throbber
Hindawi Publishing Corporation
`BioMed Research International
`Volume 2015, Article ID 479140, 6 pages
`http://dx.doi.org/10.1155/2015/479140
`
`Research Article
`The Effects of Bifidobacterium breve on Immune Mediators and
`Proteome of HT29 Cells Monolayers
`
`Borja Sánchez,1 Irene González-Rodríguez,1 Silvia Arboleya,1 Patricia López,2 Ana Suárez,2
`Patricia Ruas-Madiedo,1 Abelardo Margolles,1 and Miguel Gueimonde1
`1 Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos L´acteos de Asturias,
`Consejo Superior de Investigaciones Cient´ıficas (IPLA-CSIC), Paseo R´ıo Linares s/n, Villaviciosa, 33300 Asturias, Spain
`2 Department of Functional Biology, Immunology Area, University of Oviedo, Oviedo, 33006 Asturias, Spain
`
`Correspondence should be addressed to Miguel Gueimonde; mgueimonde@ipla.csic.es
`
`Received 15 May 2014; Revised 3 October 2014; Accepted 4 October 2014
`
`Academic Editor: Riitta Korpela
`
`Copyright © 2015 Borja S´anchez et al. This is an open access article distributed under the Creative Commons Attribution License,
`which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`The use of beneficial microorganisms, the so-called probiotics, to improve human health is gaining popularity. However, not all of
`the probiotic strains trigger the same responses and they differ in their interaction with the host. In spite of the limited knowledge
`on mechanisms of action some of the probiotic effects seem to be exerted through maintenance of the gastrointestinal barrier
`function and modulation of the immune system. In the present work, we have addressed in vitro the response of the intestinal
`epithelial cell line HT29 to the strain Bifidobacterium breve IPLA20004. In the array of 84 genes involved in inflammation tested,
`the expression of 12 was modified by the bifidobacteria. The genes of chemokine CXCL6, the chemokine receptor CCR7, and,
`specially, the complement component C3 were upregulated. Indeed, HT29 cells cocultivated with B. breve produced significantly
`higher levels of protein C3a. The proteome of HT29 cells showed increased levels of cytokeratin-8 in the presence of B. breve.
`Altogether, it seems that B. breve IPLA20004 could favor the recruitment of innate immune cells to the mucosa reinforcing, as well
`as the physical barrier of the intestinal epithelium.
`
`1. Introduction
`
`Probiotics are live microorganisms which when administered
`in adequate amounts confer a health benefit on the host [1],
`the genus Bifidobacterium being among the most widely used.
`These microorganisms are common members of the human
`gut microbiota and they predominate in breast-fed infants
`[2]. Several beneficial health effects have been attributed
`to specific probiotic strains [3]. Although the knowledge
`on probiotic mechanisms of action is still limited some of
`these beneficial effects are exerted through their role in the
`maintenance of the gastrointestinal barrier function and by
`modulating the immune system [4, 5].
`The interest in the immunomodulatory properties of
`probiotic bacteria derives from the observations that intesti-
`nal microbiota plays a critical role in the development and
`regulation of the immune system [6]. It is known that
`different probiotic bacteria present different effects upon the
`
`immune system [7, 8], making necessary the characterization
`of the effects of each specific potentially probiotic strain.
`Some strains promote Th1 responses, characterized by the
`production of IFN𝛾 and TNF𝛼, whereas other strains induce
`anti-inflammatory cytokines generating a Th2 profile [7, 8].
`To determine these properties, the direct effect of the inter-
`action of probiotic bifidobacteria with immune cells, either
`total peripheral blood mononuclear cells (PBMCs) or isolated
`immune cell types, is often studied. However, the potential
`effect of the cross talk between bifidobacteria and epithelial
`cells upon the immune system has received less attention.
`The intestinal epithelium separates microorganisms from the
`underlying immune cells. It consists of a layer of cells, mainly
`enterocytes, and a mucus layer that coats the epithelium [9].
`Moreover, different immune cells are localized in the gut
`associated lymphoid tissue, which constitute the first contact
`point between gut commensals and the immune system
`[10]. Consequently, assessing the effect of the interaction
`
`Genome Ex. 1044
`Page 1 of 6
`
`

`

`2
`
`BioMed Research International
`
`of potentially probiotic Bifidobacterium strains with the gut
`mucosa constitutes an important task for both probiotics
`selection and understanding of their mechanisms of action.
`This understanding would allow selection of specific strains
`with the desired properties for a specific application.
`Previous studies carried out on the breast-milk isolate
`Bifidobacterium breve IPLA20004 [11] by our group indi-
`cated the ability of this strain to induce Th1 polarization of
`lymphocytes and to increase the physical resistance of the
`intestinal mucosa [12, 13]. These results suggest that this strain
`may be of interest for increasing the intestinal barrier against
`pathogens, firstly by strengthening the physical resistance of
`the epithelial layer and secondly by modulating the immune
`system towards a preactivated steady state. Moreover, some
`effects of the strain on the expression of chemokines and
`their receptors have been previously suggested [13]. To this
`regard an effect on the production of chemokines by intestinal
`epithelial cells may have a direct impact on the immune
`system by affecting the recruitment of immune cells to the
`mucosa.
`For the above-mentioned reasons we decided to evaluate
`the effect of B. breve IPLA20004 on the expression of genes
`related to the inflammatory response and on the production
`of cytokines, by the human intestinal epithelial cell line HT29.
`Moreover, the effect of the strain on HT29 cells was also
`assessed by proteomic analyses.
`
`2. Materials and Methods
`2.1. Bacteria Culture Conditions. To evaluate the effects of
`the B. breve IPLA20004 on HT29 cells, cultures were freshly
`prepared by growing the microorganisms in MRS medium
`(Difco, Becton, Dickinson and Company, Le Pont de Claix,
`France) supplemented with a 0.25% L-cysteine (Sigma Chem-
`ical Co., St. Louis, MO, USA) (MRSc) at 37∘C under anaerobic
`conditions (10% H2, 10% CO2, and 80% N2) in a chamber Mac
`500 (Don Whitley Scientific, West Yorkshire, UK).
`
`2.2. HT29 Cell Line Culture Conditions. The epithelial intesti-
`nal cell
`line HT29 (ECACC number 91072201), derived
`from human colon adenocarcinoma, was purchased from the
`European Collection of Cell Cultures (Salisbury, UK). HT29
`cell culture passages 146-147 were used for the experiments.
`The cell line was maintained in McCoy’s medium supple-
`mented with 3 mM L-glutamine, 10% (v/v) heat-inactivated
`bovine fetal serum, and a mixture of antibiotics to give a
`final concentration of 50 𝜇g/mL penicillin, 50 𝜇g/mL strepto-
`mycin, 50 𝜇g/mL gentamicin, and 1.25 𝜇g/mL amphotericin
`B. All media and supplements were obtained from Sigma.
`The incubations took place at 37∘C, 5% CO2 in an SL
`water-jacketed CO2 incubator (Sheldon Mfg. Inc., Cornelius,
`Oregon, USA). Culture media were changed every two days
`and the cell line was trypsinized with 0.25% trypsin-EDTA
`solution (Sigma) following standard procedures. For gene
`expression experiments and protein profile determinations,
`105 cells/mL were seeded in 24-well plates and incubated
`to reach a confluent and differential state (reaching about
`107 HT29 cells/mL) after 13 ± 1 days.
`
`2.3. Gene Expression Analysis. B. breve IPLA20004 was
`grown overnight in MRSc, harvested by centrifugation,
`washed twice with Dulbecco’s PBS buffer (Sigma), and
`resuspended in McCoy’s medium without antibiotics. Five
`hundred 𝜇L of a bacterial suspension containing 108 cfu/mL
`(as determined by plate counting) in McCoy’s medium or
`McCoy’s medium without bacteria (control) was added to
`each well containing HT29 monolayers (bacteria/HT29 cell
`ratio 10 : 1) previously washed twice with Dulbecco’s PBS to
`remove the antibiotics. Plates were then incubated for 6 h at
`37∘C, 5% CO2 in a Heracell 240 incubator (Thermo Electron
`LDD GmbH, Langenselbold, Germany). After incubation
`the culture media were removed and stored at −80∘C, the
`monolayers were resuspended in 500 𝜇L of RNA Protect Cell
`Reagent (Qiagen GmbH, Hilden, Germany), and the cells
`were kept frozen at −80∘C until RNA extraction. At least three
`independent experiments were carried out.
`RNA from HT29 cells was extracted by using the
`RNeasy Plus Mini Kit (Qiagen) and QIAshredder homoge-
`nizer columns (Qiagen) following manufacturer instructions.
`Quality of RNA was monitored by gel electrophoresis and it
`was quantified by using an Epoch apparatus (BioTek Instru-
`ments, Inc., Winooski, VT, USA). For reverse-transcriptase
`PCR analyses 1 𝜇g of RNA was reverse-transcribed to cDNA
`by using the RT2 First Strand Kit (SABiosciences, Qiagen,
`Frederick, MD, USA), and gene expression was quantified
`by using the 96-well RT2 Profiler PCR Array for human
`inflammatory cytokines and receptors (SABiosciences) fol-
`lowing manufacturer’s instructions. The array comprises 84
`key genes involved in the inflammatory response includ-
`ing chemokine and cytokine genes (CCL1 [I-309], CCL11
`[eotaxin], CCL13 [mcp-4], CCL15 [MIP-1d], CCL16 [HCC-
`4], CCL17 [TARC], CCL18 [PARC], CCL19, CCL2 [mcp-1],
`CCL20 [MIP-3a], CCL21 [MIP-2], CCL23 [MPIF-1], CCL24
`[MPIF-2/eotaxin-2], CCL25 [TECK], CCL26, CCL3 [MIP-
`1a], CCL4 [MIP-1b], CCL5 [RANTES], CCL7 [mcp-3], CCL8
`[mcp-2], CXCL1, CXCL10 [IP-10], CXCL11 [I-TAC/IP-9],
`CXCL12 [SDF1], CXCL13, CXCL14, CXCL2, CXCL3, CXCL5
`[ENA-78/LIX], CXCL6 [GCP-2], CXCL9, IL13, IL8, IFNA2,
`IL10, IL13, IL17C, IL1A, IL1B, IL1F10, IL1F5, IL1F6, IL1F7,
`IL1F8, IL1F9, IL22, IL5, IL8, IL9, LTA, LTB, MIF, SCYE1, SPP1,
`and TNF), chemokine and cytokine receptor genes (CCR1,
`CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9,
`CX3CR1, XCR1 [CCXCR1], IL1R1, IL1RN, IL5RA, IL8RA,
`IL8RB, IL9R, IL10RA, IL10RB, and IL13RA1), other genes
`involved in the inflammatory response (ABCF1, BCL6, C3,
`C4A, C5, CEBPB, CRP, ICEBERG, LTB4R, and TOLLIP), and
`five housekeeping genes (B2 M, HPRT1, RPL13A, GAPDH,
`and ACTB) for normalization of data.
`
`2.4. Cytokines and C3a Determination. Cytokine and C3a
`levels in the cell culture supernatants of HT29 cells cultured
`with or without B. breve as indicated above were quantified
`by using the High Sensitivity ELISA Kits for human IL10,
`IL12p70, IL1𝛽, and TNF𝛼 and the Platinum ELISA Kits for
`human IL8 and C3a (eBioscience Inc., San Diego, CA, USA).
`Colour development after ELISA was measured in a Modulus
`Microplate Photometer (Turner Biosystems, Sunnyvale, CA,
`USA). All the results were expressed as pg/mL. Detection
`
`Genome Ex. 1044
`Page 2 of 6
`
`

`

`BioMed Research International
`
`3
`
`limits for the ELISA kits used were 0.05, 0.1, 0.05, 0.13, 2,
`and 70 pg/mL for IL10, IL12p70, IL1𝛽, TNF𝛼, IL8, and C3a,
`respectively.
`
`the Proteomic Profiles. B. breve
`2.5. Determination of
`IPLA20004 was grown and added to the wells containing
`HT29 as previously indicated. Plates were then incubated
`for 3 h at 37∘C, 5% CO2, gently washed three times with
`Dulbecco’s PBS buffer to remove the nonadhered bacteria,
`and the HT29 monolayers were kept for further proteomic
`analysis.
`For protein extraction and two-dimensional electropho-
`resis analysis, HT29 monolayers were disaggregated with
`440 𝜇L of lysis buffer (30 mM Tris, 7 M urea, 2 M thiourea, 4%
`(w/v) CHAPS, and 100 mM DTT; all reagents were purchased
`by GE Healthcare Life Sciences) containing complete pro-
`tease inhibitors (Roche Diagnostics, Mannheim, Germany).
`Total protein from the cell suspensions was obtained by
`sonication for one min in ice-chilled water (two cycles),
`with one min of delay between the two cycles. After adding
`2 mg of RNase A (Sigma-Aldrich) and 100 U of DNase I
`(Sigma-Aldrich), the cell lysates were incubated for 30 min at
`RT. Finally, the pellet was centrifuged for 10 min at 16,000 g
`and 4∘C to precipitate insoluble components and cell debris.
`Protein concentration was estimated using the BCA Protein
`Assay Kit (Pierce, Rockford, IL).
`Isoelectric focusing (IEF) was performed in immobilized
`pH gradient (IPG) strips containing a nonlinear pH range of
`3–10 (GE Healthcare Life Sciences), using 500 𝜇g of protein.
`When needed, lysis buffer was added up to 450 𝜇L. In all the
`cases, the IPG-buffer corresponding to pHs 3–10 was added to
`a final concentration of 0.5% (v/v). IEF was conducted at 20∘C
`for 60,000 Vhrs in an IPGphor system (GE Healthcare Life
`Sciences). Proteins were resolved by SDS-PAGE (12.5% w/v
`polyacrylamide gel) and stained with GelCode Blue Safe Pro-
`tein Stain (Pierce). Gels were scanned using ImageScanner
`(GE Healthcare Life Sciences), and spot detection and volume
`quantification were performed with ImageMaster Platinum
`software (version 5.00, GE Healthcare). The relative volume
`of each spot was obtained by determining the spot intensity
`in pixel units and normalizing that value to the sum of the
`intensities of all the spots of the gel. Each experiment was
`performed independently four times, and the differences in
`normalized volumes were analyzed statistically using paired
`Student’s 𝑡-tests (control condition versus presence of the
`bifidobacteria strain).
`
`2.6. Statistical Analyses. Differences in the measured vari-
`ables, between the control HT29 cells and those exposed to
`the B. breve strain, were evaluated by one-way ANOVA test.
`Results were represented by mean ± standard deviation. The
`SPSS 18.0 statistical software package (SPSS Inc., Chicago, IL,
`USA) was used for all determinations and a value of 𝑃 < 0.05
`was considered significant.
`
`3. Results
`3.1. Effect of B. breve IPLA20004 on the Expression of Genes
`Mediating the Inflammatory Response in HT29 Cells. When
`
`Table 1: Changes in cytokines and receptors gene expression in
`HT29 cells after exposition to bifidobacteria when compared to
`exposition to culture medium without bifidobacteria (control), as
`determined by RT-PCR.
`
`Up- or downregulation (compared to control)
`B. breve IPLA20004
`Fold regulation
`17.71
`−3.29
`−5.32
`−2.60
`−6.07
`−10.36
`3.19
`2.12
`−2.43
`−1.64
`−2.75
`−3.65
`
`𝑃
`0.001
`0.026
`0.039
`0.012
`0.006
`0.004
`0.011
`0.028
`0.015
`0.021
`0.009
`0.001
`
`Gene
`
`C3
`CCL2
`CCL25
`CCR1
`CCR4
`CCR5
`CCR7
`CXCL6
`CXCL14
`IL10
`IL13
`XCR1
`
`using the human inflammatory cytokines and receptors
`pathway focused RT-PCR array, comprising 84 key genes
`involved in the inflammatory response, we observed some
`statistically significant changes in gene expression in HT29
`cells after coincubation with B. breve IPLA20004. These
`changes were in general modest and most of the studied genes
`were expressed at low basal levels (Ct values around 30, data
`not shown), with the exception of CCL25 (Ct value of 19 in the
`control HT29 cells) and CCR1 (Ct value 26 in the control).
`The genes whose expression was significantly modified by
`the strain are shown in Table 1. The expression of chemokine
`genes CCL2, CCL25, and CXCL14 and the cytokines genes
`IL10 and IL13 genes was significantly downregulated. On
`the contrary the gene for CXCL6 chemokine was found to
`be upregulated. With regard to chemokine receptor genes,
`a statistically significant downregulation of CCR1, CCR4,
`CCR5, and XCR1 and induction of CCR7 were observed.
`Interestingly, B. breve IPLA20004 upregulated very signifi-
`cantly (17-fold) the expression of the complement component
`C3 (Table 1). No statistically significant differences were
`observed for any of the other genes analyzed in the RT-PCR
`array (data not shown).
`
`3.2. Effect of B. breve IPLA20004 on Cytokines and C3 Pro-
`duction by HT29 Cells. The levels of the different cytokines
`measured, as well as those of C3a, in supernatants of HT29
`cells are shown in Table 2. In general the levels detected
`were low, in some cases being barely over the detection
`limits of the ELISA kits used. No statistically significant
`differences between control and B. breve-exposed HT29 cells
`were observed for IL10, IL1𝛽, TNF𝛼, or IL8 levels. On the
`contrary coculture of HT29 cells with B. breve IPLA20004
`significantly increased the production of IL12p70 and C3a,
`although for the former cytokine the detected levels (0.19 and
`0.3 pg/mL for control and B. breve-exposed HT29 cells, resp.)
`
`Genome Ex. 1044
`Page 3 of 6
`
`

`

`4
`
`BioMed Research International
`
`Table 2: Effect of B. breve IPLA20004 on cytokines and C3a
`levels in HT29 cells supernatants. All the results are expressed
`as pg/mL. Control cells were exposed to culture medium without
`bifidobacteria.
`
`Control
`0.43 ± 0.44
`0.19 ± 0.03
`0.17 ± 0.17
`2.19 ± 1.45
`264.82 ± 38.41
`217.33 ± 38.37
`
`Concentration (pg/mL)
`B. breve IPLA20004
`0.83 ± 0.38
`0.30 ± 0.02
`0.26 ± 0.20
`3.35 ± 0.18
`423.51 ± 200.29
`311 ± 42.14
`
`IL10
`IL12p70
`IL1𝛽
`TNF𝛼
`IL8
`C3a
`
`𝑃
`0.302
`0.007
`0.604
`0.242
`0.249
`0.045
`
`were only slightly above the detection limit of the technique
`used and, therefore, the relevance of this observation is
`unclear.
`
`3.3. Effect of B. breve IPLA20004 on the Proteome of
`HT29 Cells. The comparison of the proteomes of HT29
`cells cocultured with or without B. breve revealed that
`two proteins were significantly (𝑃 < 0.05) upregulated
`in the HT29 cells by the strain B. breve IPLA20004 (see
`Figure 1 in the Supplementary Material available online at
`http://dx.doi.org/10.1155/2014/479140). These proteins were
`excised from the gels and identified as cytokeratin-8 (2.8
`times fold induction) and the chain A of the tapasin-ERp57
`(4.7 times fold induction).
`
`4. Discussion
`The interaction of bacteria with intestinal epithelial cells
`may play a role in immune modulation by modifying gene
`expression and local
`immune environment through, for
`instance, production of chemokines and other immune active
`molecules. Chemokines are chemotactic cytokines that guide
`the migration of cells regulating leukocyte traffic and exert
`their effects by interacting with their specific receptors that
`are selectively found on the surfaces of their target cells.
`We studied the interaction of B. breve IPLA20004 with
`colonic epithelial cells HT29 and found changes in the
`expression of genes related to the inflammatory response
`and immune cell chemotaxis in the HT29 cell line. The
`strain was observed to significantly induce the expression
`of some immunoactive molecules, such as C3 and CXCL6,
`and to downregulate the expression of others including
`CCL2, CCL25, CXCL14, IL10, or IL13. It should be noted,
`however, that in some cases such as IL10 the magnitude of
`the change in gene expression, although significant, was small
`(less than 2) and perhaps of limited biological relevance.
`The effect of probiotics, mainly Lactobacillus strains, on
`transcriptional responses of human epithelial cells has been
`previously assessed both in vitro [14–16] and in vivo [17–
`19]. Although the studies on bifidobacteria are scarcer there
`are also some examples [12, 13, 20, 21]. These studies show
`a limited response of human intestinal epithelia cells lines
`to stimulation with bifidobacteria. Nevertheless, it is still
`
`interesting to see that our results, although in vitro, suggest
`an effect of the strain B. breve IPLA20004 in a number of
`genes coding for cytokines, chemokines, and receptors, which
`is in agreement with some in vivo studies on the effect of
`probiotic lactobacilli upon gene expression patterns in the
`human small bowel [17] and supports a link between the
`interaction of bacteria with epithelial cells and the immune
`system. Interestingly, in spite of the different models used,
`some of the genes found to be modulated in this study
`have been previously reported to be modulated by probiotic
`Lactobacillus strains both in vitro using epithelial cells [22]
`and in vivo in the human small bowel mucosa [17, 19]. To
`this regard, the colonic epithelial cell line used in our study
`(HT29) may better resemble the small bowel, where the
`mucus layer is thin, than the colon where a thick mucus
`layer is known to be present which prevents the close contact
`of bacteria with the epithelial cell [9]. Moreover, coculture
`of mice primary colonic epithelial cells with L. rhamnosus
`GG induced the expression of IL1𝛽, TNF𝛼, CXCL5 (ENA-
`78), CXCL10 (IP10), CCL20 (MIP3𝛼), CCL2 (MCP1), CCL7
`(MCP3), CXCL2 (MIP2𝛼), and CCL5 (RANTES) [22], and
`our results indicated a significant downregulation of CCL2
`without affecting the other L. rhamnosus GG-induced genes.
`This may suggest a differential response to our bifidobacteria
`with regard to L. rhamnosus GG, although the influence of
`the different colonocyte models used cannot be overruled.
`Administration of L. rhamnosus GG to human volunteers
`induced the expression of some of these genes (CCL24,
`CCL2, CXCL3, CXCL13, CXCL12, CCR3, CCL19, CCL21, or
`lymphotoxin-𝛽 [LTB], among others) on the small bowel
`mucosa, whilst other Lactobacillus strains (L. acidophilus
`Lafti L10) resulted in a different expression profile (inducing
`CXCL10 and CXCL11, among others) [19]. On the contrary,
`generalizing, in our in vitro model B. breve IPLA20004
`tended either to downregulate or not to affect these genes
`which suggest a limited stimulatory activity of this strain
`when compared with the immune-stimulatory ability of
`lactobacilli. It should be noted, however, that the differences
`existing between the in vivo studies and our in vitro results
`with HT29 cells may be partly related to the different
`experimental conditions used; for instance, we performed
`the incubations under a 5% CO2 atmosphere in comparison
`with the anaerobic intestinal environment which may have an
`effect on an anaerobic microorganism such as B. breve.
`As indicated above, chemokines function mainly as
`chemoattractants for leukocytes, recruiting monocytes, neu-
`trophils, and other effectors cells from the blood to sites of
`infection or tissue damage [23]. Chemokines such as CCL2
`or CCL25 attract immune cells, such as macrophages and
`T-lymphocytes expressing their receptors (CCR2 and CCR9,
`resp.) to the tissue [23]. Actually, expression of CCR9 has
`been found to be involved in the homing to the intestine of
`thymic T-cells [24]. On the other hand, the only chemokine
`gene found to be upregulated in our study was that of
`CXCL6 (human granulocyte chemotactic protein-2, GCP-
`2). This chemokine attracts and activates neutrophils [25]
`being, together with IL8, the only CXC-family chemokine
`recognized by both CXCR1 and CXCR2 receptors. IL8 is the
`most active protein chemoattracting neutrophil, although in
`
`Genome Ex. 1044
`Page 4 of 6
`
`

`

`BioMed Research International
`
`5
`
`our study its gene was not found to be significantly upreg-
`ulated and IL8 determination in the supernatants showed
`higher, but not statistically significant, values. Moreover, a
`recent study demonstrated an increased production of the
`chemokine CXCL16 (not studied in this work) in germ-
`free animals, which resulted in an increased recruitment of
`immune cells to the intestinal mucosa [26]. This underlines
`the importance of chemokines in immune cells recruitment
`and the modulation of their production by the intestinal
`microbiota.
`Interestingly, the complement component C3 was among
`the most strongly upregulated genes in the small bowel
`mucosa after administration of L. rhamnosus GG to healthy
`volunteers [17]. Similarly, the expression of this gene in HT29
`cells was the most clearly upregulated by our B. breve strain
`and a significantly higher production of C3 by the epithelial
`cell line was confirmed by means of ELISA tests. C3 is the
`most abundant complement protein in serum, it enhances
`phagocytosis promoting innate immunity, and it is also
`important for an effective antibody response, thus constitut-
`ing a link between the complement system and the acquired
`immune response [27]. In our study the downregulation of
`the expression of genes such as CCL2 or CCL25 together
`with the upregulation of the expression of CXCL6 and C3
`by colonic cells suggests a local effect by suppressing the
`recruitment to the mucosa of lymphocytes and by increasing
`that of the innate immunity cells such as neutrophils and
`mastocytes. However, the limitations of our study design do
`not allow the establishment of firm conclusions on whether
`the differences obtained with regard to the reports by other
`authors are due to the different strains used or to the models’
`responsiveness.
`Finally, in order to complement the data on the inter-
`action between B. breve IPLA20004 and HT29 cells we
`performed a proteomic approach. This analysis allowed us
`to detect the overproduction of cytokeratin-8 (CK-8) or type
`I cytoskeletal 8, a keratin protein encoded by the krt8 gene;
`this protein is located in the nucleoplasm and the cytoplasm
`where, as a part of the cytoskeleton, it is known to help to link
`the contractile machinery to dystrophin at the costamere in
`striated muscle cells [28]. Interestingly, this strain has been
`previously found to increase the transepithelial resistance
`of the HT29 cell monolayer [13] which may be correlated
`with this induction of changes in the cytoskeleton. Moreover,
`the chain A of the tapasin-ERp57 was also overproduced.
`The heterodimer formed by tapasin-ERp57,
`linked by a
`stable disulfide bond, is part of the major histocompatibility
`complex (MHC) class I peptide-loading complex [29]. This
`heterodimer has been shown as the functional unit for load-
`ing MHC class I molecules with high-affinity peptides [30].
`It has been shown that upregulation of tapasin may facilitate
`optimal peptide loading on the MHC class I molecule [31],
`although the putative functions in enterocytes have passed
`unnoticed until now.
`In this study we have determined the effects of B.
`breve IPLA20004 on intestinal epithelial cells, observing a
`potential improvement of the epithelial barrier. This, together
`with previous studies carried out on the interaction of the
`strain with immune cells indicating a Th1 profile [8, 12] or
`
`showing an increase of the transepithelial resistance of the
`colonic epithelial cells monolayer [13], suggests the interest
`in conducting experiments in which both polarized epithelial
`cells and immune cells are cocultured.
`In summary, our results suggest that this strain offers
`possibilities for increasing the intestinal barrier against
`pathogens in populations in which the barrier may be
`compromised. This could be achieved by two independent
`mechanisms: firstly by strengthening the cell cytoskeleton
`and, therefore, the physical resistance of the epithelial layer
`and secondly by modulating the immune environment at
`local mucosal level towards a “prestimulated” innate immune
`response by recruiting immune cells.
`
`Conflict of Interests
`All the authors have declared no conflict of interests.
`
`Acknowledgments
`This work was financed by Projects PIE201370E019 from
`CSIC and AGL2009-09445 and AGL2013-43770R from the
`Spanish “Ministerio de Economia y Competitividad” Silvia
`Arboleya was funded by a predoctoral JAE Fellowship from
`CSIC. Irene Gonz´alez-Rodr´ıguez was the recipient of a FPI
`grant and Borja S´anchez of a Juan de la Cierva postdoctoral
`contract, both from the Spanish “Ministerio de Ciencia e
`Innovaci´on.”
`
`References
`[1] Food and Health Agricultural Organization of the United
`Nations and World Health Organization, “Guidelines for the
`evaluation of probiotics in food,” Working Group Report,
`FAO/WHO, Ontario, Canada, 2002.
`[2] T. Yatsunenko, F. E. Rey, M. J. Manary et al., “Human gut
`microbiome viewed across age and geography,” Nature, vol. 486,
`no. 7402, pp. 222–227, 2012.
`[3] M. C. Collado, M. Gueimonde, and G. P´erez-Mart´ınez, “Cur-
`rent and future applications of probiotics,” Current Nutrition
`and Food Science, vol. 7, no. 3, pp. 170–180, 2011.
`[4] M. G. Gareau, P. M. Sherman, and W. A. Walker, “Probiotics
`and the gut microbiota in intestinal health and disease,” Nature
`reviews. Gastroenterology & hepatology, vol. 7, no. 9, pp. 503–514,
`2010.
`[5] N. Binns, Probiotics, Prebiotics and the Gut Microbiota, ILSI
`Europe Concise Monograph Series, International Life Sciences
`Institute, 2013.
`[6] C. L. Maynard, C. O. Elson, R. D. Hatton, and C. T.
`Weaver, “Reciprocal interactions of the intestinal microbiota
`and immune system,” Nature, vol. 489, no. 7415, pp. 231–241,
`2012.
`[7] H. R. Christensen, H. Frøkiær, and J. J. Pestka, “Lactobacilli
`differentially modulate expression of cytokines and maturation
`surface markers in murine dendritic cells,” The Journal of
`Immunology, vol. 168, no. 1, pp. 171–178, 2002.
`[8] P. L´opez, M. Gueimonde, A. Margolles, and A. Su´arez, “Distinct
`Bifidobacterium strains drive different immune responses in
`vitro,” International Journal of Food Microbiology, vol. 138, no.
`1-2, pp. 157–165, 2010.
`
`Genome Ex. 1044
`Page 5 of 6
`
`

`

`6
`
`BioMed Research International
`
`[24] A. Cebula, M. Seweryn, G. A. Rempala et al., “Thymus-
`derived regulatory T cells contribute to tolerance to commensal
`microbiota,” Nature, vol. 497, no. 7448, pp. 258–262, 2013.
`[25] A. Wuyts, S. Struyf, K. Gijsbers et al., “The CXC chemokine
`GCP-2/CXCL6 is predominantly induced in mesenchymal cells
`by interleukin-1𝛽 and is down-regulated by interferon-𝛾: com-
`parison with interleukin-8/CXCL8,” Laboratory Investigation,
`vol. 83, no. 1, pp. 23–34, 2003.
`[26] T. Olszak, D. An, S. Zeissig et al., “Microbial exposure during
`early life has persistent effects on natural killer T cell function,”
`Science, vol. 336, no. 6080, pp. 489–493, 2012.
`[27] A. Sahu and J. D. Lambris, “Structure and biology of comple-
`ment protein C3, a connecting link between innate and acquired
`immunity,” Immunological Reviews, vol. 180, pp. 35–48, 2001.
`[28] M. R. Stone, A. O’Neill, D. Catino, and R. J. Bloch, “Specific
`interaction of the actin-binding domain of dystrophin with
`intermediate filaments containing keratin 19,” Molecular Biology
`of the Cell, vol. 16, no. 9, pp. 4280–4293, 2005.
`[29] P. A. Wearsch and P. Cresswell, “In vitro reconstitution of the
`MHC class I peptide-loading complex,” Methods in Molecular
`Biology, vol. 960, pp. 67–79, 2013.
`[30] P. A. Wearsch and P. Cresswell, “Selective loading of high-
`affinity peptides onto major histocompatibility complex
`class I molecules by the tapasin-ERp57 heterodimer,” Nature
`Immunology, vol. 8, no. 8, pp. 873–881, 2007.
`[31] R. Kaneno, G. V. Shurin, F. M. Kaneno, H. Naiditch, J. Luo, and
`M. R. Shurin, “Chemotherapeutic agents in low noncytotoxic
`concentrations increase immunogenicity of human colon can-
`cer cells,” Cellular Oncology, vol. 34, no. 2, pp. 97–106, 2011.
`
`[9] A. Swidsinski, V. Loening-Baucke, H. Lochs, and L. P. Hale,
`“Spatial organization of bacterial flora in normal and inflamed
`intestine: a fluorescence in situ hybridization study in mice,”
`World Journal of Gastroenterology, vol. 11, no. 8, pp. 1131–1140,
`2005.
`[10] P. Brandtzaeg, “The gut as communicator between environment
`and host: Immunological consequences,” European Journal of
`Pharmacology, vol. 668, supplement 1, pp. S16–S32, 2011.
`[11] S. Arboleya, P. Ruas-Madiedo, A. Margolles et al., “Char-
`acterization and in vitro properties of potentially probiotic
`Bifidobacterium strains isolated from breast-milk,” International
`Journal of Food Microbiology, vol. 149, no. 1, pp. 28–36, 2011.
`[12] P. L´opez, I. Gonz´alez-Rodr´ıguez, M. Gueimonde, A. Margolles,
`and A. Su´arez, “Immune response to Bifidobacterium bifidum
`strains support Treg/Th17 plasticity,” PLoS ONE, vol. 6, no. 9,
`Article ID e24776, 2011.
`[13] P. L´opez, I. Gonz´alez-Rodr´ıguez, B. S´anchez et al., “Interaction
`of Bifidobacterium bifidum LMG13195 with HT29 Cells Influ-
`ences regulatory-T-cell-associated chemokine receptor expres-
`sion,” Applied and Environmental Microbiology, vol. 78, no. 8,
`pp. 2850–2857, 2012.
`[14] D. R. Mack, S. Ahrne, L. Hyde, S. Wei, and M. A. Hollingsworth,
`“Extracellular MUC3 mucin secretion follows adherence of
`Lactobacillus strains to intestinal epithelial cells in vitro,” Gut,
`vol. 52, no. 6, pp. 827–833, 2003.
`[15] M. Schlee, J. Harder, B. K¨oten, E. F. Stange, J. Wehkamp, and
`K. Fellermann, “Probiotic lactobacilli and VSL#3 induce entero-
`cyte 𝛽-defensin 2,” Clinical and Experimental Immunology, vol.
`151, no. 3, pp. 528–535, 2008.
`[16] R. Paolillo, C. R. Carratelli, S. Sorrentino, N. Mazzola, and A.
`Rizzo, “Immunomodulatory effects of Lactobacillus plantarum
`on human colon cancer cells,” International Immunopharmacol-
`ogy, vol. 9, no. 11, pp. 1265–1271, 2009.
`[17] S. di Caro, H. Tao, A. Grillo et al., “Effects of Lactobacillus GG
`on genes expression pattern in small bowel mucosa,” Digestive
`and Liver Disease, vol. 37, no. 5, pp. 320–329, 2005.
`[18] F. J. Troost, P. Van Baarlen, P. Lindsey et al., “Identification
`of the transcriptional response of human intestinal mucosa to
`Lactobacillus plantarum WCFS1 in vivo,” BMC Genomics, vol. 9,
`article 374, 2008.
`[19] P.

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket