Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Internalin AB-expressing recombinant Lactobacillus casei protects Caco-2 cells from Listeria monocytogenes-induced damages under simulated intestinal conditions

  • Moloko G. Mathipa,

    Roles Data curation, Formal analysis, Investigation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa, Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, Indiana, United States of America

  • Arun K. Bhunia,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Methodology, Project administration, Resources, Supervision, Validation, Writing – review & editing

    Affiliations Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, Indiana, United States of America, Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States of America, Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, United States of America

  • Mapitsi S. Thantsha

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Project administration, Supervision, Validation, Writing – review & editing

    mapitsi.thantsha@up.ac.za

    Affiliation Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa

Abstract

Background

Listeria monocytogenes is an intracellular foodborne pathogen that employs a number of strategies to survive challenging gastrointestinal conditions. It proliferates in the gut and subsequently causes listeriosis in high-risk individuals. Therefore, inhibition of its adherence to the intestinal receptors is crucial in controlling its infection. In this study, the effect of our previously developed recombinant Lactobacillus casei strain expressing invasion protein, Internalin AB of L. monocytogenes (LbcInlAB) on epithelial infection processes of the latter under simulated intestinal conditions was investigated.

Materials and methods

The confluent Caco-2 cell monolayer was pre-exposed to different L. casei strains at a multiplicity of exposure (MOE) of 10 for various periods before infection with L. monocytogenes at a multiplicity of infection (MOI) of 10 under simulated intestinal conditions. Subsequently, L. monocytogenes adhesion, invasion, and translocation, cytotoxicity and impact on tight junction integrity of the Caco-2 cells were analyzed.

Results

Under the simulated gastrointestinal condition, LbcInlAB showed a significant increase (p<0.0001) in adherence to, invasion and translocation through the Caco-2 cells when compared with the wild type strain. Although LbcInlAB strain exhibited enhanced inhibition of L. monocytogenes, it was not able to displace L. monocytogenes cells already attached to the monolayer. Additionally, pre-exposure to LbcInlAB reduced L. monocytogenes-mediated cytotoxicity and protected the tight junction barrier function.

Conclusion

The recombinant L. casei expressing InlAB shows potential for use as a prophylactic intervention strategy for targeted control of L. monocytogenes during the intestinal phase of infection.

Introduction

Listeria monocytogenes is abundant in nature and has proven to proliferate under various environmental conditions [1] including temperatures between -1.5 to 45°C and broad pH range of 4.0 to 9.6. It is well suited to survive in foods, and transit through the gastrointestinal tract. In immunocompromised individuals and pregnant women, it causes a disease generally referred to as listeriosis, while it results in self-limiting gastroenteritis in healthy individuals. L. monocytogenes employs different survival strategies in the challenging microenvironments of the gastrointestinal tract [24]. It enters the body through gastrointestinal mucosal surfaces to cause infection [5]. During disease progression, L. monocytogenes employs the Listeria adhesion protein (LAP), to aid adhesion and transmigration across the epithelial barrier in the gut [6,7]. It also uses the surface protein Internalin A (InlA) [8,9] and InlB [10,11] to attach to and gain entry into host cells for systemic spread. This invasion has also been shown to be associated with murine M cells both in vivo and in vitro [12].

Attenuated strains of foodborne pathogens such as L. monocytogenes have been used as vaccines for their control. However, such strains present the two most important risks. Firstly, the attenuated strain has a potential for reversion to its virulent phenotype post administration. Secondly, they can be virulent in partially immunocompetent (young infants; elderly) or immunocompromised individuals as they retain residual virulence [13]. These risks prompted an interest in a search for alternative strategies for pathogen control. Non-pathogenic transient bacteria in the digestive tract were then suggested as an alternative that can be used to substitute the attenuated pathogens [14,15]. Probiotics are candidates of choice as they have been reported to confer a health benefit on the host (FAO/WHO, 2002) [16] and to competitively inhibit foodborne pathogen infection [17,18]. Their advantages include tolerance to acid and bile salts allowing for survival, transition through the gastrointestinal tract (GIT) and colonization of the mucosal surface [19]. However, several studies have reported that probiotics sometimes fail to produce desirable effects. McCarthy et al. [20] reported the same levels of the anti-inflammatory cytokine TGF-β in untreated mice suffering from colitis and those to which probiotic Lactobacillus salivarius and Bifidobacterium infantis were orally administered. Koo et al. [21] reported inefficiency of wild type probiotic strains in preventing the attachment of L. monocytogenes to intestinal epithelial monolayers in vitro.

In an effort to enhance the effectiveness or antipathogenic effects of probiotics, bioengineering has been used as an alternative strategy [19,22], where a virulent gene of the specific pathogen is cloned and expressed to create a recombinant probiotic strain that is subsequently used to competitively exclude that pathogen. The bioengineered probiotics were reported by different researchers to possess enhanced antipathogenic effects when compared to their wild-type counterparts [21,23,24]. Chu et al. [23] reported the ability of a recombinant L. acidophilus strain carrying the K99 fimbriae from enterotoxigenic Escherichia coli (ETEC) to reduce attachment of ETEC to the porcine intestinal brush border in a dose-dependent manner. In another study, Wu et al. [25] cloned and expressed the heat stable (ST) and heat labile (LT) enterotoxins of ETEC under the nisin-inducible promoter into probiotic Lactobacillus reuteri. They reported that the recombinant strain successfully prevented enterotoxicity in a mouse model. Koo et al. [21] cloned and expressed the Listeria adhesion protein (LAP) from L. monocytogenes into probiotic L. paracasei and demonstrated that the recombinant strain was able to exclude Listeria colonization and epithelial cell damage in vitro. Depending on the virulence genes that are cloned and expressed, these genetically engineered probiotics can be used either as prophylactic or treatment alternatives for the specific pathogens. Due to promising results reported in these aforementioned studies, we also recently cloned and expressed the invasion operon inlAB into Lactobacillus casei (LbcInlAB) for the inhibition of L. monocytogenes adhesion, invasion and translocation in vitro, using the Caco-2 cells grown and maintained in the cell culture media, Dulbecco's Modified Eagle's Medium (DMEM) supplemented with fetal bovine serum (FBS) [26]. The resultant recombinant probiotic showed enhanced inhibition of L. monocytogenes compared to the wild type L. casei. Although the results were positive, they were still not appropriate for making inferences about how the recombinant L. casei would affect the L. monocytogenes intestinal infection phase, as the media used did not sufficiently simulate the intestinal conditions.

The Caco-2 cell line, a colon adenocarcinoma cell origin differentiates into the enterocyte-like cell and is a widely used model to assess intestinal permeability [27,28]. Different media such as the buffered salt solution, Hanks’ balanced salt solution (HBSS) buffered with HEPES (10 mM) at pH 7.4, and the DMEM containing amino acids and vitamins, both supplemented with glucose, have been commonly used in permeability studies. However, these media have been criticized for supporting cell growth, which renders them inappropriate models for the epithelial infection process [29,30]. This raised the need for alternative media that can address this shortcoming and thereby give a better simulation of the intestinal conditions. In order to address this, simulated intestinal fluids (SIF), namely, the Fed State SIF (FeSSIF) originally proposed for evaluation of drug dissolution kinetics [31], and Fasted State SIF (FaSSIF), were evaluated for their compatibility with Caco-2 monolayer. It was found that FeSSIF exhibited cytotoxicity to Caco-2 while FaSSIF was compatible with the Caco-2 monolayer [27,28,32]. Brouwers et al., [33] reported that data generated using FaSSIF were similar to that obtained with actual human intestinal aspirates collected in the fasted state. Hence, in order to determine the effect of the recombinant LbcInlAB on the epithelial infection process of L. monocytogenes in vitro, we used a Caco-2 cell culture model and FaSSIF as the medium.

Materials and methods

Bacterial strains, plasmids and growth conditions

Listeria monocytogenes F4244 (serovar 4b, epidemic strain), Lactobacillus casei (LbcWT), L. casei expressing InlAB (LbcInlAB) and LAP (LbcLAP, unpublished), and L. casei carrying the pLP401T empty vector (LbcV) were used in this study. Cloning and expression of inlAB gene of L. monocytogenes in L. casei (LbcInlAB) has been reported before [26] where the pLP401T vector [34] contains the origin of replication of Lactobacillus and the α-amylase promoter gene (Pamy). The promoter is mannitol inducible, therefore, modified MRS supplemented with 1% mannitol was used to induce the expression of InlAB in L. casei [21]. The vector also has an anchor peptide (117 aa) gene of L. casei and transcription terminator of the cbh (conjugated bile acid hydrolase) gene (Tcbh). The surface expression of InlAB on LbcInlAB is envisaged to covalently link to the cell wall/peptidoglycan via anchor peptide and LPxTG motif [26]. L. monocytogenes culturewas grown in Tryptone Soy broth supplemented with 0.6% yeast extract (TSA-YE) at 37°C for 18 h. LbcWT was grown in de Man Rogosa Sharpe (MRS) broth while LbcV and recombinant LbcInlAB and LbcLAP strains were grown anaerobically at 37°C for 16 h in MRS broth containing 2 μg/ml erythromycin.

Preparation of the FaSSIF

The fasted state simulated intestinal fluid (FaSSIF) was prepared as per Dressman et al., [35]. Briefly, 0.78 g potassium hydrogen phosphate (KH2PO4), 3.28 g of potassium chloride (KCl), 5 mM sodium taurocholate (representative bile salt) and 1.5 mM lecithin were suspended in 150 ml distilled water. The pH of the solution was adjusted to pH 6.8 with 1M NaOH or 1M HCl, and then its volume was made up to 200 ml with distilled water. The FaSSIF was sterilized by filtering through a 0.2 μm filter to avoid thermal denaturation of the media components. The FaSSIF was stored at 4°C and then used within 24 h post preparation.

Adhesion and invasion of Caco-2 cells by recombinant L. casei

Human colon adenocarcinoma cell line Caco-2 (HTB37; American Type Culture Collection) was cultured in Dulbecco’s modified eagle’s medium (DMEM with high glucose, HyCloneTM, GE, Logan, UT) supplemented with 10% Fetal Bovine Serum (FBS Atlanta Biologicals, GA) (D10F). The cells were grown in flasks (Greiner- Bio-One) for up to 10–12 days, trypsinized and then seeded in 12 well plates at a density of 1 × 105 cells/well. The plates were incubated at 37°C in the presence of 7% CO2 in a cell culture incubator for 10–12 days to allow for monolayer formation and cell differentiation (106 cells/ well) [21,36].

Overnight (18 h) bacteria pre-cultivated in their respective broths were washed twice with PBS, their absorbance adjusted to OD600 = 1 and then they were suspended in FaSSIF to a final concentration of 1 × 107 CFU/ml (MOE or MOI, 10). The Caco-2 cell monolayers were washed three times with DMEM and then exposed separately to the L. casei strains (LbcWT, LbcV, LbcInlAB or LbcLAP) and L. monocytogenes and incubated at 37ºC with 5% CO2 for 1 h [21]. Excess media were removed and the cell monolayers were washed three times with DMEM. To enumerate the adhered bacterial cells, cell monolayers were treated with 0.1% Triton X-100, incubated at 37ºC for 10 min. For the invasion assay, the monolayers were exposed to L. monocytogenes and L. casei and then washed as was done in the adhesion assay, treated with gentamycin (50 μg/ml, 1 h) and with 0.1% Triton X-100 (37ºC, 10 min). The lysed cell suspensions from both adhesion and invasion experiments were serially diluted in PBS before plating on MRS, MRS supplemented with erythromycin (2 μg/ml) and Modified Oxford (MOX) agar for LbcWT, recombinant L. casei, and L. monocytogenes, respectively. All the plates were incubated at 37ºC for 24–48 h before bacterial enumeration.

Determination of L. monocytogenes exclusion by L. casei

The competitive exclusion assay was done as per Koo et al. [21] with minor modifications. The absorbance of the bacterial cultures was adjusted to OD600 = 1 after they were washed twice with PBS, and then they were suspended in FaSSIF to a final concentration of 1 × 107 CFU/ml (MOI, 10). For competitive adhesion, L. monocytogenes was co-inoculated with each of the L. casei strains (LbcWT, LbcV, LbcInlAB or LbcLAP) to Caco-2 cell monolayer and incubated for 1 h. Adherent bacteria were enumerated as before.

In the inhibition of adhesion assay, the Caco-2 cell monolayers were first inoculated with each L. casei strain and incubated for 1 h. Unbound bacteria were removed by washing of the wells four times using DMEM. L. monocytogenes was then added to the wells and plates were incubated for one more hour. Adhered bacteria were released and plated as above. For displacement of adhesion, Caco-2 monolayers were first inoculated with L. monocytogenes and incubated for 1 h. Then unbound bacteria were washed off as in the inhibition of adhesion assay. L. casei strains were then added to the wells and plates incubated for another 1 h. Adhered bacteria were released and plated on MRS, MRS supplemented with 2 μg/ml of erythromycin and MOX agar plates for enumeration of LbcWT, recombinant L. casei, and L. monocytogenes, respectively [21].

Inhibition of L. monocytogenes adhesion and invasion by L. casei

Bacteria were pre-cultivated in their respective broths for 18 h. The bacterial cultures were washed twice with PBS after adjusting their absorbance to OD600 = 1, followed by their resuspension in FaSSIF to the final concentration of 1×107 CFU/ml (MOE/MOI, 10). The Caco-2 cell monolayers were washed and then exposed to the L. casei strains for 1, 4, 16 and 24 h at 37°C in the humidified incubator with 5% CO2. Excess medium in the wells containing unbound L. casei was removed and replaced with 500 μl of L. monocytogenes suspended in FaSSIF, and the plates incubated for 1 h at 37°C with 5% CO2. The cells were then washed thrice using DMEM. To enumerate the adhered bacterial cells, cell monolayers were treated with 0.1% Triton X-100, incubated at 37ºC for 10 min before plating onto the respective microbiological media as already mentioned.

For inhibition of L. monocytogenes invasion, the Caco-2 cell monolayers were washed three times with DMEM and then exposed to each L. casei strain for 1, 4, 16 and 24 h at 37°C with 5% CO2. Excess L. casei cells were removed and replaced with 500 μl of L. monocytogenes suspended in FaSSIF and then incubated for 1 h at 37°C with 5% CO2. To remove the non-adhered bacteria, the cell monolayers were washed three times with DMEM and then treated for 1 h with gentamycin (50 μg/ml). The invading bacterial counts were determined by plating as above.

Caco-2 cytotoxicity assay

To determine Caco-2 cytotoxicity induced by L. monocytogenes after pre-exposure to L. casei over time, we performed the LDH assay [21]. The supernatants after infection with L. monocytogenes for 1 h were collected and used to analyze for lactate dehydrogenase (LDH) enzyme release. Caco-2 cells that were treated with 500 μl of 0.1% Triton X-100 per well were used as positive control while those treated with DMEM were used as the negative control. From the supernatants collected, 100 μl were transferred to the 96-well flat bottom plate in triplicates and was analyzed using Pierce LDH cytotoxicity assay kit (Thermo Scientific, USA) following the protocol from the manufacturer.

Transcellular translocation of L. casei strains and subsequent inhibition of L. monocytogenes transepithelial translocation by recombinant L. casei

The Caco-2 cells were grown in 12 well trans-well filter inserts (3-μm pore size) for 20–25 days to reach confluence [6]. TEER of Caco-2 cells was quantified using the Millicell ERS system (Millipore, Billerica, MA) and a TEER value of more than 200 was used for all the experiments. Overnight (18 h) bacteria pre-cultivated in their respective broths were used. The bacterial cultures were washed twice with PBS and then suspended in FaSSIF (MOE, 10). For determining baseline translocation by L. casei or L. monocytogenes, the cell monolayers were washed three times with DMEM and then the bacteria were added separately to the apical wells, followed by incubation of microwell plates at 37°C with 5% CO2 for 2 h. The liquid from the basal well was collected, serially diluted in PBS and then plated for the enumeration of viable cells (CFU/ ml).

For the inhibition of L. monocytogenes translocation, L. casei strains were first added to the apical wells and incubated for 1, 4, 16 and 24 h at 37°C with 5% CO2. The liquid from the basal wells was collected, serially diluted in PBS and then plated onto MRS plates for enumeration of L. casei as described. Subsequently, excess L. casei cells were removed and replaced with 500 μl of L. monocytogenes suspended in FaSSIF (MOI, 10) and then incubated for 2 h at 37°C with 5% CO2. The liquid from the basal wells was removed and serially diluted in PBS and then plated on MOX plates for the enumeration L. monocytogenes.

Epithelial tight junction integrity analysis

Transepithelial electrical resistance (TEER) of Caco-2 cells was measured before and after the exposure to the bacteria using the Millicell ERS system (Millipore, Billerica, MA). Furthermore, we analyzed the epithelial tight junction integrity as per Koo et al. [21]. After exposure to L. monocytogenes, the tight junction permeability was analyzed using DextranFITC (Mr 3–5 kDa; Sigma) permeability through the transwell filter inserts. Fluorescence of the samples collected from the apical and basolateral chambers was read in a SpectraMax Gemini EM fluorescent plate reader (Molecular Devices; Sunnyvale, CA).

Statistical analysis

All data were analyzed using Prism 7 software (GraphPad Software Inc., United States), and significance was assigned at p < 0.05. Where appropriate, Tukey’s multiple comparisons, with p<0.005 as the significant difference was used to identify statistically significant differences.

Results

Adhesion, invasion and translocation profiles of Listeria monocytogenes (Lm) and Lactobacillus casei (Lbc) strains

Probiotics and foodborne pathogens colonize the gastrointestinal tract to exert beneficial effects or cause infection, respectively. It was therefore imperative that we determine how the expression of L. monocytogenes invasion genes by L. casei would affect its ability to adhere to, invade and translocate the Caco-2 cells under simulated intestinal conditions when compared to L. monocytogenes. Fig 1A depicts the adhesion profiles of the L. casei strains and L. monocytogenes to Caco-2 cells in simulated intestinal fluid. There were no statistically significant differences in the adhesion of L. monocytogenes F4244 versus LbcWT (p = 0.4436) or LbcV (p = 0.9914) to the Caco-2 cells, which showed adhesion percentages of 7%, 8%, and 7.8%, respectively. Conversely, recombinant L. casei strains expressing the different genes of L. monocytogenes adhered to Caco-2 cells at levels significantly higher than those of L. monocytogenes (p = 0.0002 for LbcInlAB vs L. monocytogenes and p <0.0001 for LbcLAP vs L. monocytogenes). It is worth noting, adhesion of LbcLAP was significantly higher than that of LbcInlAB (p = 0.0229).

thumbnail
Fig 1.

Adhesion (A), Invasion (B) and Translocation (C) of Listeria monocytogenes (Lm) and Lactobacillus casei (LbcWT, LbcV, LbcInlAB and LbcLAP) to Caco-2 cells. Percentages were calculated relative to the inocula that were added to the Caco-2 cells. Data are average (SD) of three independent experiments performed in duplicate. Error bars are standard deviations of averages of the three independent experiments. Bars marked with different letters (a, b, c) indicate significant difference at p<0.05.

https://doi.org/10.1371/journal.pone.0220321.g001

Invasion (Fig 1B) and translocation (Fig 1C) profiles of the L. casei strains and L. monocytogenes in simulated intestinal conditions were investigated. The strains LbcWT and LbcV displayed similar trends in invasion and translocation through the Caco-2 cells, both showing 0.08% and 0.13% for invasion and translocation, respectively. There was an increase in levels of both invasion and translocation by the recombinant L. casei strains (LbcInlAB and LbcLAP). LbcInlAB invaded and translocated through the Caco-2 cells at levels significantly higher compared to those of LbcWT and LbcV. Invasion and translocation levels for LbcLAP were not significantly different from those of LbcWT and LbcV (p<0.79), but significantly lower than those for LbcInlAB (p<0.0001). L. monocytogenes was able to invade and translocate the Caco-2 cell monolayer at significantly higher levels than all the L. casei strains. What was worth noting is that invasion and translocation of LbcInlAB through the Caco-2 cells was at significantly higher levels than all the other L. casei strains.

Mechanisms of L. monocytogenes exclusion by the L. casei strains

Probiotics employ competitive adhesion, inhibition or displacement mechanisms to inhibit or reduce adhesion of pathogens to the intestinal cells. In order to determine which mechanism is employed by L. casei strains (LbcWT, LbcV, LbcInlAB and LbcLAP) against L. monocytogenes, we evaluated each under simulated intestinal conditions (Fig 2). Adhesion of L. monocytogenes to Caco-2 cells in the absence of the L. casei strains was recorded as 100% in all the assays and was used to calculate the relative adhesion in the presence of the L. casei strains.

thumbnail
Fig 2. Competitive exclusion of Listeria monocytogenes (Lm) adhesion to Caco-2 cells by L. casei strains (LbcWT, LbcV, LbcInlAB and LbcLAP), analyzed by three different exclusion mechanisms.

(A) Competitive adhesion: Caco-2 cells were exposed to L. casei strains with Lm simultaneously, (B) Inhibition of adhesion: Caco-2 cells were pre-exposed to L. casei strains for 1 h before infection with Lm, and (C) Displacement of adhesion: Caco-2 cells were infected with Lm for 1 h before L. casei strains (1 h). Adhesion of Lm alone to Caco-2 cells was presented as 100% and percentage adhesion was calculated relative to that. Data are averages of three experiments run in duplicates. Error bars are standard deviations of averages of the three independent experiments. Bars marked with different letters (a, b, c, d) indicate significant difference at p<0.05.

https://doi.org/10.1371/journal.pone.0220321.g002

Fig 2A shows that adhesion of L. monocytogenes to Caco-2 cells was insignificantly reduced when it was co-inoculated with LbcWT and LbcV (p = 0.9941). We recorded reductions of 5.67% and 6% in adhesion of L. monocytogenes by LbcWT and LbcV, respectively. When co-inoculated with the recombinant strains (LbcInlAB and LbcLAP), there was a significant reduction (p< 0.0001) in the adhesion of L. monocytogenes. There was a 20.48% and 22.34% adhesion reduction by LbcInlAB and LbcLAP, respectively. Although both LbcInlAB and LbcLAP reduced the adhesion of L. monocytogenes, there was no statistical difference in their reduction levels (p = 0.2620).

In the inhibition of adhesion assay (Fig 2B), adhesion of L. monocytogenes to the Caco-2 cells was reduced by 2.92% and 3.05% due to their pre-exposure to LbcWT and LbcV, respectively. The reductions recorded were significant for both LbcWT (p = 0.0494) and LbcV (p = 0.0391), however, as expected, there was no significant difference when comparing the inhibition of L. monocytogenes adhesion by LbcWT vs. LbcV (p = 0.4588). Adhesion of L. monocytogenes was reduced by 18.9% and 14.4% due to pre-exposure of the Caco-2 cells to the recombinant strains LbcInlAB and LbcLAP, respectively. Interestingly, these recorded reduction levels were significantly higher when compared to the adhesion of L. monocytogenes alone (p<0.0001). Furthermore, there was a significant difference (p< 0.0033) in the reduction of adhesion between the two recombinant strains, with LbcLAP showing higher inhibition of adhesion than LbcInlAB. Significant differences (p<0.0001) were also obtained when comparing inhibition of adhesion by LbcWT or LbcV versus LbcInlAB or LbcLAP. In the displacement of adhesion experiment (Fig 2C), there were no significant differences in the adhesion of L. monocytogenes alone when compared to in the presence of any of the L. casei strains. Furthermore, there were no statistical differences among all the L. casei strains in the displacement of L. monocytogenes. Thus, the results show that inhibition of adhesion is the mechanism of competition used by the recombinant L. casei strains to reduce interaction of L. monocytogenes with Caco-2 cells.

Inhibition of L. monocytogenes adhesion, invasion, and translocation by L. casei over time

In order to determine how inhibition of L. monocytogenes adhesion to Caco-2 in FaSSIF will be influenced by duration of pre-exposure of the cell monolayer to L. casei strains, we investigated adhesion, invasion, and translocation of Caco-2 cells under simulated intestinal conditions by L. monocytogenes over a 24 h period. The effect of different exposure periods to L. casei strains on adhesion of L. monocytogenes to Caco-2 cells is presented in Fig 3A. Adhesion of L. monocytogenes was more reduced the longer the Caco-2 cells were pretreated with LbcWT, with reductions of 3.29%, 4.51%, and 12.96% recorded for 4, 16 and 24 h pre-exposure times, respectively. Significant reductions due to LbcWT were recorded after 4 h (p = 0.0007) and 16–24 h (p<0.0001). Improved reductions were obtained due to pre-exposure to recombinant L. casei strains, with reduced levels of 14.36% and 18.58% after 1 h, as well as of 57.66% and 61.52% after 24 h, recorded for LbcInlAB and LbcLAP, respectively. Contrary to what was observed for LbcWT, significant reductions in adhesion (p<0.0001) were obtained for LbcInlAB and LbcLAP for all exposure periods. Furthermore, even though prolonged exposure to either of the recombinant L. casei strains enhanced inhibition of adhesion, it was interesting to observe that pre-exposure to LbcLAP maintained significantly higher reduction levels than LbcInlAB throughout the 24 h (p<0.0001) period.

thumbnail
Fig 3.

Inhibition of Listeria monocytogenes (Lm) adhesion (A), invasion (B) and translocation (C) by L. casei strains (LbcWT, LbcInlAB and LbcLAP) over time. Caco-2 cells were pre-exposed to the L. casei strains for 1, 4, 16 and 24 h before infection with Lm for 1 h for adhesion and invasion and 2 h for translocation. Data are averages of three experiments run in duplicates. Error bars are standard deviations of averages of the three independent experiments. For each time point bars marked with different letters (a, b, c, d, e, f, g, h, I, j, k, l) indicate significant difference at p<0.05.

https://doi.org/10.1371/journal.pone.0220321.g003

Similar trends were observed for invasion (Fig 3B) and translocation (Fig 3C) of Caco-2 cells by L. monocytogenes subsequent to their prolonged pre-exposure to L. casei strains. Pre-exposure of the Caco-2 cells to LbcWT for 1 to 16 h showed no significant reduction of invasion (p = 0.3088); however, the 24-h exposure time resulted in a significant reduction (p<0.0001) (Fig 3B). Translocation of L. monocytogenes was significantly reduced (p<0.0001) by this strain from 1 h up to 24 h pre-exposure times (Fig 3C). No significant reduction in invasion was obtained due to pre-exposure of the Caco-2 cells to LbcInlAB or LbcLAP for 1–4 h, while the significant reduction (p<0.0001) was evident for 16 h to 24 h pre-exposure period to these strains. Similarly, when comparing the recombinant L. casei strains, there were no significant differences at 1 and 4 h pre-exposure, however, they exhibited significant differences after 16 and 24 h pre-exposure (p<0.0001). Pre-exposure to LbcInlAB and LbcLAP for 24 h showed significant (p<0.0001) reductions of L. monocytogenes invasion, with 48.96% and 32.22% reductions recorded for LbcInlAB and LbcLAP, respectively (Fig 3B).

For translocation assays (Fig 3C), after 24 h of pre-exposure, reductions of 17.81% and 15.67% were recorded for LbcInlAB and LbcLAP, respectively. Prolonged exposure of the Caco-2 cells to the recombinants showed an even significantly (p<0.0001) enhanced reduction of translocation. Overall, the results indicate that the longer the Caco-2 cells were pre-exposed to L. casei strains before their infection with L. monocytogenes, the more adhesion, invasion, and translocation of L. monocytogenes were reduced.

Cytotoxicity of L. monocytogenes to Caco-2 cells in the presence of L. casei strains

L. monocytogenes-mediated cytotoxicity to the Caco-2 was investigated (Fig 4) using the lactate dehydrogenase (LDH) assay. In the absence of L. casei strains, L. monocytogenes treatment for 1 h induced 70.25% cytotoxicity to Caco-2 cells. Pre-exposure of the cells to L. casei strains resulted in a reduction of cell cytotoxicity. L. monocytogenes induced 63.7% and 65.42% cytotoxicity levels after 1 h and 24 h, respectively, when Caco-2 cells were pre-exposed to LbcWT while 8.45% and 30.45% cytotoxicity levels were recorded after pre-exposure to LbcInlAB for 1 and 24 h, respectively (Fig 4). When the cells were pre-exposed to LbcLAP for 1 and 24 h, L. monocytogenes induced only 0.34% and 18.25% cytotoxicity levels for these exposure periods, respectively. These data indicate that recombinant L. casei strains provide significant protection (p<0.0001) against the cytotoxic effect of L. monocytogenes than LbcWT.

thumbnail
Fig 4. Cytotoxicity of Listeria monocytogenes (Lm) on Caco-2 cells pre-exposed to L. casei over time.

Cytotoxicity value for L. monocytogenes treatment in the absence of L. casei strains was 70.25%. Data are averages of three experiments run in duplicates. Error bars are standard deviations of averages of the three independent experiments. Bars marked with different letters (a, b, c, d, e, f) indicate significant difference at p<0.05.

https://doi.org/10.1371/journal.pone.0220321.g004

Epithelial tight junction integrity analysis

The integrity of the Caco-2 cells infected with L. monocytogenes alone or after their exposure to L. casei strains in the FaSSIF was measured using the transepithelial electrical resistance (TEER) (Fig 5) and DextranFITC (Fig 6) analyses. The results obtained for both analyses complemented those for cytotoxicity test. When the Caco-2 cells were pre-treated with L. casei strains for all exposure periods tested, there were lower TEER reduction changes than that of cells treated with L. monocytogenes alone (Fig 5). TEER reduction changes were also lower due to pre-exposure to recombinant L. casei strains than that due to their wild type counterpart. When comparing recombinant strains, TEER reductions were lower for LbcLAP than LbcInlAB. These results showed that under simulated intestinal conditions, recombinant L. casei strains protected the integrity of tight junctions between the Caco-2 cells, with LbcLAP showing better protection than LbcInlAB. However, as was also observed for cytotoxicity analysis, prolonged exposure of Caco-2 cells to the probiotics in FaSSIF had negative effects on Caco-2 cells as it resulted in higher TEER reductions when compared to shorter exposure periods. Nevertheless, even after 24 h, the TEER reductions for Caco-2 cells pre-exposed to L. casei strains were still lower than that of L. monocytogenes treatment alone.

thumbnail
Fig 5. Caco-2 cell permeability analysis using transepithelial electrical resistance (TEER) in simulated intestinal conditions.

TEER reduction by L. monocytogenes in the absence of L. casei strains was 20.5%. Bars marked with different letters (a, b, c, d, e, f, g, h) indicate significant difference at p<0.05, error bars are standard deviations of average TEER reductions of the three independent experiments.

https://doi.org/10.1371/journal.pone.0220321.g005

thumbnail
Fig 6. Tight junction integrity analysis using DextranFITC permeability assays.

DextranFITC recovery after exposure to L. monocytogenes alone was 3.72± 0.03%. Bars marked with different letters (a, b, c, d, e, f) indicate significant difference at p<0.05, error bars are standard deviations of average DextranFITC recovered from the three independent experiments.

https://doi.org/10.1371/journal.pone.0220321.g006

The results of the DextranFITC permeability analysis indicated that pre-treatment of the Caco-2 cells with L. casei strains reduced their permeability induced by L. monocytogenes since the amount of DextranFITC recovered from the basal chamber of the transwell plate was always higher for cells infected with L. monocytogenes alone than that for those pre-exposed to L. casei strains (Fig 6). Comparing the amount of dye recovered in the basal chamber for cells pre-exposed to L. casei strains, recombinants (LbcInlAB and LbcLAP) showed better protection than LbcWT, however, LbcLAP showed better protection than LbcInlAB. These differences are in agreement with those found in the inhibition of translocation and the TEER reduction tests, meaning that LbcLAP was better at protecting the integrity of Caco-2 cells under simulated intestinal conditions. The amount of dye recovered increased with an increase in the duration of exposure to L. casei strains, with levels higher after 24 h than after 2 h for all the strains. These results confirmed observations from the cytotoxicity and TEER reduction assays, which showed that prolonged exposure of Caco-2 cells to the L. casei strains in FaSSIF affected them negatively.

Discussion

There is a dire need for safe alternative methods for the prevention or treatment of foodborne diseases due to the ever-increasing development of microbial resistance to antibiotics. Inactivated or attenuated pathogens were initially used as vaccines; however, the risks associated with their use sparked an increased interest in using safe nonpathogenic bacteria as a substitute [15]. These risks include possible reversion of the attenuated pathogen to virulent phenotypes in hosts as well as the possibility for becoming virulent, especially in immunocompromised individuals [15,37]. Probiotics have been reported to offer beneficial health-promoting effects on the host and are generally regarded-as-safe (GRAS), attributes that predispose them as an attractive alternative [38]. The use of probiotics for inhibition of pathogens has been reported in the literature, however, with inconsistent outcomes whereby in certain cases, they have been reported to be less effective against some pathogens. In an effort to eliminate or lessen the limitations of conventional probiotics and enhance the efficacy of probiotics against pathogens, probiotic bioengineering, which is used to design and develop recombinant probiotic strains harboring certain virulence factors of the targeted pathogen, is currently being explored.

We used probiotic L. casei strain for bioengineering since it does not possess any virulent genes and has been used as probiotic before [39]. We used pLP401T as a vector to clone and express InlAB and this vector has been used before [21,40] and the recombinant L. casei strains exhibited desired beneficial effects compared to the wild type strain. Furthermore, the recombinant strains did not exhibit any cytotoxic response in the in vitro experiments [21,26] including this study suggesting these recombinant strains possibly does not have negative effect. However, animal studies are needed to fully validate their health beneficial effects. For successful application of recombinant probiotic strains, it is necessary to design and construct new suitable expression vectors that will not contain antibiotic markers for selection, preferably integrative vectors.

These genetically engineered probiotics, depending on the virulence genes they express and their mechanism of action can be used as either prophylactics or therapeutics for control of the specific pathogens. Although there have been concerns regarding probiotic bioengineering, it was reported that the probiotics retain their GRAS status even after the expression of heterologous genes [41].

During the infection process, L. monocytogenes employs the LAP for its adhesion and transcellular migration across the epithelial barrier [57] and invasion proteins, InlA and InlB to invade a wider range of mammalian cells [3]. Therefore, in order to construct a probiotic strain with an enhanced ability for targeted control of L. monocytogenes, we have recently cloned and expressed InlAB into L. casei which showed enhanced ability to adhere to, invade and translocate through Caco-2 cells in vitro compared to that of the wild type strain [26]. Internalins belong to a group of surface exposed leucine rich repeat proteins. They comprise of N-terminal cap domain and also harbour both an N-terminal signal peptide and a C-terminal LPxTG motif followed by a hydrophobic transmembrane region, marking them as extracellular proteins, covalently attached to the bacterial cell wall peptidoglycan [42]. Cloning of inlAB using pLP401T allowed anchoring and surface expression of InlAB on the recombinant LbcInlAB strain [26,34].

One shortcoming of our earlier study is that the experiments were performed in vitro using the Caco-2 cells grown and maintained in the standard cell culture media, DMEM supplemented with fetal bovine serum. In its disease progression, L. monocytogenes has to overcome diverse suboptimal microenvironments that usually constitute the host’s defense system [43] in order to colonize the host GI tract and cause infection. These conditions include but are not limited to low acid in the stomach and high bile concentration in the small intestine. In an effort to better understand the epithelial infection processes of L. monocytogenes as influenced by the recombinant L. casei strains, in the current study, we investigated the effect of LbcInlAB on the interaction between L. monocytogenes and Caco-2 cells in FaSSIF. Gamboa and Leong [44] reported that SIF has osmolality that is similar to that of human cells thus making this fluid a better medium to be used in in vitro intestinal model. The results in Fig 1 shows that expression of InlAB and LAP by the recombinant LbcInlAB and LbcLAP exhibited an enhanced adhesion (versus L. monocytogenes and LbcWT), invasion and translocation as opposed to LbcWT. The increased adhesion of recombinant strains when compared to L. monocytogenes is potentially due to high plasmid copy numbers in the recombinants. However, it was worth noting that LbcLAP showed a better adhesion capability as opposed to LbcInlAB, which exhibited superior invasion potentials instead. These results are in correlation with the function of the LAP and InlAB genes in the pathogen adhesion and invasion. Guimarães et al. [45] cloned and expressed the invasion gene InlA into Lactococcus lactis and reported that the recombinant showed an enhanced ability to invade epithelial cells. In a different study, Koo et al. [21] cloned and expressed the LAP into probiotic Lactobacillus paracasei and reported that the resultant recombinant strain exhibited enhanced adhesion to the Caco-2 cells. Although these studies reported the enhancement of probiotics through genetic engineering, the cells that they used were maintained in media that supported the growth of the epithelial cells, which is undesirable in such studies. In the current study, we used the FaSSIF, which does not stimulate the growth of epithelial cells, therefore presents a better simulation of the in vivo intestinal conditions when compared to the abovementioned studies.

Researchers elsewhere have investigated the intestinal phase of L. monocytogenes infection process in artificial gastrointestinal fluid broth systems [4648]. These studies reported on the behavior of L. monocytogenes when it was introduced on its own to these conditions. In the current study, we investigated the intestinal infection phase of L. monocytogenes in FaSSIF in the presence of L. casei strains. We looked at the ability of the L. casei strains to competitively exclude L. monocytogenes using three different mechanisms: competitive adhesion, inhibition, and displacement of adhesion, under simulated intestinal conditions (Fig 2). The results revealed that during competitive adhesion and inhibition of adhesion, adhesion of L. monocytogenes to Caco-2 cells was reduced by the L. casei strains, with recombinant LbcInlAB and LbcLAP exhibiting an enhanced reduction compared to LbcWT. Our results were in agreement with previous studies that reported on the competitive exclusion of pathogens. It has been shown that some probiotics share binding specificities with some pathogens [49,50], making it possible for direct competition between the probiotics with specific pathogens for receptor sites on the host cell [51]. Lee and Puong [52] reported that the inhibition of pathogens by probiotics could be due to the interaction of specific adhesins and receptors present in both probiotic and pathogen, affording the ability to compete for attachment to the same receptors. It has to be highlighted that although our study investigated the intestinal phase of L. monocytogenes infection processes in presence of recombinant L. casei strain, it still does not sufficiently simulate the intestinal conditions, specifically with regard to the presence of other intestinal microbiota, which may influence the results currently reported. An efficient platform for deciphering clearer conclusions pertaining to the actual effects of the recombinant L. casei strain on L. monocytogenes intestinal infection phase will be through the animal studies in these missing inhabitants of the GIT will be present. Converse to the results for competitive adhesion and inhibition of adhesion, all the L. casei strains failed to displace L. monocytogenes cells that had already adhered to Caco-2 cells similar to previous reports [53,54]. This observation negates the suitability of the recombinant L. casei strains as a therapeutic intervention for L. monocytogenes infection.

Previously, Barmpalia-Davis et al. [55] reported that artificial gastrointestinal conditions closely simulate the dynamics of GIT. Bernbom et al. [56] reported that in order to eliminate the influence of the indigenous microflora during pathogenesis studies and thereby simplify results interpretation, in vitro models of the intestinal system can be used. Taking these studies into consideration, we further examined how the presence of L. casei strains affected the different stages in the infection cycle of L. monocytogenes under simulated intestinal conditions over various exposure times (Fig 3). Prolonged exposure of the Caco-2 cells to L. casei strains showed enhanced inhibition of L. monocytogenes. Koo et al. [21] also reported that prolonged exposure of the Caco-2 cells to recombinant L. paracasei expressing LAP showed enhanced inhibition of L. monocytogenes adhesion, invasion, and translocation. Furthermore, it was worth noting that in all stages of infection the recombinant L. casei strains were better at inhibiting L. monocytogenes than LbcWT. Consequent to studying the inhibition of translocation, we also monitored the tight junction integrity using electrical resistance (Fig 5), and DextranFITC (Fig 6) permeability assays in the presence of L. casei strains. In agreement to the results observed for the inhibition of adhesion, invasion, and translocation, tight junction integrity of Caco-2 cells was better preserved in the presence of L. casei strains under simulated intestinal conditions. L. monocytogenes translocation has been reported to potentially occur in the stomach [57], the small intestine [6,5860] and the large intestine [61] in murine models. The enhanced protection of the tight junction by the recombinant L. casei strains in simulated intestinal conditions will result in a reduction of L. monocytogenes translocation, therefore, inhibiting Listeria infection. Our findings reported here are likely to be a closer reflection of the effects of the recombinant LbcInlAB on intestinal disease progression of L. monocytogenes in vivo. Nevertheless, this does not eliminate the need for animal studies.

In summary, the current study shows that the recombinant L. casei strain expressing InlAB can minimize the interaction of L. monocytogenes with the Caco-2 cells under simulated intestinal conditions. Additionally, it shows that different stages (adhesion, invasion, and translocation) of the L. monocytogenes infection cycle can be targeted depending on the virulence genes cloned and expressed. Future in vivo studies are recommended to confirm the purported effects of the L. casei strains under the actual conditions. These studies should address among others, safety issues associated with the use of the recombinant strains and investigate the apparent reduction of disease progression and/or disease severity in the presence of other GIT microorganisms.

Supporting information

S1 Table. Raw data for adhesion to Caco-2 cells.

https://doi.org/10.1371/journal.pone.0220321.s001

(XLSX)

S2 Table. Raw data for invasion of Caco-2 cells.

https://doi.org/10.1371/journal.pone.0220321.s002

(XLSX)

S3 Table. Raw data for translocation through Caco-2 cells.

https://doi.org/10.1371/journal.pone.0220321.s003

(XLSX)

S4 Table. Raw data for competitive adhesion of L. monocytogenes.

https://doi.org/10.1371/journal.pone.0220321.s004

(XLSX)

S5 Table. Raw data for inhibition of adhesion L. monocytogenes.

https://doi.org/10.1371/journal.pone.0220321.s005

(XLSX)

S6 Table. Raw data for displacement of adhesion L. monocytogenes.

https://doi.org/10.1371/journal.pone.0220321.s006

(XLSX)

S7 Table. Raw data for inhibition of adhesion over time.

https://doi.org/10.1371/journal.pone.0220321.s007

(XLSX)

S8 Table. Raw data for inhibition of invasion over time.

https://doi.org/10.1371/journal.pone.0220321.s008

(XLSX)

S9 Table. Raw data for inhibition of translocation over time.

https://doi.org/10.1371/journal.pone.0220321.s009

(XLSX)

S10 Table. Raw data for summary cytotoxicity, TEER and FITC.

https://doi.org/10.1371/journal.pone.0220321.s010

(XLSX)

Acknowledgments

The authors declare no conflict of interest. The research was supported in part by the funds from The National Research Foundation (grant # 102107) with the University of Pretoria for providing financial aid to MM to conduct research at Purdue University, the U.S. Department of Agriculture National Institute of Food and Agriculture (Hatch accession no. 1000028), Purdue AgSEED program and USDA Agricultural Research Service, under Agreement No. 59-8072-6-001. Any opinions, findings, conclusion, or recommendations expressed in this publication are those of the author(s) and do not necessarily reflect the view of the U.S. Department of Agriculture.

References

  1. 1. Smith JL, Liu Y, Paoli GC (2012) How does Listeria monocytogenes combat acid conditions? Can J Miccrobiol 59: 141–152.
  2. 2. Horn N, Bhunia AK (2018) Food-Associated Stress Primes Foodborne Pathogens for the Gastrointestinal Phase of Infection. Front Microbiol 9: 1962. pmid:30190712
  3. 3. Hamon M, Bierne H, Cossart P (2006) Listeria monocytogenes: a multifaceted model. Nat Rev Microbiol 4: 423–434. pmid:16710323
  4. 4. Schuppler M, Loessner MJ (2010) The opportunistic pathogen Listeria monocytogenes: pathogenicity and interaction with the mucosal immune system. Int J Inflamation 2010: 704321.
  5. 5. Drolia R, Bhunia AK (2019) Crossing the intestinal barrier via Listeria adhesion protein and internalin A. Trends Microbiol 27: 408–425. pmid:30661918
  6. 6. Drolia R, Tenguria S, Durkes AC, Turner JR, Bhunia AK (2018) Listeria adhesion protein induces intestinal epithelial barrier dysfunction for bacterial translocation. Cell Host & Microbe 23: 470–484.
  7. 7. Jagadeesan B, Koo OK, Kim KP, Burkholder KM, Mishra KK, Aroonnual A, et al. (2010) LAP, an alcohol acetaldehyde dehydrogenase enzyme in Listeria promotes bacterial adhesion to enterocyte-like Caco-2 cells only in pathogenic species. Microbiology 156: 2782–2795. pmid:20507888
  8. 8. Gaillard JL, Berche P, Frehel C, Gouin E, Cossart P (1991) Entry of L. monocytogenes into cells is mediated by internalin, a repeat protein reminiscent of surface-antigens from Gram-positive cocci. Cell 65: 1127–1141. pmid:1905979
  9. 9. Mengaud J, Ohayon H, Gounon P, Mege RM, Cossart P (1996) E-cadherin is the receptor for internalin, a surface protein required for entry of L. monocytogenes into epithelial cells. Cell 84: 923–932. pmid:8601315
  10. 10. Braun L, Ohayon H, Cossart P (1998) The InlB protein of Listeria monocytogenes is sufficient to promote entry into mammalian cells. Mol Microbiol 27: 1077–1087. pmid:9535096
  11. 11. Chiba S, Nagai T, Hayashi T, Baba Y, Nagai S, Koyasu S (2011) Listerial invasion protein internalin B promotes entry into ileal Peyer's patches in vivo. Microbiol Immunol 55: 123–129. pmid:21204945
  12. 12. Jensen VB, Harty JT, Jones BD (1998) Interactions of the invasive pathogens Salmonella typhimurium, Listeria monocytogenes, and Shigella flexneri with M cells and murine Peyer's patches. Infect Immun 66: 3758–3766. pmid:9673259
  13. 13. Mercenier A, Muller-Alouf H, Grangette C (2000) Lactic acid bacteria as live vaccines. Curr Issues Mol Biol 2: 17–26. pmid:11464916
  14. 14. Marelli B, Perez AR, Banchio C, de Mendoza D, Magni C (2011) Oral immunization with live Lactococcus lactis expressing rotavirus VP8* subunit induces specific immune response in mice. J Virol Methods 175: 28–37. pmid:21530589
  15. 15. Tarahomjoo S (2012) Development of vaccine delivery vehicles based on lactic acid bacteria. Mol Biotechnol 51: 183–199. pmid:21901278
  16. 16. FAO/WHO (2002) Guidelines for the evaluation of probiotics in food. London, Ontario.
  17. 17. Behnsen J, Deriu E, Sassone-Corsi M, Raffatellu M (2013) Probiotics: properties, examples, and specific applications. Cold Spring Harbor Perspect Biol 3: a010074.
  18. 18. Amalaradjou MAR, Bhunia AK (2012) Modern approaches in probiotics research to control foodborne pathogens. Adv Food Nutr Res 67: 185–239. pmid:23034117
  19. 19. Amalaradjou MAR, Bhunia AK (2013) Bioengineered probiotics, a strategic approach to control enteric infections. Bioengineered 4: 291–299.
  20. 20. McCarthy J, O’mahony L, O’callaghan L, Sheil B, Vaughan EE, Fitzsimons N, et al. (2003) Double blind, placebo controlled trial of two probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. Gut 52: 975–980. pmid:12801954
  21. 21. Koo OK, Amalaradjou MAR, Bhunia AK (2012) Recombinant probiotic expressing Listeria adhesion protein attenuates Listeria monocytogenes virulence in vitro. PLoS One 7: e29277. pmid:22235279
  22. 22. Sola-Oladokun B, Culligan EP, Sleator RD (2017) Engineered Probiotics: Applications and Biological Containment. Annu Rev Food Sci Technol 8: 353–370. pmid:28125354
  23. 23. Chu H, Kang S, Ha S, Cho K, Park SM, Han KH, et al. (2005) Lactobacillus acidophilus expressing recombinant K99 adhesive fimbriae has an inhibitory effect on adhesion of enterotoxigenic Escherichia coli. Microbiol Immunol 49: 941–948. pmid:16301804
  24. 24. Sánchez B, López P, González-Rodríguez I, Suárez A, Margolles A, Urdaci MC (2011) A flagellin-producing Lactococcus strain: interactions with mucin and enteropathogens. FEMS Microbiol Lett 318: 101–107. pmid:21323981
  25. 25. Wu C-M, Chung T-C (2007) Mice protected by oral immunization with Lactobacillus reuteri secreting fusion protein of Escherichia coli enterotoxin subunit protein. FEMS Immunol Med Microbiol 50: 354–365. pmid:17651125
  26. 26. Mathipa MG, Thantsha MS, Bhunia AK (2019) Lactobacillus casei expressing Internalins A and B reduces Listeria monocytogenes interaction with Caco‐2 cells in vitro. Microb Biotechnol 12: 715–7129. pmid:30989823
  27. 27. Ingels F, Deferme S, Destexhe E, Oth M, Van den Mooter G, Augustijns P (2002) Simulated intestinal fluid as transport medium in the Caco-2 cell culture model. Int J Pharm 232: 183–192. pmid:11790502
  28. 28. Fossati L, Dechaume R, Hardillier E, Chevillon D, Prevost C, Bolze S, et al. (2008) Use of simulated intestinal fluid for Caco-2 permeability assay of lipophilic drugs. Int J Pharm 360: 148–155. pmid:18539418
  29. 29. Kapitza SB, Michel BR, van Hoogevest P, Leigh MLS, Imanidis G (2007) Absorption of poorly water soluble drugs subject to apical efflux using phospholipids as solubilizers in the Caco-2 cell model. Eur J Pharm Biopharm 66: 146–158. pmid:17071065
  30. 30. Lind ML, Jacobsen J, Holm R, Müllertz A (2007) Development of simulated intestinal fluids containing nutrients as transport media in the Caco-2 cell culture model: assessment of cell viability, monolayer integrity and transport of a poorly aqueous soluble drug and a substrate of efflux mechanisms. Eur J Pharm Sci 32: 261–270. pmid:17890067
  31. 31. Galia E, Nicolaides E, Hörter D, Löbenberg R, Reppas C, Dressman JB (1998) Evaluation of various dissolution media for predicting in vivo performance of class I and II drugs. Pharm Res 15: 698–705. pmid:9619777
  32. 32. Ingels F, Beck B, Oth M, Augustijns P (2004) Effect of simulated intestinal fluid on drug permeability estimation across Caco-2 monolayers. Int J Pharm 274: 221–232. pmid:15072798
  33. 33. Brouwers J, Tack J, Lammert F, Augustijns P (2006) Intraluminal drug and formulation behavior and integration in in vitro permeability estimation: a case study with amprenavir. J Pharm Sci 95: 372–383. pmid:16374852
  34. 34. Pouwels PH, Vriesema A, Martinez B, Tielen FJ, Seegers JF, Leer RJ, et al. (2001) Lactobacilli as vehicles for targeting antigens to mucosal tissues by surface exposition of foreign antigens. Methods Enzymol 336: 369–389. pmid:11398413
  35. 35. Dressman JB, Amidon GL, Reppas C, Shah VP (1998) Dissolution testing as a prognostic tool for oral drug absorption: immediate release dosage forms. Pharm Res 15: 11–22. pmid:9487541
  36. 36. Natoli M, Leoni BD, D’Agnano I, Zucco F, Felsani A (2012) Good Caco-2 cell culture practices. Toxicol In Vitro 26: 1243–1246. pmid:22465559
  37. 37. Nabel GJ (2013) Designing Tomorrow's Vaccines. N Engl J Med 368: 551–560. pmid:23388006
  38. 38. Wyszyńska A, Kobierecka P, Bardowski J, Jagusztyn-Krynicka EK (2015) Lactic acid bacteria—20 years exploring their potential as live vectors for mucosal vaccination. Appl Microbiol Biotechnol 99: 2967–2977. pmid:25750046
  39. 39. Jacouton E, Chain F, Sokol H, Langella P, Bermúdez-Humarán LG (2017) Probiotic strain Lactobacillus casei BL23 prevents colitis-associated colorectal cancer. Front Immunol 8.
  40. 40. Kajikawa A, Igimi S (2009) Reduction of tumor necrosis factor alpha-inducing capacity of recombinant Lactobacillus casei via expression of Salmonella OmpC. Appl Environ Microbiol 75: 2727–2734. pmid:19270120
  41. 41. Kumar M, Yadav AK, Verma V, Singh B, Mal G, Nagpal R, et al. (2016) Bioengineered probiotics as a new hope for health and diseases: an overview of potential and prospects. Future Microbiol 11: 585–600. pmid:27070955
  42. 42. Cossart P, Pizarro-Cerdá J, Lecuit M (2003) Invasion of mammalian cells by Listeria monocytogenes: functional mimicry to subvert cellular functions. Trends Cell Biol 13: 23–31. pmid:12480337
  43. 43. Gahan C, Hill C (2014) Listeria monocytogenes: survival and adaptation in the gastrointestinal tract. Front Cell Infect Microbiol 4: 9. pmid:24551601
  44. 44. Gamboa JM, Leong KW (2013) In vitro and in vivo models for the study of oral delivery of nanoparticles. Adv Drug Delivary Rev 65: 800–810.
  45. 45. Guimaraes VD, Gabriel JE, Lefevre F, Cabanes D, Gruss A, Cossart P, et al. (2005) Internalin-expressing Lactococcus lactis is able to invade small intestine of guinea pigs and deliver DNA into mammalian epithelial cells. Microbes Infect 7: 836–844. pmid:15878681
  46. 46. Begley M, Gahan CGM, Hill C (2002) Bile stress response in Listeria monocytogenes LO28: adaptation, cross-protection, and identification of genetic loci involved in bile resistance. Appl Environ Microbiol 68: 6005–6012. pmid:12450822
  47. 47. King T, Ferenci T, Szabo EA (2003) The effect of growth atmosphere on the ability of Listeria monocytogenes to survive exposure to acid, proteolytic enzymes and bile salts. Int J Food Microbiol 84: 133–143. pmid:12781937
  48. 48. Formato G, Geornaras I, Barmpalia IM, Skandamis PN, Belk KE, Scanga JA, et al. (2007) Effect of acid adaptation on growth during storage at 10 C and resistance to simulated gastric fluid of Listeria monocytogenes inoculated onto bologna formulated with or without antimicrobials. J Food Prot 70: 65–69. pmid:17265862
  49. 49. Fujiwara S, Hashiba H, Hirota T, Forstner JF (2001) Inhibition of the binding of enterotoxigenic Escherichia coli Pb176 to human intestinal epithelial cell line HCT-8 by an extracellular protein fraction containing BIF of Bifidobacterium longum SBT2928: suggestive evidence of blocking of the binding receptor gangliotetraosylceramide on the cell surface. Int J Food Microbiol 67: 97–106. pmid:11482574
  50. 50. Neeser JR, Granato D, Rouvet M, Servin A, Teneberg S, Karlsson KA (2000) Lactobacillus johnsonii La1 shares carbohydrate-binding specificities with several enteropathogenic bacteria. Glycobiology 10: 1193–1199. pmid:11087711
  51. 51. Collado MC, Gueimonde M, Salminen S (2010) Probiotics in adhesion of pathogens: mechanisms of action. Bioactive Foods in Promoting Health: Elsevier. pp. 353–370.
  52. 52. Lee YK, Puong KY (2002) Competition for adhesion between probiotics and human gastrointestinal pathogens in the presence of carbohydrate. Brit J Nutr 88: S101–S108. pmid:12215184
  53. 53. Lee Y-K, Puong K-Y, Ouwehand AC, Salminen S (2003) Displacement of bacterial pathogens from mucus and Caco-2 cell surface by lactobacilli. J Med Microbiol 52: 925–930. pmid:12972590
  54. 54. Gueimonde M, Jalonen L, He F, Hiramatsu M, Salminen S (2006) Adhesion and competitive inhibition and displacement of human enteropathogens by selected lactobacilli. Food Res Int 39: 467–471.
  55. 55. Barmpalia-Davis IM, Geornaras I, Kendall PA, Sofos JN (2008) Differences in survival among 13 Listeria monocytogenes strains in a dynamic model of the stomach and small intestine. Appl Environ Microbiol 74: 5563–5567. pmid:18586963
  56. 56. Bernbom N, Licht TR, Brogren CH, Jelle B, Johansen AH, Badiola I, et al. (2006) Effects of Lactococcus lactis on composition of intestinal microbiota: Role of nisin. Appl Environ Microbiol 72: 239–244. pmid:16391049
  57. 57. Conlan JW (1997) Critical roles of neutrophils in host defense against experimental systemic infections of mice by Listeria monocytogenes, Salmonella Typhimurium, and Yersinia enterocolitica. Infect Immun 65: 630–635. pmid:9009323
  58. 58. Macdonald TT, Carter PB (1980) Cell-mediated immunity to intestinal infection. Infect Immun 28: 516–523. pmid:6772561
  59. 59. Marco AJ, Prats N, Ramos JA, Briones V, Blanco M, Dominguez L, et al. (1992) A microbiological, histopathological and immunohistological study of the intragastric inoculation of Listeria monocytogenes in mice. J Comp Pathol 107: 1–9. pmid:1430342
  60. 60. Pron B, Boumaila C, Jaubert F, Sarnacki S, Monnet J, Berche P, et al. (1998) Comprehensive study of the intestinal stage of listeriosis in a rat ligated ileal loop system. Infect Immun 66: 747–755. pmid:9453636
  61. 61. Nishikawa S, Hirasue M, Miura T, Yamada K, Sasaki S, Nakane A (1998) Systemic dissemination by intrarectal infection with Listeria monocytogenes in mice. Microbiol Immunol 42: 325–327. pmid:9623921