

Research frontiers

With a long and safe history of application, lactic acid bacteria have acquired the status of “Generally Regarded As Safe”. However, published reports of rare infections involving Lactobacilli or Bifidobacteria are available. The strains selected as probiotics are needed to be assessed carefully and comprehensively. This study may contribute to a better biosafety assessment of probiotic bacteria.

Innovations and breakthroughs

This is the first study to assess the biosafety of probiotic bacteria based on complete genome sequences. Through bioinformatics analysis of the genome sequences, the authors found that although the strain was safe based on phenotype, the information derived from complete bacterial genome sequences revealed some putative unfavourable genes that should be paid attention to.

Applications

The study provides a comprehensive assessment on potential risk factors of a probiotic strain based on complete genome sequences. The information related to biosafety derived from the genome of JDM301 will contribute to a wider and deeper insight into the safety of probiotic bacteria.

Peer review

This is a very nice and comprehensive study assessing the genomic stability, potential of antibiotic resistance, virulence and production of harmful metabolites. This adds valuable information to current knowledge about probiotics.

Footnotes

Peer reviewer: Tauseef Ali, MD, Assistant Professor, Section of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, 920 SL Young Blvd, Oklahoma City, OK 73104, United States

S- Editor Tian L L- Editor Ma JY E- Editor Zhang DN

References
1.  Ventura M, Canchaya C, Tauch A, Chandra G, Fitzgerald GF, Chater KF, van Sinderen D. Genomics of Actinobacteria: tracing the evolutionary history of an ancient phylum. Microbiol Mol Biol Rev. 2007;71:495-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 784]  [Cited by in F6Publishing: 597]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
2.  Marco ML, Pavan S, Kleerebezem M. Towards understanding molecular modes of probiotic action. Curr Opin Biotechnol. 2006;17:204-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 199]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
3.  Schell MA, Karmirantzou M, Snel B, Vilanova D, Berger B, Pessi G, Zwahlen MC, Desiere F, Bork P, Delley M. The genome sequence of Bifidobacterium longum reflects its adaptation to the human gastrointestinal tract. Proc Natl Acad Sci USA. 2002;99:14422-14427.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Sela DA, Chapman J, Adeuya A, Kim JH, Chen F, Whitehead TR, Lapidus A, Rokhsar DS, Lebrilla CB, German JB. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Natl Acad Sci USA. 2008;105:18964-18969.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Borriello SP, Hammes WP, Holzapfel W, Marteau P, Schrezenmeir J, Vaara M, Valtonen V. Safety of probiotics that contain lactobacilli or bifidobacteria. Clin Infect Dis. 2003;36:775-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 302]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
6.  O'Brien J, Crittenden R, Arthur C. Ouwehand and Seppo Salminen. Safety evaluation of probiotics. Trends in Food Sci and Technol. 1999;10:418-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 24]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
7.  McBain AJ, Macfarlane GT. Investigations of bifidobacterial ecology and oligosaccharide metabolism in a three-stage compound continuous culture system. Scand J Gastroenterol Suppl. 1997;222:32-40.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Ruiz-Moyano S, Martín A, Benito MJ, Casquete R, Serradilla MJ, Córdoba MD. Safety and functional aspects of pre-selected lactobacilli for probiotic use in Iberian dry-fermented sausages. Meat Sci. 2009;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
9.  Wei YX, Zhang ZY, Liu C, Zhu YZ, Zhu YQ, Zheng H, Zhao GP, Wang S, Guo XK. Complete genome sequence of Bifidobacterium longum JDM301. J Bacteriol. 2010;192:4076-4077.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
10.  Benson DA, Karsch-Mizrachi I, Lipman DJ, Ostell J, Wheeler DL. GenBank. Nucleic Acids Res. 2008;36:D25-D30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 655]  [Cited by in F6Publishing: 750]  [Article Influence: 44.1]  [Reference Citation Analysis (0)]
11.  Delcher AL, Harmon D, Kasif S, White O, Salzberg SL. Improved microbial gene identification with GLIMMER. Nucleic Acids Res. 1999;27:4636-4641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1911]  [Cited by in F6Publishing: 1917]  [Article Influence: 76.7]  [Reference Citation Analysis (0)]
12.  Guo FB, Ou HY, Zhang CT. ZCURVE: a new system for recognizing protein-coding genes in bacterial and archaeal genomes. Nucleic Acids Res. 2003;31:1780-1789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 123]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
13.  Bose M, Barber RD. Prophage Finder: a prophage loci prediction tool for prokaryotic genome sequences. In Silico Biol. 2006;6:223-227.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Grissa I, Vergnaud G, Pourcel C. The CRISPRdb database and tools to display CRISPRs and to generate dictionaries of spacers and repeats. BMC Bioinformatics. 2007;8:172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 732]  [Cited by in F6Publishing: 701]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
15.  Lowe TM, Eddy SR. tRNAscan-SE: a program for improved detection of transfer RNA genes in genomic sequence. Nucleic Acids Res. 1997;25:955-964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3182]  [Cited by in F6Publishing: 3181]  [Article Influence: 117.8]  [Reference Citation Analysis (0)]
16.  Ghai R, Hain T, Chakraborty T. GenomeViz: visualizing microbial genomes. BMC Bioinformatics. 2004;5:198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
17.  Chen L, Yang J, Yu J, Yao Z, Sun L, Shen Y, Jin Q. VFDB: a reference database for bacterial virulence factors. Nucleic Acids Res. 2005;33:D325-D328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 709]  [Cited by in F6Publishing: 1021]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
18.  Liu B, Pop M. ARDB--Antibiotic Resistance Genes Database. Nucleic Acids Res. 2009;37:D443-D447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 814]  [Cited by in F6Publishing: 832]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
19.  Ventura M, Turroni F, Zomer A, Foroni E, Giubellini V, Bottacini F, Canchaya C, Claesson MJ, He F, Mantzourani M. The Bifidobacterium dentium Bd1 genome sequence reflects its genetic adaptation to the human oral cavity. PLoS Genet. 2009;5:e1000785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
20.  Barrangou R, Briczinski EP, Traeger LL, Loquasto JR, Richards M, Horvath P, Coûté-Monvoisin AC, Leyer G, Rendulic S, Steele JL. Comparison of the complete genome sequences of Bifidobacterium animalis subsp. lactis DSM 10140 and Bl-04. J Bacteriol. 2009;191:4144-4151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
21.  Philippe H, Douady CJ. Horizontal gene transfer and phylogenetics. Curr Opin Microbiol. 2003;6:498-505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 138]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
22.  Ammor MS, Flórez AB, Mayo B. Antibiotic resistance in non-enterococcal lactic acid bacteria and bifidobacteria. Food Microbiol. 2007;24:559-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 267]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
23.  Delgado S, Flórez AB, Mayo B. Antibiotic susceptibility of Lactobacillus and Bifidobacterium species from the human gastrointestinal tract. Curr Microbiol. 2005;50:202-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 133]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
24.  Moubareck C, Gavini F, Vaugien L, Butel MJ, Doucet-Populaire F. Antimicrobial susceptibility of bifidobacteria. J Antimicrob Chemother. 2005;55:38-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 121]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
25.  D'Aimmo MR, Modesto M, Biavati B. Antibiotic resistance of lactic acid bacteria and Bifidobacterium spp. isolated from dairy and pharmaceutical products. Int J Food Microbiol. 2007;115:35-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 112]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
26.  Wilson JW, Schurr MJ, LeBlanc CL, Ramamurthy R, Buchanan KL, Nickerson CA. Mechanisms of bacterial pathogenicity. Postgrad Med J. 2002;78:216-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 163]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
27.  von Ossowski I, Reunanen J, Satokari R, Vesterlund S, Kankainen M, Huhtinen H, Tynkkynen S, Salminen S, de Vos WM, Palva A. Mucosal adhesion properties of the probiotic Lactobacillus rhamnosus GG SpaCBA and SpaFED pilin subunits. Appl Environ Microbiol. 2010;76:2049-2057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 154]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
28.  Emanuelsson O, Brunak S, von Heijne G, Nielsen H. Locating proteins in the cell using TargetP, SignalP and related tools. Nat Protoc. 2007;2:953-971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2543]  [Cited by in F6Publishing: 2438]  [Article Influence: 143.4]  [Reference Citation Analysis (0)]
29.  Ventura M, Lee JH, Canchaya C, Zink R, Leahy S, Moreno-Munoz JA, O'Connell-Motherway M, Higgins D, Fitzgerald GF, O'Sullivan DJ. Prophage-like elements in bifidobacteria: insights from genomics, transcription, integration, distribution, and phylogenetic analysis. Appl Environ Microbiol. 2005;71:8692-8705.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Brussow H. Phages of dairy bacteria. Annu Rev Microbiol. 2001;55:283-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 149]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
31.  Touchon M, Rocha EP. Causes of insertion sequences abundance in prokaryotic genomes. Mol Biol Evol. 2007;24:969-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 177]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
32.  Godde JS, Bickerton A. The repetitive DNA elements called CRISPRs and their associated genes: evidence of horizontal transfer among prokaryotes. J Mol Evol. 2006;62:718-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 232]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
33.  O'Driscoll J, Heiter DF, Wilson GG, Fitzgerald GF, Roberts R, van Sinderen D. A genetic dissection of the LlaJI restriction cassette reveals insights on a novel bacteriophage resistance system. BMC Microbiol. 2006;6:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
34.  O'Driscoll J; FEEDAP. Prepared by the Panel on Additives and Products or Substances used in Animal Feed on the Update of the criteria used in the assessment of bacterial resistance to antibiotics of human or veterinary importance. EFSA. 2008;732:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
35.  Scott KP, Melville CM, Barbosa TM, Flint HJ. Occurrence of the new tetracycline resistance gene tet(W) in bacteria from the human gut. Antimicrob Agents Chemother. 2000;44:775-777.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Masco L, Van Hoorde K, De Brandt E, Swings J, Huys G. Antimicrobial susceptibility of Bifidobacterium strains from humans, animals and probiotic products. J Antimicrob Chemother. 2006;58:85-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 96]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
37.  Munakata S, Arakawa C, Kohira R, Fujita Y, Fuchigami T, Mugishima H. A case of D-lactic acid encephalopathy associated with use of probiotics. Brain Dev. 2010;32:691-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
38.  Bongaerts G, Bakkeren J, Severijnen R, Sperl W, Willems H, Naber T, Wevers R, van Meurs A, Tolboom J. Lactobacilli and acidosis in children with short small bowel. J Pediatr Gastroenterol Nutr. 2000;30:288-293.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Uchida H, Yamamoto H, Kisaki Y, Fujino J, Ishimaru Y, Ikeda H. D-lactic acidosis in short-bowel syndrome managed with antibiotics and probiotics. J Pediatr Surg. 2004;39:634-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
40.  Garai G, Dueñas MT, Irastorza A, Martín-Alvarez PJ, Moreno-Arribas MV. Biogenic amines in natural ciders. J Food Prot. 2006;69:3006-3012.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Silla Santos MH. Biogenic amines: their importance in foods. Int J Food Microbiol. 1996;29:213-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1053]  [Cited by in F6Publishing: 932]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
42.  Garai G, Dueñas MT, Irastorza A, Moreno-Arribas MV. Biogenic amine production by lactic acid bacteria isolated from cider. Lett Appl Microbiol. 2007;45:473-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Benno Y, Mitsuoka T. Impact of Bifidobacterium longum on human fecal microflora. Microbiol Immunol. 1992;36:683-694.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Kim Y, Lee D, Kim D, Cho J, Yang J, Chung M, Kim K, Ha N. Inhibition of proliferation in colon cancer cell lines and harmful enzyme activity of colon bacteria by Bifidobacterium adolescentis SPM0212. Arch Pharm Res. 2008;31:468-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 86]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
45.  Choi SS, Kang BY, Chung MJ, Kim SD, Park SH, Kim JS, Kang CY, Ha NJ. Safety assessment of potential lactic acid bacteria Bifidobacterium longum SPM1205 isolated from healthy Koreans. J Microbiol. 2005;43:493-498.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Jones BV, Begley M, Hill C, Gahan CG, Marchesi JR. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc Natl Acad Sci USA. 2008;105:13580-13585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 660]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
47.  Tanaka H, Doesburg K, Iwasaki T, Mierau I. Screening of lactic acid bacteria for bile salt hydrolase activity. J Dairy Sci. 1999;82:2530-2535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 188]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
48.  Vankerckhoven V, Huys G, Vancanneyt M, Vael C, Klare I, Romond MB, Entenza JM, Moreillon P, Wind RD, Knol J. Biosafety assessment of probiotics used for human consumption: recommendations from the EU-PROSAFE project. Trends in Food Sci and Technol. 2008;19:102-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 136]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
49.  Nagengast FM, Grubben MJ, van Munster IP. Role of bile acids in colorectal carcinogenesis. Eur J Cancer. 1995;31A:1067-1070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 261]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
50.  Marteau P, Gerhardt MF, Myara A, Bouvier E, Trivin F, Rambaud JC. Metabolism of Bile Salts by Alimentary Bacteria During Transit in the Human Small Intestine. Micro Ecol in Heal and Dis. 1995;8:151-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
51.  Jones ML, Chen H, Ouyang W, Metz T, Prakash S. Microencapsulated Genetically Engineered Lactobacillus plantarum 80 (pCBH1) for Bile Acid Deconjugation and Its Implication in Lowering Cholesterol. J Biomed Biotechnol. 2004;2004:61-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 64]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
52.  Knaust A, Weber MV, Hammerschmidt S, Bergmann S, Frosch M, Kurzai O. Cytosolic proteins contribute to surface plasminogen recruitment of Neisseria meningitidis. J Bacteriol. 2007;189:3246-3255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 82]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
53.  Lähteenmäki K, Kuusela P, Korhonen TK. Bacterial plasminogen activators and receptors. FEMS Microbiol Rev. 2001;25:531-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
54.  Candela M, Miccoli G, Bergmann S, Turroni S, Vitali B, Hammerschmidt S, Brigidi P. Plasminogen-dependent proteolytic activity in Bifidobacterium lactis. Microbiology. 2008;154:2457-2462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
55.  Candela M, Bergmann S, Vici M, Vitali B, Turroni S, Eikmanns BJ, Hammerschmidt S, Brigidi P. Binding of human plasminogen to Bifidobacterium. J Bacteriol. 2007;189:5929-5936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 85]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
56.  Candela M, Centanni M, Fiori J, Biagi E, Turroni S, Orrico C, Bergmann S, Hammerschmidt S, Brigidi P. DnaK from Bifidobacterium animalis subsp. lactis is a surface-exposed human plasminogen receptor upregulated in response to bile salts. Microbiology. 2010;156:1609-1618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 78]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
57.  Luck SN, Turner SA, Rajakumar K, Sakellaris H, Adler B. Ferric dicitrate transport system (Fec) of Shigella flexneri 2a YSH6000 is encoded on a novel pathogenicity island carrying multiple antibiotic resistance genes. Infect Immun. 2001;69:6012-6021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 98]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
58.  Satin B, Del Giudice G, Della Bianca V, Dusi S, Laudanna C, Tonello F, Kelleher D, Rappuoli R, Montecucco C, Rossi F. The neutrophil-activating protein (HP-NAP) of Helicobacter pylori is a protective antigen and a major virulence factor. J Exp Med. 2000;191:1467-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 229]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
59.  Verbelen C, Dufrêne YF. Direct measurement of Mycobacterium-fibronectin interactions. Integr Biol (Camb). 2009;1:296-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
60.  Lee JH, Karamychev VN, Kozyavkin SA, Mills D, Pavlov AR, Pavlova NV, Polouchine NN, Richardson PM, Shakhova VV, Slesarev AI. Comparative genomic analysis of the gut bacterium Bifidobacterium longum reveals loci susceptible to deletion during pure culture growth. BMC Genomics. 2008;9:247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 146]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
61.  Gaillot O, Pellegrini E, Bregenholt S, Nair S, Berche P. The ClpP serine protease is essential for the intracellular parasitism and virulence of Listeria monocytogenes. Mol Microbiol. 2000;35:1286-1294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 214]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
62.  Alfaleh K, Anabrees J, Bassler D. Probiotics reduce the risk of necrotizing enterocolitis in preterm infants: a meta-analysis. Neonatology. 2010;97:93-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
63.  McFarland LV. Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease. Am J Gastroenterol. 2006;101:812-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 563]  [Cited by in F6Publishing: 472]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
64.  Mach T. Clinical usefulness of probiotics in inflammatory bowel diseases. J Physiol Pharmacol. 2006;57 Suppl 9:23-33.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Parvez S, Malik KA, Ah Kang S, Kim HY. Probiotics and their fermented food products are beneficial for health. J Appl Microbiol. 2006;100:1171-1185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 815]  [Cited by in F6Publishing: 700]  [Article Influence: 38.9]  [Reference Citation Analysis (0)]