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

Cell-Free Culture Supernatant of Bifidobacterium breve CNCM I-4035 Decreases Pro-Inflammatory Cytokines in Human Dendritic Cells Challenged with Salmonella typhi through TLR Activation

  • Miriam Bermudez-Brito,

    Affiliation Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, Granada, Spain

  • Sergio Muñoz-Quezada,

    Affiliation Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, Granada, Spain

  • Carolina Gomez-Llorente,

    Affiliation Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, Granada, Spain

  • Esther Matencio,

    Affiliation Global Centre for Child Nutrition Technology, Hero Group, Alcantarilla, Murcia, Spain

  • Maria J. Bernal,

    Affiliation Global Centre for Child Nutrition Technology, Hero Group, Alcantarilla, Murcia, Spain

  • Fernando Romero,

    Affiliation Global Centre for Child Nutrition Technology, Hero Group, Alcantarilla, Murcia, Spain

  • Angel Gil

    agil@ugr.es

    Affiliation Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, Granada, Spain

Abstract

Dendritic cells (DCs) constitute the first point of contact between gut commensals and our immune system. Despite growing evidence of the immunomodulatory effects of probiotics, the interactions between the cells of the intestinal immune system and bacteria remain largely unknown. Indeed,, the aim of this work was to determine whether the probiotic Bifidobacterium breve CNCM I-4035 and its cell-free culture supernatant (CFS) have immunomodulatory effects in human intestinal-like dendritic cells (DCs) and how they respond to the pathogenic bacterium Salmonella enterica serovar Typhi, and also to elucidate the molecular mechanisms involved in these interactions. Human DCs were directly challenged with B. breve/CFS, S. typhi or a combination of these stimuli for 4 h. The expression pattern of genes involved in Toll-like receptor (TLR) signaling pathway and cytokine secretion was analyzed. CFS decreased pro-inflammatory cytokines and chemokines in human intestinal DCs challenged with S. typhi. In contrast, the B. breve CNCM I-4035 probiotic strain was a potent inducer of the pro-inflammatory cytokines and chemokines tested, i.e., TNF-α, IL-8 and RANTES, as well as anti-inflammatory cytokines including IL-10. CFS restored TGF-β levels in the presence of Salmonella. Live B.breve and its supernatant enhanced innate immune responses by the activation of TLR signaling pathway. These treatments upregulated TLR9 gene transcription. In addition, CFS was a more potent inducer of TLR9 expression than the probiotic bacteria in the presence of S. typhi. Expression levels of CASP8 and IRAK4 were also increased by CFS, and both treatments induced TOLLIP gene expression. Our results indicate that the probiotic strain B. breve CNCM I-4035 affects the intestinal immune response, whereas its supernatant exerts anti-inflammatory effects mediated by DCs. This supernatant may protect immune system from highly infectious agents such as Salmonella typhi and can down-regulate pro-inflammatory pathways.

Introduction

Probiotic bacteria including lactobacilli and bifidobacteria are part of a normal intestinal microbiota in humans and generally considered as potentially beneficial to various aspects of host metabolism [1]. Bifidobacterium sp. are among the most relevant probiotic microorganisms because they colonize the intestinal tract soon after birth, are present at high levels in the guts of infants and adults and promote beneficial effects on intestinal ecology and immune responses [2], [3]. Several mechanisms for the favorable influence of probiotic bacteria on the intestinal mucosa have been suggested including the secretion of antimicrobial products, resistance to pathogen colonization, barrier function enhancement and maintenance, modulation of epithelial cell signal transduction and innate and adaptive immunomodulation [3], [4]. The beneficial effects of specific probiotic strains have been established for the treatment and prevention of many diseases [5], including diarrhea [6], the alleviation of lactose intolerance [7] and postoperative complications [8], antimicrobial [9] and anticolorectal cancer activity [10], [11] and for reducing irritable bowel symptoms [12] and increasing the relapse time for some inflammatory bowel diseases [13].

The probiotic properties of commensal bacteria including lactobacilli and bífidobacteria are likely to be determined at least in part by their effects on dendritic cells (DCs) [1], a complex, heterogeneous group of multifunctional antigen-presenting cells (APCs) that comprise a critical arm of the immune system [14], [15]. These cells play a critical role in the orchestration of the adaptive immune response by inducing tolerance and adaptive immunity. Understanding the direct interaction between commensal bacteria and DCs is particularly important to know how the immune system of the gut is locally able to distinguish these bacteria from pathogens and to elicit a tolerogenic response [16]. The primary response to these bacteria is triggered by the innate pattern recognition receptors (PRRs), which bind pathogen-associated molecular patterns (PAMPs). PRRs comprise Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), adhesion molecules and lectins [4], [17]. The binding of microbe-associated molecules to these receptors can activate APCs and initiate a signaling transduction cascade that leads to the release of cytokines and initiation of the acquired immune response [18].

Mucosal DCs appear to have unique properties that distinguish them from peripheral DCs [19]. However, to date, probiotic activity has often been tested in monocyte-derived DCs (MoDCs) or murine DCs, which are quite different from human gut DCs [20]. For this reason, in this study, we used intestinal-like human DCs that were developed from umbilical cord blood CD34+ progenitor cells. These human DCs are Langerhans-like cells that extend dendritic processes and sample antigens similarly to the lamina propria DCs in the gut that sample luminal antigens [21].

We have previously reported some of the probiotic properties of Bifidobacterium breve CNCM I-4035, a novel bifidobacteria strain isolated from the feces of newborns that were exclusively breast-fed [22], [23]. In the present work, we studied the immunomodulatory effects of B.breve CNCM I-4035 and its cell-free culture supernatant (CFS) on human intestinal-like DCs and how the treated DCs interact and respond to the pathogenic bacteria Salmonella enterica serovar Typhi at molecular level.

Materials and Methods

Ethic Statement

The ethical Committee of Granadás University approved this study.

Bifidobacterium breve was obtained from feces of breast-fed newborns, in a previous work [24]. Briefly, 12 healthy, exclusively breast-fed infants, aged 1 month, were selected for the study at the Clinic Hospital of the University of Granada. This study was conducted according to the guidelines laid down in the Declaration of Helsinki and all procedures involving human subjects were approved by the Ethical Committee of the University of Granada. Written informed consent was obtained from the parents after a careful explanation of the nature of the study.

Preparation of bacteria and cell-free culture supernatant

B. breve CNCM I-4035was isolated from the feces of breast-fed newborns and previously selected for its in vitro probiotic characteristics [22], [23]. B. breve CNCM I-4035 was routinely anaerobically cultured for 24 hours at 37°C in de Man-Rogosa-Sharpe (MRS) broth medium (Oxoid, Basingstoke, United Kingdom) supplemented with 0.05% (wt/vol) cysteine (Sigma-Aldrich, St. Louis, MO) to promote the growth of B. breve. The supernatant of the culture medium was collected by centrifugation at 12,000×g for 10 min, neutralized to pH 7.0 by the addition of 1 N NaOH and concentrated ten-fold by lyophilization. The supernatants were passed through a 0.22-μm pore size filter unit (Minisart hydrophilic syringe filter; Sartorius Stedim Biotech GmbH, Goettingen, Germany) and stored at −20°C until use. The supernatant was added to the DC culture medium at a concentration of 7% v/v.

Salmonella enterica serovar Typhi CECT 725 was provided by the Spanish Type Culture Collection (CECT; Burjassot, Spain) and aerobically cultured in tryptone soy broth (Panreac Química, Barcelona, Spain).

For experiments, S. typhi was cultured for 8 h at 37°C in tryptone soy broth and then subcultured 1:500 in RPMI 1640 medium (Sigma-Aldrich) containing 10% fetal bovine serum (FBS; Gibco Invitrogen, Paisley, United Kingdom) at 37°C overnight.

Cell preparation

DCs generated from umbilical cord blood CD34+ progenitor cells (hematopoietic stem cells) were supplied by MatTek Corporation (Ashland, MA). These cells were seeded in 24-well plates in DC maintenance medium (DC-MM; MatTek) containing cytokines and antibiotics.

Bacterial co-culture and DC stimulation

Cell cultures were seeded in 24-well plates at a density of 2×105 DCs/well. For incubations, DC-MM was replaced with RPMI-1640 medium. DCs were co-incubated with B. breve CNCM I-4035 bacteria (107 CFU/ml) or CFS as well as S. typhi (106 CFU/ml) or a combination of these treatments for 4 h at 37°C in a 5% CO2/95% air atmosphere. After incubation, the DCs were washed with PBS, and DC-MM (containing cytokines and antibiotics) was added to the wells and incubated for an additional 20 h. Cell supernatants and cells were collected for cytokine analysis and RNA extraction, respectively. Escherichia coli lipopolysaccharide (LPS; Sigma-Aldrich) was applied at a concentration of 20 ng/ml as a positive control. Negative-control cultures contained unstimulated DCs.

Cytokine and chemokine quantification in culture supernatants

Cytokine production was measured by immunoassay with the MILLIplexTM kit (Linco Research Inc., MO) using the Luminex 200 system according to the manufacturer's instructions. IL-1β, IL-6, IL-8, IL-10, IL-12(p40), IL-12(p70), TNF-α, IFN-γ, MCP-1/CCL2, MIP-1α/CCL3, RANTES/CCL5, MDC/CCL22 and TGF-β were analyzed. We performed three independent experiments.

Reverse transcriptase (RT) reaction and polymerase chain reaction (PCR)

Total RNA was isolated from cells using the RNAqueous Kit (Ambion, Paisley, United Kingdom) and additional Turbo DNase treatment (Ambion) according to the manufacturer's recommendations. The RNA quality was verified using a Model 2100 Bioanalyzer (Agilent, Santa Clara, USA), and the RNA concentration was determined using a Rediplate 96 Ribogreen RNA Quantitation Kit (Gibco, Invitrogen). The total RNA was reversed-transcribed using an RT2 First-strand Kit (SABiosciences Corporation, Frederick, MD). Real-time PCR was performed using an RT2 Real-time PCR SYBR Green/ROX Kit (SABiosciences) on an ABI Prism 7500 sequence detector (Applied Biosystems, Foster City, CA). Real-time RT-PCR analysis of the samples was performed using a Human TLR Signaling Pathway PCR Array (SABiosciences), which includes primer pairs specific for the following 20 genes related to TLR-mediated signaling pathways: TLR1, TLR2, TLR3, TLR4, TLR5, TLR9, MYD88, TNF-α, IRAK-1, IRAK-4, TOLLIP, CASP8, IL-10, TAK-1, JNK, NFKBIA, NFKB-1, TBK-1, MAPK14 and IRF-3. The housekeeping gene GAPDH was used as a control. The thermal profile for all reactions was: 1 cycle of 95°C for 10 min and 40 cycles of 95°C for 15 s and 60°C for 1 min. The expression levels of the target genes were normalized to those of untreated DCs (control).

Statistical analysis

The results shown are the mean ± SEM of three independent experiments.

The differences in cytokine levels and gene expression between treatments were compared using the Mann-Whitney U-test. Analyses were performed using NCSS 2007 software (Kaysville, UT). P<0.05 was considered significant and is indicated with an asterisk in the figures.

Differences between DCs treated with Salmonella and Salmonella plus B. breve CNCM I-4035 or its CFS were also evaluated. P<0.05 was considered to significant and is indicated in the figures with a pound sign (#).

Results

Supernatant of B. breve CNCM I-4035 decreases cytokine release in human DCs co-cultured with S. typhi

The addition of pathogenic bacteria (S. typhi CECT 725) or LPS to DCs markedly affected the expression of pro-inflammatory cytokines (Figures 1 and 2). These treatments lead to a strong secretion of IL-1β, IL-6, IL-8, IL-12p40 and TNF-α compared to the controls. Accordingly, as illustrated in figure 3, the release of MCP-1, MIP-1α, RANTES and MDC to the culture medium was significantly elevated by either Salmonella or LPS stimulation.

thumbnail
Figure 1. Effects of B. breve CNCM I-4035 and its cell-free culture supernatant on the secretion of pro-inflammatory cytokines by intestinal-like human dendritic cells.

Dendritic cells (DCs) were incubated for 4 h with the B. breve CNCM I-4035 probiotic (Prob) or its cell-free supernatant (CFS), Salmonella (Sal) or both and further incubated for 20 h in medium containing antibiotics. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. Negative-control cultures contained unstimulated DCs. Culture supernatants were collected, and the cytokine levels were assessed using an immunoassay. The production of IL-1β, IL-6, IL-8, IL-12(p40) and IL-12(p70) was measured. The data shown are the mean value ± SEM for three independent experiments. *, p<0.05 compared with the negative control; #, p<0.05 compared with S. typhi; N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g001

thumbnail
Figure 2. Measurement of anti-inflammatory cytokines and TNF-α in DCs after exposure to B. breve, Salmonella or a combination of the two.

Dendritic cells (DCs) were incubated for 4 h with the B. breve CNCM I-4035 (Prob) probiotic or its cell-free supernatant (CFS), Salmonella (Sal) or both and then incubated for 20 h in medium containing antibiotics. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. Negative-control cultures contained unstimulated DCs. Culture supernatants were collected, and the cytokine levels were assessed by an immunoassay in which the production of IL-10, TNF-α, TGF-β1 and TGF-β2 was measured. The data shown are the mean value ± SEM of three independent experiments. *, p<0.05 compared with controls; #, p<0.05 compared with S. typhi; N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g002

thumbnail
Figure 3. Measurement of chemokines and IFNγ in DCs after exposure to B. breve, Salmonella or a combination of the two.

Dendritic cells (DCs) were incubated for 4 h with the B. breve CNCM I-4035 (Prob) probiotic or its cell-free supernatant (CFS), Salmonella (Sal) or a combination of the two and further incubated for 20 h in medium containing antibiotics. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. Negative control cultures contained unstimulated DCs. Culture supernatants were collected, and the cytokine and chemokine levels were assessed by an immunoassay. The production of IFNγ and the chemokines MCP-1/CCL2, MIP-1α/CCL3, RANTES/CCL5 and MDC/CCL22 was measured. The data shown are the mean values ± SEM of three independent experiments. *, p<0.05 compared with controls; #, p<0.05 compared with S.typhi; N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g003

In DCs, B.breve CNCM I-4035 (live bacteria) and its CFS exerted different behaviors with regard to cytokine induction in response to B. breve CNCM I-4035 stimulation. The CFS decreased the release of pro-inflammatory cytokines (e.g., IL-6 and IL-12p40) and chemokines (e.g., RANTES/CCL5 and MIP-1α/CCL3) in human intestinal DCs challenged with S. typhi (Figures 1 to 3). In contrast, the B. breve CNCM I-4035 strain (live bacteria) was a potent inducer of pro-inflammatory cytokines (e.g., IL-8 and IL-6; Figures 1 and 2), chemokines (e.g., MDC/CCL22; Figure 3) and some anti-inflammatory cytokines (e.g., IL-10; Figure 2). Moreover, DCs interacting with the CFS, in absence of pathogenic bacteria, released low amounts of pro-inflammatory cytokines (e.g., IL-6 and IL-12p40) and chemokines (e.g., MDC and RANTES). In contrast, B.breve (live bacteria) stimulation increased overall cytokine and chemokine production, namely IL-6, IL-8 and MDC (Figure 1 to 3). In addition that treatment also produced high levels of IL-10 (Figure 2).

As shown in Figure 2, the Bifidobacterium strain was a potent TGF-β1 inducer. Interestingly, CFS restored TGF-β levels in the presence of Salmonella. In contrast, live B.breve was unable to increase TGF-β1 production. Finally, we did not detect TGF-β2 and TGF-β3 expression.

The effects of live B.breve CNCM I-4035 or its CFS, S.typhi, or a combination of the two on the production of pro-inflammatory cytokines, anti-inflammatory cytokines and chemokines by intestinal-like human DCs are summarized in tables 1, 2 and 3, respectively. The data shown are the mean value ± SEM of three independent experiments.

thumbnail
Table 1. Effects of live B.breve CNCM I-4035 (Prob) or its cell-free culture supernatant (CFS), S.typhi, or a combination of the two on the secretion of IL-1β, IL-6, IL-8, IL-12p40 and IL-12p70 by intestinal-like human dendritic cells.

https://doi.org/10.1371/journal.pone.0059370.t001

thumbnail
Table 2. Effects of live B.breve CNCM I-4035 (Prob) or its cell-free culture supernatant (CFS), S.typhi, or a combination of the two on the secretion of IL-10, TNF-α and TGF-β1 by intestinal-like human dendritic cells.

https://doi.org/10.1371/journal.pone.0059370.t002

thumbnail
Table 3. Effects of live B.breve CNCM I-4035 (Prob) or its cell-free culture supernatant (CFS), S.typhi, or a combination of the two on the secretion of chemokines MCP-1, MIP-1α, RANTES, MDC and IFN-γ by intestinal-like human dendritic cells.

https://doi.org/10.1371/journal.pone.0059370.t003

Differences between B. breve CNCM I-4035 and its supernatant were observed in the induction of TLR signaling pathway components in human DCs, particularly TLR9 expression

S. typhi induced the expression of other TLR genes including TLR1, TLR2 and TLR5 (Figure 4) and upregulated TLR9 gene expression in human DCs (Figure 5) and. A similar effect was observed for IRAK4, TAK1, JNK (Figures 5 and 6) and IL-10 (Figure 7).

thumbnail
Figure 4. Expression of TLR genes in DCs in the presence of B. breve, Salmonella or a combination of the two.

Comparison of the expression of TLR1, TLR2, TLR3, TLR4 and TLR5 in dendritic cells (DCs) in the presence of the probiotic (Prob), its supernatant (CFS), Salmonella (Sal) or a combination of these stimuli. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. The fold change (Fc) represents the ratio of the expression in treated DCs to that in control cells. *, p<0.05 compared with controls; #, p<0.05 compared with S. typhi. N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g004

thumbnail
Figure 5. Expression of TLR signaling pathway components in DCs treated with B. breve, Salmonella or a combination of the two.

Comparison of the expression of TLR9, MYD88, IRAK-1, IRAK-4 and TOLLIP in dendritic cells (DCs) in the presence of the probiotic (Prob) B. breve, its supernatant (CFS), Salmonella (Sal) or a combination of these stimuli. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. The fold change (Fc) represents the ratio of the expression in treated DCs to that in control cells. *, p<0.05 compared with controls; #, p<0.05 compared with S. typhi. N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g005

thumbnail
Figure 6. Expression of TLR signaling pathway components in DCs treated with B. breve, Salmonella or a combination of the two.

Comparison of the expression of CASP8, TAK-1, JNK, IRF-3 and MAPK14 in dendritic cells (DCs) in the presence of the probiotic (Prob) B. breve, its supernatant (CFS), Salmonella (Sal) or a combination of these stimuli. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. The fold change (Fc) represents the ratio of the expression in treated DCs to that in control cells. *, p<0.05 compared with controls; #, p<0.05 compared with S. typhi. N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g006

thumbnail
Figure 7. Expression of TLR signaling pathway components in DCs treated with B. breve, Salmonella or a combination of the two.

Comparison of the expression of NFKBIA, NFKB-1, TBK-1, IL-10 and TNF-α in dendritic cells (DCs) in the presence of the probiotic (Prob) B. breve, its supernatant (CFS), Salmonella (Sal) or a combination of these stimuli. E. coli lipopolysaccharide (LPS; 20 ng/ml) was used as a positive control. The fold change (Fc) represents the ratio of the expression in the treated DCs to that in the control cells. *, p<0.05 compared with controls; #, p<0.05 compared with S. typhi. N.D. indicates not detected.

https://doi.org/10.1371/journal.pone.0059370.g007

Differences between the probiotic bacteria B. breve CNCM I-4035 and its CFS were observed with regard to TLR expression in DCs (Figures 4 and 5). Both stimuli induced strong TLR9 transcription. In addition, CFS was a more potent inducer of TLR9 expression than live B. breve CNCM I-4035 in the presence of S. typhi (Figure 5). CFS and live B. breve CNCM I-4035 both induced strong and sustained TLR2 transcription (Figure 4). The live bacteria upregulated TLR4, whereas CFS upregulated TLR1 and TLR5 (Figure 4).

Interestingly, in response to stimulation with strain B. breve CNCM I-4035 and Salmonella, TLR1, TLR2 and TLR5 expression was decreased, whereas exposure of the DCs to the probiotic and Salmonella upregulated TLR3 gene expression (Figures 4 and 5). Upon stimulation with CFS plus Salmonella, the expression of the TLR genes increased (Figures 4 and 5). Both treatments induced the expression of TOLLIP (Figure 5), JNK (Figure 6), TBK1 and TNF-α (Figure 7). We also observed differences between the treatments. CFS induced the expression of IRAK4, MYD88 (Figure 5) and CASP8 (Figure 6), whereas these genes were downregulated by B. breve CNCM I-4035.

Discussion

Intestinal DCs, are known to sample microbes that continuously bombard the intestinal mucosa via PRRs such as TLRs and NLRs [25]. However, the underlying mechanisms are poorly understood. One main difficulty is the assessment of the interaction with intestinal immune system components, particularly DCs, which are key players in mucosal immunity [26]. A few studies have addressed the effects of bifidobacteria on human immunocompetent cells [27][29]; however, to the best of our knowledge, this is the first study to analyze the immune response to human intestinal-like DCs developed from CD34+ progenitor cells isolated from the umbilical cord blood.

The main finding of this study was that B. breve CNCM I-4035 and its supernatant could modify the release of cytokines by DCs in specific and differing manners. The CFS exhibited an anti-inflammatory behavior by decreasing pro-inflammatory cytokines (e.g., IL-6 and IL-12p40) and chemokines (e.g., RANTES/CCL5 and MIP-1α/CCL3) in DCs challenged with S. typhi. However, CFS did not increase IL-10 production. This observation is in contrast with that of Hoarau et al., who reported that the B. breve C50 supernatant induces high IL-10 levels in DCs [30]. It is important to note that the B. breve supernatant by itself was a poor cytokine inducer (both inflammatory and non-inflammatory) in our study but had an important impact on the ability of human DCs to secrete lower levels of inflammatory cytokines in response to Salmonella, which suggests that this supernatant may have immunomodulatory properties and may be used to dampen inflammatory responses. These results are consistent with a recent study indicating similar effects for CFS from a novel probiotic strain isolated from the feces of newborns that were exclusively breast-fed (Lactobacillus paracasei CNCM I-4034), i.e., decreased pro-inflammatory cytokines and chemokines in human DCs challenged with S. typhi [23], [31]. Altogether, these data indicate that soluble bacteria product(s) released by B.breve CNCM I-4035 possess anti-inflammatory activity and should be identified. Work in progress in our lab using a proteomic view indicates that this bacterium secretes a number of proteins able to interact with the gut associated immune system (unpublished data).

In contrast to CFS, B. breve CNCM I-4035 (live bacteria) was a potent inducer of the pro-inflammatory cytokines (e.g., IL-8) and chemokines tested (e.g., MDC/CCL22). Our results are in agreement with several studies that reported that some members of the Bifidobacterium genus are inducers of IL-6 [32] and, to a lesser degree, IL-12 [33]. Similarly to most Bifidobacterium strains, live B. breve CNCM I-4035 stimulated the production of high levels of IL-10 [34]. This increase may have an anti-inflammatory effect [35]. The co-incubation of DCs with live B. breve and S. typhi strongly increased the release of pro-inflammatory cytokines, particularly IL-6, IL-8 and TNF-α, as well as IL-10. IL-10 and TNF-α are pleiotropic cytokines that are produced by immune cells. These cytokines are mutually regulated and play opposing roles in inflammatory responses; therefore, their relative balance is of central relevance for controlling immune deviation [36], [37]. We observed that IL-10 production was higher than TNF-α release; therefore, it appears that B. breve CNCM I-4035 (live bacteria) promotes anti-inflammatory effects to restore homeostasis and prevents Salmonella-induced inflammation. In addition, it has been suggested that the high genomic cytosine and guanosine (CG) content of the Bifidobacterium species (approximately 60%) increases IL-10 production [38]. Moreover, in line with other studies, live B. breve CNCM I-4035 did not stimulate IL-12 p70, which is a characteristic effect of bifidobacteria. However, IL-12p40 expression was increased in the presence of S. typhi. This result was predictable because IL-12 induction is strongly correlated with TNF-α production [32].

TGF-β expression analysis demonstrated that the Bifidobacterium strain is a potent TGF-β1 inducer. CFS restored TGF-β levels in the presence of Salmonella. TGF-β1 is a pleiotropic cytokine known to inhibit immune responses at several levels including inhibition of T cell proliferation and differentiation [39], [40] and inhibition of DC maturation [41]. Therefore, the elevation in TGF-β1 production could be responsible for the observed anti-inflammatory effects upon probiotic stimulation.

TLRs are pattern recognition receptors that recognize microbial components and initiate an innate immune response (4). B. breve and its supernatant possess different abilities to regulate the TLR signaling pathway. Our results demonstrate that live B. breve CNCM I-4035 and its CFS stimulated TLR9 expression in the presence and absence of Salmonella. Plantinga et al. reported that cytokine induction by B. breve and lactobacilli is strongly dependent on TLR9 [42]. Their genomic DNA was identified as one of the anti-inflammatory components [43]. Ghadimi et al. reported that TLR9 signaling may at least in part mediate the anti-inflammatory effects of natural-commensal origin DNA [44]. Our results, consistent with several authors, indicate that TLR9 activation is one of the major pathways responsible for the anti-inflammatory effects of probiotics [43][45]. In our study, TLR9 gene expression in response to the bacterial supernatant was significantly higher than that in response to B. breve CNCM I-4035 in DCs challenged with Salmonella. In consequence, this could explain the anti-inflammatory effects of the CFS compared with B.breve (live bacteria). Moreover, it has been proposed that the high frequency of CpG motifs in the DNA of the Bifidobacterium genus may play an important role in the immunostimulatory properties of commensal or probiotic bifidobacterial strains.

The strong upregulation of the TLR2 gene in the presence of B. breve is not surprising because peptidoglycan and lipoteichoic acid, components of the cell wall of Gram-positive bacteria are TLR-2 ligands. A recent study reported that TLR2 recognition had the opposite effect of TLR9 recognition as it induced the expression of TNF-α, IL-1β and IFNγ [42]. This effect may explain the cytokine profile induced by the bacteria in the absence of Salmonella, which is characterized by IL-10 and TNF-α secretion. Moreover, TLR2 has also been implicated in the induction of regulatory T-cell responses, which further emphasizes the immunosuppressive potential of TLR2 signaling. However, the involvement of TLR2 in this process remains unclear, although an immunoregulatory role of TLR2 in the recognition of probiotic strains has been described [30], [46], [47].

In line with several previous studies, our results suggest that probiotic live bacteria increase the expression of TLR2 and TLR9 to activate an innate immune response [48][51] and provide immunostimulation, whereas its CFS increases the expression of TLR9 and TLR5 to exert anti-inflammatory effects. In addition, Uematsu et al. [52] proposes that microbiota induce Ig A production through a mechanism mediated by TLR5. The major role of Ig A is to maintain a balance between the host and microbiota [53], [54].

In contrast, in the presence of Salmonella, the CFS increased the expression of CASP8 and IRAK4, whereas these genes were downregulated by the live bacteria. Both treatments induced TOLLIP gene expression. In this context, signal propagation is necessary to amplify the initiating signal to trigger the nuclear mobilization of transcription factors and induce gene expression. TOLLIP is an adaptor molecule that can bind to TLR2 and TLR4 to inhibit MyD88 binding and activation [55], [56]. In addition, TOLLIP binds to and is phosphorylated by IRAK, which suppresses its ability to function in the TLR pathway [57].

Finally, our results coincide with those of another study indicating that live probiotic bacteria affect the intestinal immune response, whereas secreted components exert anti-inflammatory effects in the gastrointestinal tract [58]. This supernatant may protect immune system from highly infectious agents such as Salmonella typhi and can down-regulate pro-inflammatory pathways.

Author Contributions

Conceived and designed the experiments: AG CGL. Performed the experiments: MBB SMQ. Analyzed the data: MBB. Contributed reagents/materials/analysis tools: EM MJB FR. Wrote the paper: MBB AG.

References

  1. 1. Verbeek R, Bsibsi M, Plomp A, van Neerven RJ, te Biesebeke R, et al. (2010) Late rather than early responses of human dendritic cells highlight selective induction of cytokines, chemokines and growth factors by probiotic bacteria. Benef. Microbes. 1: 109–19.
  2. 2. Guarner F, Malaguelada JR (2003) Gut flora in health and disease. Lancet. 361: 512–519.
  3. 3. Collado MC, Isolauri E, Salminen S, Sanz Y (2009) The impact of probiotic on gut health. Curr. Drug. Metab. 10: 68–78.
  4. 4. Bermudez-Brito M, Plaza-Díaz J, Muñoz-Quezada S, Gomez-Llorente C, Gil A (2012) Mechanisms of action of probiotics. Ann. Nutr. Metab. 61: 160–174.
  5. 5. Yan F, Polk DB (2011) Probiotics and immune health. Curr. Opin. Gastroenterol. 27: 496–501.
  6. 6. Huang JS, Bousvaros A, Lee JW, Diaz A, Davidson EJ (2002) Efficacy of probiotic use in acute diarrhea in children: a meta-analysis. Dig. Dis. Sci. 47: 2625–34.
  7. 7. Pelletier X, Laure-Boussuge S, Donazzolo Y (2001) Hydrogen excretion upon ingestion of dairy products in lactose-intolerant male subjects: importance of the live flora. Eur. J. Clin. Nutr. 55: 509–12.
  8. 8. Woodard GA, Encarnacion B, Downey JR, Peraza J, Chong K, et al. (2009) Probiotics improve outcomes after Roux-en-Y gastric bypass surgery: a prospective randomized trial. J. Gastrointest. Surg. 13: 1198–204.
  9. 9. Karska-Wysocki B, Bazo M, Smoragiewicz W (2010) Antibacterial activity of Lactobacillus acidophilus and Lactobacillus casei against methicillin-resistant Staphylococcus aureus (MRSA). Microbiol. Res. 165: 674–86.
  10. 10. Liong MT (2008) Safety of probiotics: translocation and infection. Nutr. Rev. 66: 192–202.
  11. 11. Rafter J, Bennett M, Caderni G, Clune Y, Hughes R, et al. (2007) Dietary synbiotics reduce cancer risk factors in polypectomized and colon cancer patients. Am. J. Clin. Nutr. 85: 488–96.
  12. 12. Moayyedi P, Ford AC, Talley NJ, Cremonini F, Foxx-Orenstein AE, et al. (2010) The efficacy of probiotics in the treatment of irritable bowel syndrome: a systematic review. Gut. 59: 325–32.
  13. 13. Golowczyc MA, Mobili P, Garrote GL, Abraham AG, De Antoni GL (2007) Protective action of Lactobacillus kefir carrying S-layer protein against Salmonella enterica serovar enteritidis. Int. J. Food. Microbiol. 118: 264–73.
  14. 14. Ohnmacht C, Pullner A, King SB, Drexler I, Meier S, et al. (2009) Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity. J. Exp. Med. 206: 549–59.
  15. 15. Kushwah R, Hu J (2011) Role of dendritic cells in the induction of regulatory T cells. Cell. Biosci. 1: 20.
  16. 16. Rizzello V, Bonaccorsi I, Dongarrà ML, Fink LN, Ferlazzo G (2011) Role of natural killer and dendritic cell crosstalk in immunomodulation by commensal bacteria probiotics. J. Biomed. Biotechnol. 2011: 473097.
  17. 17. Gómez-Llorente C, Muñoz S, Gil A (2010) Role of Toll-like receptors in the development of immunotolerance mediated by probiotics. Proc. Nutr. Soc. 69: 381–9.
  18. 18. Hua MC, Lin TY, Lai MW, Kong MS, Chang HJ, et al. (2010) Probiotic Bio-Three induces Th1 and anti-inflammatory effects in PBMC and dendritic cells. World. J. Gastroenterol. 16: 3529–40.
  19. 19. Kelsall BL, Rescigno M (2004) Mucosal Dendritic Cells in Immunity and Inflammation. Nat. Immunol. 5: 1091–5.
  20. 20. Tsilingiri K, Barbosa T, Penna G, Caprioli F, Sonzogni A, et al. (2012) Probiotic and postbiotic activity in health and disease: comparison on a novel polarised ex-vivo organ culture model. Gut. 61: 1007–15.
  21. 21. Ayehunie S, Snell M, Child M, Klausner M (2009) A plasmacytoid dendritic cell (CD123+/CD11c−) based assay system to predict contact allergenicity of chemicals. Toxicology. 264: 1–9.
  22. 22. Vieites Fernández JM, Muñoz Quezada S, Llamas Company I, Maldonado Lozano J, Romero Braquehais R, et al. (2010) PCT AX090006WO.
  23. 23. Muñoz-Quezada S, Chenoll E, Vieites Fernández JM, Genovés S, Maldonado J, et al. (2013) Isolation, identification and characterization of three novel probiotic strains (Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036) from faeces of exclusively breast milk fed infants. Br. J. Nutr. 109: S51–62.
  24. 24. Muñoz-Quezada S, Bermudez-Brito M, Chenoll E, Genovés S, Gómez-Llorente C, et al. (2013) Competitive inhibition of three novel bacteria isolated from faeces of breast milk-fed infants against selected enteropathogens. Br. J. Nutr. 109: S63–9.
  25. 25. Stagg AJ, Hart AL, Knight SC, Kamm MA (2003) The dendritic cell: its role in intestinal inflammation and relationship with gut bacteria. Gut. 52: 1522–9.
  26. 26. Evrard B, Coudeyras S, Dosgilbert A, Charbonnel N, Alamé J, et al. (2011) Dose-dependent immunomodulation of human dendritic cells by the probiotic Lactobacillus rhamnosus Lcr35. PLoS One 6: e18735.
  27. 27. Boyle RJ, Robins-Browne RM, Tang ML (2006) Probiotic use in clinical practice: what are the risks? Am. J. Clin. Nutr. 83: 1256–1264.
  28. 28. Young SL, Simon MA, Baird MA, Tannock GW, Bibiloni R, et al. (2004) Bifidobacterial species differentially affect expression of cell surface markers and cytokines of dendritic cells harvested from cord blood. Clin. Diagn. Lab. Immunol. 11: 686–690.
  29. 29. López P, Gueimonde M, Margolles A, Suárez A (2010) Distinct Bifidobacterium strains drive different immune responses in vitro. Int. J. Food. Microbiol. 138: 157–165.
  30. 30. Hoarau C, Lagaraine C, Martin L, Velge-Roussel F, Lebranchu Y (2006) Supernatant of Bifidobacterium breve induces dendritic cell maturation, activation, and survival through a Toll-like receptor 2 pathway. J. Allergy. Clin. Immunol. 117: 696–702.
  31. 31. Bermudez-Brito M, Muñoz-Quezada S, Gomez-Llorente C, Matencio E, Bernal MJ, et al. (2012) Human intestinal dendritic cells decrease cytokine release against Salmonella infection in the presence of Lactobacillus paracasei upon TLR activation. PloS One 7: e43197.
  32. 32. Weiss G, Christensen HR, Zeuthen LH, Vogensen FK, Jacobsen M, et al. (2011) Lactobacilli and bifidobacteria induce differential interferon-β profiles in dendritic cells. Cytokine. 56: 520–30.
  33. 33. Turroni F, Van Sinderen D, Ventura M (2011) Genomics and ecological overview of the genus Bifidobacterium. Int. J. Food. Microbiol. 149; 37–44.
  34. 34. Borchers AT, Selmi C, Meyers FJ, Keen CL, Gershwin ME (2009) Probiotics and immunity. J. Gastroenterol. 44: 26–46.
  35. 35. Madsen K (2006) Probiotics and the immune response. J. Clin. Gastroenterol. 40: 232–4.
  36. 36. Aujla SJ, Dubin PJ, Kolls JK (2007) Th17 cells and mucosal host defense. Semin Immunol. 19: 377–82.
  37. 37. Dubin PJ, Kolls JK (2008) Th17 cytokines and mucosal immunity. Immunol Rev. 226: 160–71.
  38. 38. Medina M, Izquierdo E, Ennahar S, Sanz Y (2007) Differential immunomodulatory properties of Bifidobacterium longum strains: relevance to probiotic selection and clinical applications. Clin. Exp. Immunol. 150: 531–8.
  39. 39. Marie JC, Letterio JJ, Gavin M, Rudensky AY (2005) TGF-beta 1 maintains suppressor function and Foxp3 expression in CD4+ CD25+ regulatory T cells. J Exp Med. 201: 1061–7.
  40. 40. Nakamura K, Kitani A, Fuss I, Pedersen A, Harada N, et al. (2004) TGF-beta 1 plays an important role in the mechanism of CD4+ CD25+ regulatory T cell activity in both humans and mice. J Immunol. 172: 834–4240.
  41. 41. Strobl H, Knapp W (1999) TGF-beta 1 regulation of dendritic cells. Microbes Infect 1: 1283–90.
  42. 42. Plantinga TS, van Maren WW, van Bergenhenegouwen J, Hameetman M, Nierkens S, et al. (2011) Differential Toll-like receptor recognition and induction of cytokine profile by Bifidobacterium breve and Lactobacillus strains of probiotics. Clin. Vaccine. Immunol. 18: 621–8.
  43. 43. Hiramatsu Y, Satho T, Irie K, Shiimura S, Okuno T, et al. (2012) Differences in TLR9-dependent inhibitory effects of H(2)O(2)-induced IL-8 secretion and NF-kappa B/I kappa B-alpha system activation by genomic DNA from five Lactobacillus species. Microbes Infect. 2012 pii S1286–4579(12)00274–2.
  44. 44. Ghadimi D, Vrese Md, Heller KJ, Schrezenmeir J (2010) Effect of natural commensal-origin DNA on toll-like receptor 9 (TLR9) signaling cascade, chemokine IL-8 expression, and barrier integritiy of polarized intestinal epithelial cells. Inflamm Bowel Dis. 16: 410–27.
  45. 45. Lavelle EC, Murphy C, O'Neill LA, Creagh M (2010) The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. Mucosal. Immunol. 3: 17–28.
  46. 46. Kaji R, Kiyoshima-Shibata J, Nagaoka M, Nanno M, Shida K (2010) Bacterial teichoic acids reverse predominant IL-12 production induced by certain Lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J Immunol. 184: 3505–13.
  47. 47. Zeuthen LH, Fink LN, Frøkiaer H (2008) Toll-like receptor 2 and nucleotide-binding oligomerization domain-2 play divergent roles in the recognition of gut-derived lactobacilli and bifidobacteria in dendritic cells. Immunology. 124: 489–502.
  48. 48. Vizoso Pinto MG, Rodriguez Gomez M, Seifert S, Watzl B, Holzapfel WH, et al. (2009) Lactobacilli stimulate the innate immune response and modulate the TLR expression of HT29 intestinal epithelial cells in vitro. Int. J. Food. Microbiol. 33: 86–93.
  49. 49. Tao Y, Drabik KA, Waypa TS, Musch MW, Alverdy JC, et al. (2006) Soluble factors from Lactobacillus GG activate MAPKs and induce cytoprotective heat shock proteins in intestinal epithelial cells. Am. J. Physiol. 290: 1018–30.
  50. 50. Kim YG, Ohta T, Takahashi T, Kushiro A, Nomoto K, et al. (2006) Probiotic Lactobacillus casei activates innate immunity via NF-kappaB and p38 MAP kinase signaling pathways. Microbes. Infect. 8: 994–1005.
  51. 51. Voltan S, Castagliuolo I, Elli E, Longo S, Brun P, et al. (2007) Aggregating phenotype in Lactobacillus crispatus determines intestinal colonization and TLR2 and TLR4 modulation in murine colonic mucosa. Clin. Vaccine. Immunol. 14: 1138–48.
  52. 52. Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, et al. (2008) Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat. Immunol. 9: 769–76.
  53. 53. Feng T, Elson CO, Cong Y (2011) Treg cell-Ig A axis in maintenance of host immune homeostasis with microbiota. Int. Immunopharmacol. 11: 589–592.
  54. 54. Hansen J, Gulati A, Sartor RB (2010) The role of mucosal immunity and host genetics in defining intestinal commensal bacteria. Curr. Opin. Gastroenterol. 26: 564–71.
  55. 55. Bulut Y, Faure E, Thomas L, Equils O, Arditi M (2001) Cooperation of Toll-like receptor 2 and 6 for cellular activation by soluble tuberculosis factor and Borrelia burgdorferi outer surface protein A lipoprotein: role of Toll-interacting protein and IL-1 receptor signaling molecules in Toll-like receptor 2 signaling. J. Immunol. 167: 987–94.
  56. 56. Zhang G, Ghosh S (2002) Negative regulation of toll-like receptor-mediated signalling by Tollip. J. Biol. Chem. 277: 7059–65.
  57. 57. Burns K, Clatworthy J, Martin L, Martinon F, Plumpton C, et al. (2000) Tollip, a new component of the IL-1RI pathway, links IRAK to the IL-1 receptor. Nat Cell Biol. 2: 346–51.
  58. 58. Adams CA (2010) The probiotic paradox: live and dead cells are biological response modifiers. Nutr. Res. Rev. 23: 37–46.