Anaerobe xxx (2013) 1e10

Contents lists available at ScienceDirect

Anaerobe journal homepage: www.elsevier.com/locate/anaerobe

Clinical microbiology

In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves Dobroslava Bujnakova, Eva Strakova*, Vladimir Kmet Institute of Animal Physiology, Slovak Academy of Sciences, Soltesovej 4/6, 040 01 Kosice, Slovak Republic

a r t i c l e i n f o

a b s t r a c t

Article history: Received 15 May 2013 Received in revised form 16 September 2013 Accepted 25 October 2013 Available online xxx

A total of 73 chicken and calves isolates were diagnosed using matrix-assisted laser desorption ionization-time-of flight mass spectrometry (Maldi-Tof MS). After a preliminary subtractive screening based on the high acid tolerance at pH 2.5 and bile resistance at 0.3% oxgall, twenty isolates belonging to the species Lactobacillus salivarius, Lactobacillus agilis, Lactobacillus reuteri, Lactobacillus murinus and Lactobacillus amylovorus were in vitro screened for the safety assessment and probiotic properties, including antibiotics susceptibility patterns, biochemical activity and potential for competitive exclusion of biofilm producing pathogens determined by crystal violet and/or quantitative Fluorescent in situ Hybridisation (FISH) assays utilizing 50 Cy 3 labelled probe Enter1432 for enteric group. Antibiotic susceptibility testing was performed according to the ISO norm 10932. The sixteen strains were susceptible to certain antimicrobial agents, except for two chicken (L. salivarius 12K, L. agilis 13K) and two calves (L. reuteri L10/1, L. murinus L9) isolates with the presence non wild-type ECOFFs (epidemiological cut-off) for gentamicin (256 mg ml1), tetracycline (128 mg ml1), kanamycin (256 mg ml1) and streptomycin (96 mg ml1). The two referenced chicken isolates gave positive aac(60 )Ie-aph(200 )Ia and tet(L) PCR results. The wild-type ECOFFs isolates were subjected to the apiZYM analysis for enzyme profile evaluation and amino acid decarboxylase activities determined by qualitative plate method and multiplex PCR for the detection of four genes involved in the production of histamine (histidine decarboxylase, hdc), tyramine (tyrosine decarboxylase, tyrdc) and putrescine (via eithers ornithine decarboxylase, odc, or agmatine deiminase, agdi). From examined strains only two chicken isolates (L. reuteri 14K; L. salivarius 15K) had no harmful b-glucuronidase, b-glucosidase activities connected with detrimental effects in the gastrointestinal tract and together no amino acid decarboxylase activities and no genes associated with biogenic amines production though only chicken L. salivarius 15K whole cells and acid supernatants shown strong suppressive potential against biofilm-forming Klebsiella and Escherichia coli. Our results highlight that above-mentioned isolate L. salivarius 15K fulfils the principle requirements of a qualified probiotic and may be seen as a reliable candidate for further validation studies in chicken. Ó 2013 Elsevier Ltd. All rights reserved.

Keywords: Lactobacilli Escherichia coli Klebsiella Maldi-Tof MS b-Glucuronidase Antibiotic resistance

1. Introduction Antibiotics used as growth promoters in animal feeds have been banned and nowadays, the possibility of using alternative additives instead of antibiotics is being researched. One such opportunity is probiotics. In recent years, worldwide interest in the use of probiotic bacteria for health promotion and disease prevention has increased significantly in scientific community, consumers and food producers. This interest is based on the knowledge that the targeted use of microorganisms with suitable properties may have beneficial effect on

* Corresponding author. Tel.: þ421 915 865 646. E-mail address: [email protected] (E. Strakova).

animal and human health. Bacterial strains most commonly used as probiotics are Lactobacillus, Lactococcus, Enterococcus and Bifidobacterium [1] which belong to the lactic acid bacteria. Before a screening of promising probiotic functional efficacy it is necessary to provide of preliminary in vitro screening to ensure the safety aspects of tested strains. Probiotic microorganisms should be free of undesirable traits, such as transmissible antibiotic resistance [2,3] (to avoid spreading resistance determinants in intestinal pathogenic or opportunistic bacteria), specific amino acid decarboxylase activities and thus, the possibility to synthesize biogenic amines with potential health risks to consumers [4] and harmful biochemical activities such as a-chymotrypsin, b-glucosidase, bglucuronidase, and N-acetyl-ß-glucosaminidase activities, which are often associated with intestinal diseases and involved in

1075-9964/$ e see front matter Ó 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

2

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

generating carcinogens and tumour promoters [5]. The next important criterion is survival under gastrointestinal condition (acid pH and bile salt). Though the safety and efficacy of probiotics needs to be demonstrated in controlled clinical trials [6], their primary selection is based on a series of well-defined in vitro tests [7,8]. With the aim of selecting new promising probiotic candidates with minimized undesirable properties, twenty chicken and calves lactobacilli isolates were subjected to comprehensive in vitro analyses to assess their key safety and functionality. 2. Materials and methods 2.1. Bacterial isolates and growth condition From 73 chicken and calves isolates identified using Maldi-Tof MS, twenty selected lactobacilli belonged to the five following species: Lactobacillus salivarius (n ¼ 7), Lactobacillus agilis (n ¼ 2), Lactobacillus reuteri (n ¼ 9) and Lactobacillus amylovorus (n ¼ 1), Lactobacillus murinus (n ¼ 1). All lactobacilli isolates were routinely grown in MRS medium (Oxoid, England) by anaerobic incubation at 37  C for 48 h. The pathogens used in the experiments were Klebsiella pneumonii KPC (carbapenemase positive strain); uropathogenic Escherichia coli K188 and E. coli DH5a/pCIB10B (ibeA positive strain), which was obtained from VA Medical Center, Minneapolis, United States. The strains of E. coli and K. pneumonii were grown in MacConkey agar (Oxoid, England) overnight at 37  C. 2.2. Bacterial identification 2.2.1. Maldi-Tof MS bacterial identification Maldi-Tof MS was performed on a Microflex LT instrument (Bruker Daltonik GmbH, Leipzig, Germany) as described Bessede et al. [9]. To identify microorganisms, the raw spectra obtained for each isolate were imported into BioTyper software, version 2.0 (Bruker Daltonik). When the obtained score was >2.00, identification was considered correct at the species level; in the range 1.7e 1.999, the identification was considered correct at the genus level; and 0.2, the biofilm production was considered as strong. 2.7.1. Inhibition of biofilm formation by different lactobacilli fractions Different fractions (acid, neutralized supernatant and whole cells) from lactobacilli were prepared essentially as described previously [24] and were used to evaluate the inhibition of biofilm formation. The selected pathogens were transferred to Nunc microplates as described before, and the different Lactobacillus sp. fractions were added to each well to complete 200 ml volume. The plates were incubated for 24 h at 37  C and biofilm was evaluated by using crystal violet assay as described earlier and/or quantitative FISH methods. Pathogens or Lactobacillus sp. were included as control. All the assays were performed by triplicate. 2.7.2. The quantitative FISH assay for biofilm-forming bacteria enumeration Before enumeration, bacterial biofilms were harvested by addition of 100 ml of 0.5% Triton X-100, fixed in 4% paraformaldehyde

Lactobacilli were subjected to a subtractive system of reliable in vitro screening methods recommended by FAO/WHO committee [7]. 3.2. Bacterial identification 3.2.1. Maldi-Tof MS bacterial identification Quality assurance programmes associated with research, development, production and validation of the health benefits of probiotic Lactobacillus require their relevant identification using modern techniques. Biochemical methods, such as API 50 CH enzymatic tests may be useful to obtain a first tentative classification at the genus level, but the identification result should in any case be confirmed by molecular methods [27]. The one suitable method is considered MALDI-Tof mass spectrometry protein fingerprinting analysis. Nevertheless, DNA based technique is considered as more accurate for bacterial identification than protein fingerprinting, the available literature indicates that misidentification by Maldi-Tof MS is most probably associated with an insufficient number of reference strains available in the Maldi-Tof MS spectral database and optimization of extraction protocols for difficult-to-treat samples is undoubtedly important for increase the accuracy of identification by the Maldi-Tof MS [28]. The Maldi Tof identification has been routinely used in our lab from 2010. The methodology of samples preparation especially for lactobacilli is empirically well designed. Moreover, our database is continually actualized and currently contains 225 lactobacilli belonging to 14 species. From total of 73 animal isolates, twenty lactobacilli with obtained scores higher than 2.3 were selected for further experiments. The dendrogram generated from cluster analysis of MaldiTof mass spectra of these strains pointed out distinctive clusters consisting of the same species lactobacilli (Fig. 1). The nine L. reuteri spectra constituted the first cluster that could potentially be subdivided into four smaller groups. A distance level of 400 separated the most extreme L. reuteri spectra, whereas the sub-clustering reduced the maximal distance level in all groups to approximately 200. A second cluster regrouped L. murinus, L. amylovorus and two L. agilis spectra despite a high distance level of 500 from among the L. murinus, L. amylovorus and L. agilis. Distance level of 400 is between L. murinus and L. amylovorus, e contra distance level lesser than 100 among L. agilis. Etceteras seven strains belonging to

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

4

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

Fig. 1. MSP dendrogram of MALDI-TOF mass spectral profiles generated by the MALDI Biotyper; Strains clustering with distance level lower than 500 could be classified up to species level.

L. salivarius were separated to three sub-clusters and with an additional distinct spectrum of one single strain L. salivarius 15K with a maximum distance level of 300. Distance levels on horizontal axis of MSP dendrogram indicating similarity were under 500 which have been described as reliably classified bacteria [29]. Clusters under a distance level of 100 indicating a high similarity between the isolates. 3.2.2. Genotypic bacterial identification The all of tested strains gave positive results with genus-specific primers (LBLMA 1-rev and R16-1). The separation by a multiplex PCR into four groups based on the nucleotide sequences of the 16Se 23S rRNA intergenic spacer region and adjacent 23S rRNA gene showed that sixteen strains identified by Maldi-Tof MS as L. salivarius or L. reuteri belonged to group IV, such as is described in research article written by Song et al. [11]. The seven L. salivarius strains together with finally selected L. salivarius 15K and L. salivarius 12K (aac(60 )-aph(20 )-Ia positive strain) subjected to PCR with species-specific primers for L. salivarius (For-Sal-3; Rev-Sal-1) gave positive 97 bp amplicons, which confirmed results obtaining by Maldi-Tof MS. 3.3. Tolerance to acid and bile Tolerance to gastric conditions is one of the in vitro tests frequently recommended for the evaluation of the probiotic attributes of an individual strain [30]. The twenty chosen isolates expressed high acid and bile tolerance after 4-h exposure, in doing so bacterial viability varied between 48 and 86%, respectively 52%e 91%. The results are presented in Table 1. Since viability and survival of probiotic bacteria during passage through the stomach is a considerable parameter to reach the intestine and provide potentially beneficial effect, the very important step of the probiotic potential study is focused on the selection of acid and bile tolerant isolates. The pH in stomach ranges from 1, during fasting, to 4.5, after a meal, and food ingestion can take up to 3 h. Since Lactobacillus strains are known to survive at pH 4.6, lower pH values are usually examined. The studies done so far indicate that majority of examined strains were completely resistant to pH 3 after 3 h of exposure,

but most of the strains displayed loss of viability when exposed to pH 1 or 2 for 1 h. These results are in agreement with those obtained from previous similar studies, where Lactobacillus strains were able to retain their viability when exposed to pH values of 2.5e4.0, but displayed loss of viability at lower pH values [31,32]. On the other hand, it was, however, observed before that although some strains did not survive in vitro in gastric juice with pH 2 for more than 15 min, they reached the colon in viable state and exerted a beneficial effect in vivo. It is important to consider also the buffering capacity of the ingested food or using of encapsulated delivery systems which can improve acid-sensitive strain viabilities during gastric transit [33]. Actually, ingested bacteria are rarely

Table 1 Effect of acid and bile on Lactobacillus sp. viability. Strains

Viable counts of bacteria (log CFU ml1) pH 2.5

0.3% oxgall t4

t0 L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L.

reuteri L4 reuteri L4/1 reuteri L5 reuteri L5/1 amylovorus L6 reuteri L6/1 reuteri L7 reuteri L8 murinus L9 reuteri L10/1 agilis 4K salivarius 5K salivarius 6K salivarius 8K salivarius 9K salivarius 10K salivarius 12K agilis 13K reuteri 14K salivarius 15K

6.37 6.40 6.78 5.78 5.48 7.58 7.47 6.14 5.14 6.77 5.77 6.55 6.48 5.99 6.18 7.58 6.77 6.20 7.12 6.99

                   

0.14 0.32 0.38 0.19 0.22 0.47 0.28 0.25 0.25 0.14 0.38 0.22 0.65 0.14 0.22 0.54 0.30 0.14 0.37 0.51

5.04 5.37 5.15 4.17 3.97 5.98 5.00 4.52 4.12 3.24 3.74 4.87 4.75 4.47 4.17 5.98 4.98 5.34 5.68 5.97

t0                    

0.10 0.32 0.27 0.11 0.40 0.12 0.12 0.04 0.04 0.16 0.18 0.12 0.37 0.11 0.20 0.12 0.39 0.10 0.45 0.12

(79%) (84%) (76%) (72%) (72%) (79%) (67%) (74%) (80%) (48%) (65%) (74%) (73%) (74%) (67%) (79%) (74%) (86%) (80%) (85%)

6.33 6.35 6.21 5.55 6.54 6.48 6.21 6.54 6.00 6.33 5.33 6.05 6.56 5.75 6.52 6.33 6.48 6.93 6.98 6.85

t4                    

0.21 0.12 0.25 0.38 0.02 0.09 0.33 0.15 0.55 0.21 0.41 0.12 0.07 0.18 0.62 0.09 0.50 0.29 0.69 0.40

5.14 5.73 5.05 4.07 4.97 4.78 4.99 5.32 5.02 4.14 3.14 3.37 4.13 5.17 4.97 5.78 4.64 6.14 5.68 5.47

                   

0.17 0.22 0.25 0.10 0.39 0.12 0.48 0.24 0.44 0.47 0.17 0.22 0.25 0.10 0.20 0.12 0.12 0.17 0.42 0.22

(81%) (90%) (81%) (73%) (76%) (74%) (80%) (81%) (84%) (65%) (59%) (52%) (78%) (89%) (76%) (91%) (72%) (89%) (81%) (80%)

t0eViable counts (log CFU ml1  SEM) of each strain at 0 h. t4eViable counts (log CFU ml1  SEM) of each strain at 4 h (% viability of selected lactobacilli). in grey selected L. salivarius 15K.

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

5

Table 2 Minimal inhibitory concentration (MIC) (in mg ml1) of selected intestinal lactobacilli strains to 16 antibiotics determined by the VETMIC Lact 1 and 2 system. Strain

L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L. L.

reuteri L4 reuteri L4/1 reuteri L5 reuteri L5/1 amylovorus L6 reuteri L6/1 reuteri L7 reuteri L8 murinus L9 reuteri L10/1 agilis 4K salivarius 5K salivarius 6K salivarius 8K salivarius 9K salivarius 10K salivarius 12K agilis 13K reuteri 14K salivarius 15K

Antibiotics Gm

Km

Sm

Nm

Tc

Em

Cl

Cm

Am

Pc

Va

Vi

Lz

Tm

Ci

Ri

2 4 2 2 2 4 2 2 256 256 2 2 2 2 2 2 256 256 4 4

8 8 8 8 16 8 4 4 256 256 16 4 2 8 8 4 256 256 4 8

8 16 16 8 8 16 16 8 96 96 32 8 8 8 8 8 96 96 8 8

1 2 2 1 0.5 0.5 0.5 1 2 4 8 2 4 0.5 0.5 1 2 0.5 0.5 4

8 8 8 4 4 8 2 8 128 128 4 1 1 1 1 1 128 128 4 2

0.25 0.25 0.5 0.5 0.25 0.25 0.25 0.25 0.25 0.5 0.25 0.5 0.25 0.25 0.25 0.25 0.25 0.25 0.25 0.25

0.12 0.5 0.12 0.12 0.12 0.5 0.5 0.12 0.12 0.12 0.12 0.12 0.12 0.12 0.12 0.12 0. 5 0.12 0.12 0.12

1 2 2 1 1 2 2 2 2 2 1 1 2 1 2 1 1 1 2 2

0.12 0.12 0.5 0.12 0.25 0.12 0.12 0.12 0.12 0.12 0.12 0.12 0.25 0.25 0.25 0.25 0.25 0.12 0.12 0.12

0.5 1 0.5 1 1 0.5 0.5 0.5 0.5 1 1 0.5 0.5 0.5 0.5 0.5 0.5 1 1 1

16 128 32 16 32 16 16 16 32 16 16 2 2 2 2 2 2 64 32 2

0.5 1 0.5 1 1 1 1 1 1 0.5 1 0.5 0.5 1 0.5 0.5 1 0.5 1 1

2 2 4 2 4 2 2 2 2 2 2 4 2 2 2 4 2 2 2 2

0.12 0.12 0.25 0.5 0.5 0.12 0.12 0.12 0.25 0.25 0.12 0.12 0.12 0.12 0.12 0.12 2 2 0.25 0.25

4 8 4 4 8 8 16 16 4 4 4 4 4 8 4 8 16 32 4 4

0.5 0.5 1 1 1 0.5 0.5 1 1 1 0.5 0.5 0.5 0.5 0.5 0.5 1 1 1 0.5

Gm-gentamicin; Km-kanamycin; Sm-streptomycin; Nm-neomycin; Tc-tetracycline; Em-erythromycin; Cl-clindamycin; Cm-chloramphenicol; Am-ampicillin; Pc-penicillin; Va-vancomycin; Vi-virgniamycin; Lz-linezolid; Tm-trimethoprim; Ci-ciprofloxacin; Ri-rifampicin. In grey, MIC values surpassing the microbiological breakpoints defined by the FEEDAP Panel (European Commission 2008).

subjected to pH below 3 [34]. These previous observations were reason for selection pH value 2.5 in our lactobacilli survival capability experiment. Resistance to bile salts is generally considered as an essential property for probiotic strains to survive the conditions in the small intestine. The relevant physiological concentrations of bile range are from 0.10 to 0.30% (w/v). Thus, it is necessary that efficient probiotic bacteria should be able to grow in bile salt with these concentrations [35].

tests (Liofilchem, Italy). This small antibiotic-resistant fraction justifies an antibiotic-susceptibility assay to avoid the inclusion of resistant strains in the formulation of probiotics. Although, a phenotypic test cannot confirm the presence or absence of transferable resistance genes, some of the observed levels are compatible with common transmissible determinants. These phenotypic

3.4. Antibiotic susceptibility testing and MIC determinations Antibiotic resistance of microorganisms used as probiotic agents is an area of growing concern. It is believed that antibiotic used for food-producing animals can promote the emergence of acquired antibiotic resistance in bacteria present in the intestinal microflora. Then, the antibiotic-resistant bacteria can transfer the resistance factor to other pathogenic bacteria through the exchange of genetic material [36]. One of the safety considerations in probiotic studies is the verification that a prospective probiotic strain does not contain potentially transferable resistance genes. The sixteen isolates were susceptible to tetracycline, with minimum inhibitory concentration (MIC) values ranging from 1 to 8 mg ml1, to gentamicin (2e4 mg ml1), to erythromycin (0.25e 0.5 mg ml1), to clindamycin (0.12e0.5 mg ml1), to streptomycin (8e32 mg ml1), to chloramphenicol (1e2 mg ml1), to neomycin (0.5e8 mg ml1), to kanamycin (2e16 mg ml1), to ampicillin (0.12e 0.5 mg ml1), to penicillin (0. 5 to 1 mg ml1), to vancomycin (2e 128 mg ml1), to virginiamycin (0. 5 to 1 mg ml1), to linezolid (2e 4 mg ml1), to trimethoprim (0.12e2 mg ml1), to ciprofloxacin (4e 32 mg ml1) and to rifampicin (0. 5 to 1 mg ml1), besides two chicken (L. salivarius 12K, L. agilis 13K) and two calves (L. reuteri L10/ 1, L. murinus L9) isolates with the presence of non wild-type ECOFFs with MIC levels for gentamicin (256 mg ml1), tetracycline (128 mg ml1), kanamycin (256 mg ml1) and streptomycin (96 mg ml1) (Table 2), which are regarded as resistant according to the microbiological breakpoints given by the recent FEEDAP document of the EFSA [15]. MIC values surpassing the microbiological breakpoints were confirmed moreover by using MIC strip

Fig. 2. PCR amplification of the tet (L) gene (229 bp amplicon). The PCR products were obtained using genomic DNA of the Lactobacillus salivarius 12K (lane 1); and Lactobacillus agilis 13K (lane 2); M: Mid Range DNA Ladder 100 bp to 3 kb linear scale molecular weight marker (Jena Bioscience).

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

6

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

chicken isolates, resistant to tetracycline (L. salivarius 12K, L. agilis 13K) with MIC of 128 mg ml1 were positive to efflux pump encoding tet(L)gene (Fig. 2), which is located on pL S55 plasmid as described Ammor et al. [37]. A gentamicin and a kanamycin MIC of 256 mg ml1 was found in above-mentioned chicken isolates; a kanamycin MIC of 128 mg ml1 likewise in two calves (L. reuteri L10/ 1, L. murinus L9) strains. The forenamed chicken gentamicinresistant isolate L. salivarius 12K gave positive aac(60 )Ie-aph(200 )Ia PCR result (Fig. 3) and negative ones for aph(30 )-IIIa genes, e contra the two calves kanamycin-resistant strains, which gave a negative PCR results for the both of genes. The aac(60 )Ie-aph(200 )Ia amplicon was purified, sequenced and sequence similarity was determined using BLAST in the GenBank database. This comparison showed that our sequence had 100% identity with the N-terminal region of the AAC (60 )-APH (200 ) aminoglycoside-modifying enzyme carries the acetyltransferase activity of Staphylococcus aureus (accession no. AB568461). The first report of the AAC(60 )-APH(200 ) aminoglycosidemodifying enzyme in Lactobacillus of animal origin was described by Tenorio et al. [38], who expects that it is a possibility of transfer the bifunctional gene aac(60 )Ie-aph(200 )Ia, which is a part of the composite transposon Tn4001 widely distributed among gram-positive microorganisms [39], from Enterococcus to Lactobacillus. 3.5. Determination of ability to produce biogenic amines and the multiplex PCR for detection of genes responsible for biogenic amines production

Fig. 3. PCR amplification of the aac(60 )-aph(20 )-Ia gene (348 bp amplicon). The PCR product was obtained using genomic DNA of the Lactobacillus salivarius 12K (lane 1); M: Mid Range DNA Ladder 100 bp to 3 kb linear scale molecular weight marker (Jena Bioscience).

results underline the necessity to monitor the presence of transmissible antibiotic resistance genes at the molecular level. 3.4.1. PCR Detection of resistance genes PCR-based detection of genes responsible for resistance to gentamicin [aac(60 )-aph(20 )-Ia], tetracycline [tet(L), tet(M), tet(S)], kanamycin [aph(30 )-IIIa] and streptomycin/spectinomycin [aadA] was applied to strains suspected to carry acquired resistance. Two

To ensure their safety for use in animal and their products the sixteen wild-type ECOFFs isolates were screened for potentiality to produce biogenic amines. Phenotype detection of histidine, tyrosine, ornithine decarboxylase activities determined by qualitative plate method show that four calve isolates L. reuteri (L5/1, L6/1, L7) and L. amylovorus L6 possessed histidine decarboxylase activity (Fig. 4). Though, the detection of biogenic amines producing bacteria by conventional culture techniques is often tedious and unreliable, exhibiting disadvantages such as lack of speed, appearance of false positive/negative results and low sensibility [40]. As molecular methods are fast, reliable and culture-independent, they represent an interesting alternative for the detection of bacteria producing biogenic amines. In order to rapidly screen isolates for potential biogenic amines-producing strains, a multiplex PCR method was applied to simultaneously detect four genes involved in the production of the main biogenic amines: tyramine via tyrosine decarboxylase, putrescine via ornithine decarboxylase or agmatine deiminase and histamine via histidine decarboxylase. The

Fig. 4. Phenotype detection of histidine decarboxylase activities determined by qualitative plate method-Lactobacillus reuteri L 5/1 after a) two or b) four days of incubation on decarboxylase agar supplemented with histidine and bromocresol purple as indicator dye.

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

Lactobacilli, E. coli and Klebsiella biofilm-forming capacities on abiotic surfaces were evaluated. The L. salivarius 15K (A570 ¼

0 0 0 10 20 20 5 5 5 0 10 10 10 10 30 20 0 30 10 10 5 30 0 0 30 20 5 20 0 5 0 0 30 10 0 0 0 0 10 0 10 10 0 0 0 5 0 0 40 40 40 40 40 5 40 40 40 30 40 40 40 30 5 20 20 0 5 40 20 0 10 10 5 5 5 20 5 5 5 5 5 10 0 0 10 5 20 5 40 40 5 20 5 5 0 0 30 30 20 30 40 20 40 40 20 5 0 0 5 0 10 5 10 20 20 5 5 0 0 0 5 0 5 5 10 20 0 5 20 0 0 5 10 10 10

10 30 10 20 10 10 20 5 20 20 10 40 10 20 10 20 10 5 20 5

Enzyme assayeda,b

In grey L. salivarius 15K and L. reuteri 14K without harmful enzyme activities. a Enzyme activity measured as the approximate nanomoles of substrate hydrolysed during 4-h incubation. b Harmful enzyme activities.

3.7. Screening of biofilm production in Lactobacillus sp.; E. coli; K. pneumonii

Strains

The enzyme activities of twelve lactobacilli, free of acquired resistance patterns and genes involved in the main biogenic amines production, were screened. The strains displayed various enzyme profiles as summarized in Table 3. Safety assessment includes the lack of harmful activities, such as b-glucosidase, b-glucuronidase activities, which are associated with detrimental effects in the colon by releasing aglycones and deconjugating glucuronic acid-conjugated carcinogens [41,42]. The L. salivarius (6K, 8K) and L. reuteri (L4, L4/1, L8) showed moderate b-glucosidase activity (10e20 nmol of substrate hydrolysed); L. salivarius 5K, 10K and L. reuteri L5 expressed high bglucosidase activity (30 nmol of substrate hydrolysed) and moreover L. reuteri L5, L. agilis 4K and L. salivarius (5K, 10K) had slow or moderate b-glucuronidase activity (5e10 nmol of substrate hydrolysed). In contrast, the b-galactosidase released by probiotics has been implicated in the relief of lactose maldigestion symptoms [43]. The two convenient strains L. salivarius 15K and L. reuteri 14K had high b-galactosidase activities (40 nmol of substrate hydrolysed). Simultaneously mentioned strains possessed a-fucosidase activity (through to help intestine colonization) and therewithal lack of aglucosidase activity (a trait considered positive for diabetic and obese humans) [44,45].

Table 3 Enzyme activities of Lactobacillus sp. assayed by the Api-ZYM system.

3.6. Enzyme activities

reuteri L4 reuteri L4/1 reuteri L5 reuteri L8 agilis 4K salivarius 5K salivarius 6K salivarius 8K salivarius 9K salivarius 10K reuteri 14K salivarius 15K

multiplex PCR was able to detect the corresponding a 435 bp hdc amplicons in phenotypically histidine positive strains (Fig. 5). One potential hdc target gene fragments were further confirmed by sequencing simplex PCR amplification product and sequence similarity was determined using BLAST in the GenBank database and showed 99% identity on 100% query cover with the amino acid sequence of the L. reuteri DSM 20016 (accession no. YP001272408).

L. L. L. L. L. L. L. L. L. L. L. L.

Fig. 5. Genotype detection of histidine decarboxylase, 435 bp hdc amplicons; The PCR products were obtained using genomic DNA of the Lactobacillus reuteri L5/1 (lane 1); Lactobacillus reuteri L6/1 (lane 2) Lactobacillus reuteri L7 (lane 3) and Lactobacillus amylovorus L6 (lane 4); M: Mid Range DNA Ladder 100 bp to 3 kb linear scale molecular weight marker (Jena Bioscience).

7

Naphtol-AS-BIAcid Esterase EsteraseLeuValCysa-Galactosidase b-Galactosidase b-Glucuronidaseb a-Glucosidase b-Glucosidaseb a-Fucosidase phosphohydrolase phosphatase (C4) lipase (C8) arylamidase arylamidase arylamidase

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

8

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

Table 4 The potential for exclusion of biofilm producing pathogens determined by quantitative FISH. >Strains

K188 KPC DH5a/pCIB10B

Bacterial number determined by FISHa Monoculture

L. reuteri 14K

L. salivarius 15K

4.58  0.25 3.73  0.22 5.53  0.27

4.33  0.32 (5%) 3.67  0.32 (2%) 5.10  0.18 (8%)

2.78  0.37** (39%) 2.21  0.32** (41%) 3.27  0.25*** (41%)

a number of enteric groups (Enter1432 probe) are given as log10 of number of biofilm forming bacteria per well  SEM; K188, KPC, DH5a/pCIB10B: monoculture and after treatment with whole cells L. reuteri 14K or L. salivarius 15K; experiments were performed in triplicate; significantly different from monoculture at **p < 0.01, ***p < 0.001; (% of pathogens biofilm inhibition by lactobacilli).

0.395  0.015) and L. reuteri 14K (A570 ¼ 0.264  0.025) produced strong biofilms. It is well known, that selection of probiotic is primary based on good adhesion to intestinal surfaces (in vitro-mucin, cell lines etc.). The ability to adhere to abiotic surfaces is not routinely tested in probiotic strains, even though the potential role of commensal bacteria biofilm formation in host health and disease is substantial, because allow to them to colonize of gut environment, remain more resistant and play a role in the protection of the gut epithelium from adherence of pathogenic agents. Probiotics in biofilm-like communities may be essential for long-term remodelling of the healthy composition and function of the intestinal population, therefore, probiotic strategies for the prevention and treatment of disease may require the intensive search for autochthonous strains with beneficial characteristics that include the capability to form biofilm [46]. Although biofilm formation on abiotic surfaces can’t definitely replace testing of adhesion ability to biotic surfaces, in recent years, some researchers test potential probiotic for ability of biofilm formation and/or some authors have shown a statistically significant positive correlation between adhesiveness to biotic surfaces and ability to form biofilm on abiotic surfaces [47e49]. Similarly, employed E. coli K188 (A570 ¼ 0.286  0.014); E. coli DH5a/pCIB10B (A570 ¼ 0.354  0.024) and K. pneumonii (A570 ¼ 0.249  0.035) were strong biofilm producers. Indeed, many animal bacterial pathogens exist predominantly as adherent (also called biofilm or sessile) organisms within tissue and on inert surfaces and it is well known that such infections are extremely difficult treatable, regarding to their high antimicrobial resistance [50]. The tolerance towards currently available antimicrobials calls for development of alternative treatment strategies, as bacteria, in retrospect, have a unique ability to evade the actions of classic antibiotics. 3.7.1. Inhibition of biofilm formation by different lactobacilli fractions The L. salivarius 15K and L. reuteri 14K (whole cells, acid and neutralized supernatants) different fractions competence to

inhibit E. coli and Klebsiella biofilms was tested. Our results revealed that whole cell and acid supernatant of L. salivarius 15K dramatically reduced E. coli and K. pneumonii biofilmformation. In this study, we showed that co-incubation of L. salivarius 15K whole cells together with E. coli K188; DH5a/pCIB10B and K. pneumonii resulted in a strong decrease in pathogens biofilm forming capacity (39e41% inhibition), whereas L. reuteri 14K whole cells displayed only marginal biofilm competitive exclusion potential (2e8% inhibition) (Table 4). The remarkable anti-biofilm effects of L. salivarius 15K against pathogens may be related to its stronger adhesion to abiotic surfaces in comparison with L. reuteri 14K, which could induce greater competition between the probiotic bacterium and pathogens and its strong co-aggregation potential (data not shown) may be an additional interpretation for the lack of pathogens biofilm formation [51]. Another possible mechanism of inhibition pathogens biofilms formation may be related to release of biosurfactants (not confirmed in our study), which alter surface hydrophobicity and hereby inhibit the adhesion of pathogenic microorganisms [52]. Treating E. coli K188; E. coli DH5a/pCIB10B and K. pneumonii biofilms with L. salivarius 15K acid supernatants revealed significant differences (P < 0.01, P < 0.001) in optical density between pre and post treatment. Neutralization of supernatants greatly reduced inhibition pathogens biofilms formation, suggesting that the inhibitory effect was due to acids production. The obtained results are presented in Table 5. To address the possible contribution of bacteriocins in the observed inhibition of biofilms formation, lactobacilli supernatant were treated with either trypsin or proteinase K. Protease treatment of the supernatants did not reduce inhibition of biofilms formation (data not shown), suggesting that inhibition was not due to the production of a proteinaceous component. These data support the hypothesis that ability of L. salivarius 15K to inhibit biofilms formation is caused, at least in part, to the production of acids, which caused reduction of cytoplasmic pH and metabolic activities and by this way internal bacterial osmotic disorders [53,54]. The similarly results were obtained by Juarez Tomás et al. [55]. Our results indicate that L. salivarius 15K whole cells and acid supernatant is a potent antimicrobial agent against E. coli and Klebsiella biofilms and could became one of the strategies to suppress or control biofilm forming infections, but we would like to underline the necessity for further studies. 4. Conclusion Following the subtractive in vitro screening strategy, the chicken strain L. salivarius 15K was finally selected based on promising probiotic properties without harmful characteristics. This strain could be taken in further investigation in in vivo studies to elucidate its potential health benefits and its application as novel probiotic strains in the animal (poultry) feed supplements.

Table 5 The potential for inhibition of biofilm producing pathogens determined by crystal violet assay. Strains

Crystal violet assay (A570)a Monoculture

K188 KPC DH5a/pCIB10B

0.286  0.014 0.249  0.035 0.354  0.024

L. reuteri 14K

L. salivarius 15K

AS

NS

AS

NS

0.253  0.117 0.015  0.009*** 0.152  0.023

0.267  0.02 0.199  0.039 0.325  0.045

0.004  0.022*** 0.045  0.001** 0.123  0.025**

0.317  0.045 0.230  0.059 0.360  0.065

a average values of absorbance from triplicate measurements  SEM are presented, A570eabsorbance at l ¼ 570 nm; K188, KPC, DH5a/pCIB10B: monoculture and after treatment with L. reuteri 14K or L. salivarius 15K (AS: acid supernatant; NS: neutralized supernatant); significantly different from monoculture at **p < 0.01, ***p < 0.001.

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

Acknowledgements This work is the result of the VEGA projects 2/0014/13. We would like to thank Prof. James R. Johnson and Brian Johnson from VA Medical Center, Minneapolis, United States for kindly providing the E. coli DH5a/pCIB10B. References [1] Mercenier A, Pavan S, Pot B. Probiotics as biotherapeutic agents: present knowledge and future prospects. Curr Pharm Des 2003;9(2):175e91. [2] Rönkä E, Malinen E, Saarela M, Rinta-Koski M, Aarnikunnas J, Palva A. Probiotic and milk technological properties of Lactobacillus brevis. J Food Microbiol 2003;82:63e74. [3] Hummel AS, Hertel C, Holzapfel WH, Franz C. Antibiotic resistances of starter and probiotic strains of lactic acid bacteria. Appl Environ Microbiol 2007;73: 730e9. [4] Ammor MS, Mayo B. Selection criteria for lactic acid bacteria to be used as functional starter cultures in dry sausage production: an update. Meat Sci 2007;76:138e46. [5] Parodi PW. The role of intestinal bacteria in the causation and prevention of cancer, modulation by diet and probiotics. Aust J Dairy 1999;54:103e21. [6] Reid G, Jass J, Sebulsky MT, McCormick JK. Potential uses of probiotics in clinical practice. Clin Microbiol Rev 2003;16(4):658e72. [7] Report of a joint FAO/WHO working group on drafting guidelines for the evaluation of probiotics in food, London, Ontario FAO/WHO guidelines for the evaluation of probiotics in food., http://www.who.int/foodsafety/publications/ fs_management/probiotics2/en/index.html; 2002 [accessed 2007]. [8] Sanders ME. Probiotics: definition, sources, selection, and uses. Clin Infect Dis 2008;46:58e61. [9] Bessede E, Angla-gre M, Delagarde Y, Sep Hieng S, Ménard A, Mégraud F. Matrix-assisted laser-desorption/ionization biotyper: experience in the routine of a University Hospital. Clin Microbiol Infect 2011;17:533e8. [10] Dubernet S, Desmasures N, Gueguen M. A PCR-based method for identification of lactobacilli at the genus level. FEMS Microbiol Lett 2002;214: 271e5. [11] Song YL, Kato N, Liu CX, Matsumiya Y, Kato H, Watanabe K. Rapid identification of 11 human intestinal Lactobacillus species by multiplex PCR assays using group- and species-specific primers derived from the 16S-23S rRNA intergenic spacer region and its flanking 23S rRNA. FEMS Microbiol Lett 2000;187:167e73. [12] Harrow SA, Velmurugu Ravindran RCB, Marshall JW, Tannock GW. Real-time quantitative PCR measurement of Ileal Lactobacillus salivarius populations from Broiler chickens to determine the influence of farming practices. Appl Environ Microbiol 2007;73:7123e7. [13] Anderson RC, Cookson AL, McNabb WC, Kelly WJ, Roy NC. Lactobacillus plantarum DSM 2648 is a potential probiotic that enhances intestinal barrier function. FEMS Microbiol Lett 2010;309:184e92. [14] ISO 10932/IDF 223. Milk and milk products-determination of minimal inhibitory concentration (MIC) of antibiotic applicable to bifidobacteria and non-enterococcal lactic acid bacteria (LAB); 2010. [15] EFSA. Technical guidance. Update of the criteria used in the assessment of bacterial resistance to antibiotics of human or veterinary importance. EFSA J 2008;732:1e15. [16] Maietti L, Bovini B, Huys G, Giraffa G. Incidence of antibiotic resistance and virulence determinats among Enterococcus italicus isolates from dairy products. Syst Appl Microbiol 2007;30:509e17. [17] Malhotra-Kumar S, Lammens Ch, Piessens J, Goossens H. Multiplex PCR for simultaneous detection of macrolide and tetracycline resistance determinants in Streptococci. Antimicrob Agents Chemother 2005;49(11):4798e800. [18] Vakulenko SB, Donabedian SM, Voskresenskiy AM, Zervos MJ, Lerner SA, Chow JW. Multiplex PCR for detection of aminoglycoside resistance genes in Enterococci. Antimicrob Agents Chemother 2003;47(4):1423e6. [19] Clark NC, Olsvik O, Swenson JM, Spiegel CA, Tenover FC. Detection of a streptomycin/spectinomycin adenylyltransferase gene (aadA) in Enterococcus faecalis. Antimicrob Agents Chemother 1999;43:157e60. [20] Bover-Cid S, Holzapfel WH. Improved screening procedure of biogenic amine production by lactic acid bacteria. Int J Food Microbiol 1999;53:33e41. [21] Coton M, Romano A, Spano G, Ziegler K, Vetrana C, Desmarais C, et al. Occurrence of biogenic amine-forming lactic acid bacteria in wine and cider. Food Microbiol 2010;27:1078e85. [22] Arora G, Lee BH, Lamoureux M. Characteristic of enzyme profiles of Lactobacillus casei species by rapid API-ZYM system. J Dairy Sci 1990;73:264e73. [23] Terraf MCL, Tomas MSJ, Nader-Macıas MEF, Silva C. Screening of biofilm formation by beneficial vaginal lactobacilli and influence of culture media components. J Appl Microbiol 2012;113:1517e29. [24] Schoster A, Kokotovic B, Permin A, Pedersen PD, Dal Bello F, Guardabassi L. In vitro inhibition of Clostridium difficile and Clostridium perfringens by commercial probiotic strains. Anaerobe 2013;20:36e41. [25] Amann RI, Ludwig W, Schleifer KH. Phylogenetic identification and in situ detection of indivindual microbial cells without cultivation. Microbiol Rev 1995:143e69.

9

[26] Sghir A, Gramet G, Sau A, Rochet V, Pochart P, Dore J. Quantification of bacterial groups within human fecal flora by oligonucleotide probe hybridization. Appl Environ Microbiol 2000;66:2263e6. [27] Vankerckhoven V, Huys G, Vancanneyt M, Vael C, Klare I, Romod M, et al. Biosafety assesment of probiotics used in human consumption: recommendations from the EU PROSAFE project. Trends Food Sci Technol 2008;19:102e14. [28] Bizzini A, Jaton K, Romo D, Bille J, Prod’hom G, Greub G. Matrix-assisted laser desorption IonizationeTime of flight mass spectrometry as an alternative to 16S rRNA gene sequencing for identification of difficult-to-identify bacterial strains. J Clin Microbiol 2011;49(2):693e6. [29] Sauer S, Freiwald A, Maier T, Kube M, Reiherdt R, Kostrzewa M, et al. Classification and identification of bacteria mass spectrometry and computational analysis. PloS ONE 2008;3:2843e53. [30] Guglielmotii DM, Marcó MB, Golowczyb M, Treinherimer JA, Quiberoni AL. Probiotic potential of Lactobacillus delbrueckii strain and their phage resistant mutants. Int Dairy J 2007;17:916e25. [31] De Angelis M, Siragusa S, Berloco M, Caputo L, Settanni L, Alfonsi G, et al. Selection of potential probiotic Lactobacilli from pig feces to be used as additives in pelleted feeding. Res Microbiol 2006;157:792e801. [32] Wang CY, Lin PR, Ng CC, Shyu YT. Probiotic properties of Lactobacillus strains isolated from the feces of breast-fed infants and Taiwanese pickled cabbage. Anaerobe 2010;166:578e85. [33] Huang Y, Adams MC. In vitro assessment of the upper gastrointestinal tolerance of potential probiotic dairy propionibacteria. Int J Food Microbiol 2004;91:253e60. [34] Mainville I, Arcand Y, Farnworth ER. A dynamic model that simulates the human upper gastrointestinal tract for the study of probiotics. Int J Food Microbiol 2005;99:287e96. [35] Sieladie DV, Zambou NF, Kaktcham PM, Cresci A, Fonteh F. Probiotic properties of lactobacilli strains isolated from cow milk in the western highlands of cameroon. Innovat Rom Food Biotechnol 2011;9:12e28. [36] Mathur S, Singh R. Antibiotic resistance in food lactic acid bacteriaea review. Int J Food Microbiol 2005;105:281e95. [37] Ammor MS, Gueimonde M, Danielsen M, Zagorec M, van Hoek AHAM, de los Reyes-Gavilán C, et al. Two different tetracycline resistance mechanisms, plasmid-carried tet(L) and chromosomally located transposon-associated tet(M), coexist in Lactobacillus sakei Rits 9. Appl Environ Microbiol 2008;74: 1394e401. [38] Tenorio CM, Zarazaga C, Martinez C, Torres C. Bifunctional enzyme 60 -Naminoglycoside acetyltransferase-20 -O-aminoglycoside phosphotransferase in Lactobacillus and Pediococcus isolates of animal origin. J Clin Microbiol 2001;39:824e5. [39] Vakulenko SB, Mobashery S. Versatility of aminoglycosides and prospects for their future. Clin Microbiol Rev 2003;16:430e50.  oz R. Screening of biogenic amine [40] Landeta G, De las Rivas B, Carracosa AV, Mun production by coagulase-negative staphylococci isolated during industrial Spanish dry-cured ham processed. Meat Sci 2007;77:556e61. [41] Delgado S, O’Sullivan E, Fitzegrald G, Mayo B. Substractive screening for probiotic properties of Lactobacillus species from the human gastrointestinal tract in the search for new probiotics. J Food Sci 2007;72:310e5. [42] Delgado S, O’Sullivan E, Fitzegerald G, Mayo B. In vitro evaluation of the probiotic properties of human Bifidobacterium species and selection on new probiotic candidates. J Appl Microbiol 2008;104:1119e27. [43] Hussain M, Khan NT, Wajid A, Rassol SA. Technological characterization of indigenous enerococcal population for probiotic potential. Pak J Bot 2008;40: 867e75. [44] Monteagudo-Mera A, Caro I, Rodríguez-Aparicio LB, Rúa J, Ferrero MA, García-Armesto MR. Characterization of certain bacterial strain for potential use as starter or probiotic cultures in dairy products. J Food Prot 2011;74: 1379e86. [45] Rodríguez-Díaz J, Rubio-del-Campo A, Yebra MJ. Lactobacillus casei ferments the N-acetylglucosamine moiety of fucosyl-1,3-N-acetylglucosamine and excretes L-fucose. Appl Environ Microbiol 2012;78(13):4613e9. [46] Jones SE, Versalovic J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and antiinflammatory factors. BMC Microbiol 2009;9:35. [47] Martín R, Soberón N, Vaneechoutte M, Flórez AB, Vázquez F, Suárez JE. Characterization of indigenous vaginal lactobacilli from healthy women as probiotic candidates. Int Microbiol 2008;11:261e6. [48] Pompilio A, Piccolomini R, Picciani C, D’Antonio D, Savini V, Di Bonaventura G. Factors associated with adherence to and biofilm formation on polystyrene by Stenotrophomonas maltophilia: the role of cell surface hydrophobicity and motility. FEMS Microbiol Lett 2008;287:41e7. [49] Pompilio A, Crocetta V, Confalone P, Nicoletti M, Petrucca A, Guarnieri S, et al. Adhesion to and biofilm formation on IB3-1 bronchial cells by Stenotrophomonas maltophilia isolates from cystic fibrosis patients. BMC Microbiol 2010;10:102e5. [50] Rybtke MT, Jensen PO, Hoiby N, Givskov M, Tolker-Nielsen T, Bjarnsholt T. The implication of Pseudomonas aeruginosa biofilms in infections. Inflamm Allergy Drug Targets 2011;10(2):141e57.  áková D, Vlková E, Rada V, Kme&tcaron V. Aggregation of lactobacilli [51] Bujn and bifidobacteria with Escherichia coli O157. Folia Microbiol 2004;49: 143e6. [52] Falagas ME, Makris GC. Probiotic bacteria and biosurfactants for nosocomial infection control: a hypothesis. J Hosp Infect 2009;71:301e6.

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

10

D. Bujnakova et al. / Anaerobe xxx (2013) 1e10

[53] Kotikalapudi BL. Characterization and encapsulation of probiotic bacteria using a pea-protein alginate matrix. Master thesis. Canada: University of Saskatchewan; 2009. [54] Dalié DKD, Deschamps AM, Richard-Forget F. Lactic acid bacteria e potencial for control of mould growth and mycotoxins: a review. Food Control 2010;21: 370e80.

 a V, Wiese B, Nader- Macías ME. Growth and [55] Juárez Tomás MS, Ocan lactic acid production by vaginal Lactobacillus acidophilus CRL 1259, and inhibition of uropathogenic Escherichia coli. J Med Microbiol 2003;52: 1117e24.

Please cite this article in press as: Bujnakova D, et al., In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe (2013), http://dx.doi.org/10.1016/j.anaerobe.2013.10.009

In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves.

A total of 73 chicken and calves isolates were diagnosed using matrix-assisted laser desorption ionization-time-of flight mass spectrometry (Maldi-Tof...
1MB Sizes 0 Downloads 0 Views