Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Infection and Immunity
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Host Response and Inflammation

Lactobacillus paracasei subsp. paracasei B21060 Suppresses Human T-Cell Proliferation

Ilaria Peluso, Daniele Fina, Roberta Caruso, Carmine Stolfi, Flavio Caprioli, Massimo Claudio Fantini, Giorgio Caspani, Enzo Grossi, Laura Di Iorio, Francesco Maria Paone, Francesco Pallone, Giovanni Monteleone
Ilaria Peluso
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daniele Fina
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Roberta Caruso
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carmine Stolfi
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Flavio Caprioli
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Massimo Claudio Fantini
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Giorgio Caspani
2Bracco, SpA, Milan, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Enzo Grossi
2Bracco, SpA, Milan, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laura Di Iorio
3 Cattedra di Pediatria, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Maria Paone
3 Cattedra di Pediatria, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Pallone
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Giovanni Monteleone
1Dipartimento di Medicina Interna, Università Tor Vergata, Rome, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: gi.monteleone@med.uniroma2.it
DOI: 10.1128/IAI.01172-06
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Recent studies have shown that probiotics are beneficial in T-cell-mediated inflammatory diseases. The molecular mechanism by which probiotics work remains elusive, but accumulating evidence indicates that probiotics can modulate immune cell responses. Since T cells express receptors for bacterial products or components, we examined whether different strains of lactobacilli directly regulate the functions of human T cells. CD4+ T cells were isolated from blood and intestinal lamina propria (LP) of normal individuals and patients with inflammatory bowel disease (IBD). Mononuclear cells were also isolated from Peyer's patches. Cells were activated with anti-CD3/CD2/CD28 in the presence or absence of Lactobacillus paracasei subsp. paracasei B21060, L. paracasei subsp. paracasei F19, or L. casei subsp. casei DG. Cell proliferation and death, Foxp3, intracellular pH, and cytokine production were evaluated by flow cytometry. We showed that L. paracasei subsp. paracasei B21060 but neither L. paracasei subsp. paracasei F19 nor L. casei subsp. casei DG inhibited blood CD4+ T-cell growth. This effect was associated with no change in cell survival, expression of Foxp3, or production of gamma interferon, interleukin-4 (IL-4), IL-5, and IL-10. L. paracasei subsp. paracasei B21060-mediated blockade of CD4+ T-cell proliferation required a viable bacterium and was associated with decreased MCT-1 expression and low intracellular pH. L. paracasei subsp. paracasei B21060 also inhibited the growth of Peyer's patch mononuclear cells, normal lymphocytes, and IBD CD4+ LP lymphocytes without affecting cytokine production. The data show that L. paracasei subsp. paracasei B21060 blocks T-cell growth, thus suggesting a mechanism by which these probiotics could interfere with T-cell-driven immune responses.

Human intestine surface is home to a complex and abundant bacterial flora that plays an important role in the maintenance of the health and well-being of the host (28). The luminal microflora promotes normal gastrointestinal functions, protects against pathogenic bacteria, and exerts beneficial effects on systemic metabolism (28). In addition, the indigenous commensals play a decisive role in shaping and maintaining intestinal immune homeostasis (28).

Luminal bacteria can also drive pathological inflammatory responses. This occurs in patients with Crohn's disease (CD) and patients with ulcerative colitis (UC), the major inflammatory bowel diseases (IBD) in humans. Indeed, recent insights into the nature of these diseases suggest that in IBD the tissue damage is driven by a dysregulated T-cell-mediated immune response that is directed against normal constituents of the gut microflora and that manipulation of the luminal bacteria helps limit the mucosal inflammation (4, 8, 31, 32, 35, 38). In this context, it has been shown that feeding nonpathogenic bacteria, in the form of probiotics, is beneficial in the prevention and treatment of intestinal inflammation both in humans and animal models of IBD (1, 7, 10-12). The molecular mechanism underlying the anti-inflammatory action of probiotics, however, is not known. Although effects on alteration of the gut flora, intestinal barrier integrity, and the production of antimicrobial compounds have been described, probiotics could also interfere with the mucosal immune response (32). This is substantiated by the demonstration that probiotics may enhance counter-regulatory mechanisms and regulate the synthesis of inflammatory cytokines (2, 7, 18, 34). Moreover, recent data suggest that specific lactobacillus strains might induce the expression of mu-opioid and cannabinoid receptors in intestinal epithelial cells (30).

Immune cells respond to signals from the microbial environment via pattern recognition receptors, which include Toll-like receptors (TLRs) (16, 27). Recent studies have demonstrated that TLRs can be expressed by T cells, raising the possibility that bacterium-driven signals can modulate immune responses via direct effects on T cells (17, 20). Indeed, the engagement of TLR2 on T regulatory cells (Treg) enhances their growth (37). Moreover, lipopolysaccharide, which acts through TLR4, and flagellin, which signals through TLR5, increase the suppressive functions of Treg (3, 5). Based upon these findings, we sought to determine whether probiotics directly modulate the type and extent of T-cell responses. To this end, we examined the effect of probiotics containing distinct lactobacillus strains on human peripheral and mucosal T-cell functions.

MATERIALS AND METHODS

Samples and preparation of T lymphocytes.Peripheral blood mononuclear cells were isolated from enriched buffy coats from healthy volunteer donors and used to purify CD4+ T cells by the CD4 multisort magnetic microbeads kit (Miltenyi Biotec, Bologna, Italy). The remaining CD4-negative fraction of peripheral blood mononuclear cells was used to purify CD8+ T cells by using CD8 multisort magnetic beads (Miltenyi Biotec). Cell purity was routinely evaluated by flow cytometry and ranged between 96 and 98%.

Intestinal mucosal samples were taken from three patients with CD and one patient with UC undergoing resection for a chronic disease unresponsive to medical treatment. In CD patients the primary site of disease was the terminal ileum and right colon, while in the UC patient the disease was substantial. All patients were receiving corticosteroids at the time of surgery. Mucosal samples were also taken from macroscopically and microscopically unaffected colonic areas of five patients undergoing colectomy for colon cancer. The dissected intestinal mucosa was freed of mucus and epithelial cells in sequential washing steps with dithiothreitol and EDTA and then digested for 4 h at 37°C with collagenase (all reagents were from Sigma-Aldrich, Milan, Italy). Lamina propria (LP) mononuclear cells were separated from the crude cell suspension by layering them on a Percoll solution. For CD4+ LP lymphocyte purification, LP mononuclear cells were incubated for 30 min at 4°C with CD4-magnetically labeled microbeads. The purity of CD4+ LP lymphocyte populations was >92%. Mononuclear cells were isolated from freshly obtained Peyer's patches (PP) taken from four children with irritable bowel syndrome undergoing colonoscopy for recurrent abdominal pain, as previously described (22, 25). The study received approval by the local ethical committee.

Cell culture.Cells were cultured in RPMI 1640 containing 10% fetal bovine serum and standard supplements but no antibiotics (all from Sigma-Aldrich) in 96-multiwell U-bottom plates (105 cells/well/200 μl), with or without anti-CD3/CD2/CD28-bound beads, according to the manufacturer's instructions (Myltenyi Biotec). L. paracasei subsp. paracasei F19 (SIFFRA, Florence, Italy) (14), L. paracasei subsp. paracasei B21060 (Bracco SpA, Milan, Italy) (23), and L. casei subsp. casei DG (Sofar; Trezzano Rosa, Milan, Italy) were provided by the companies as lyophilized products and stored at 4°C until used. Probiotics were dissolved in 1 ml of culture medium just prior to being used, and their concentration and viability were determined by fluorescence-activated cell sorting analysis using SYBR-green and propidium iodide (PI) (13). Probiotics were used at the initial concentration of 104 to 107 bacteria/ml, and analysis of their propagation showed a 100-fold increase in the number of both L. paracasei subsp. paracasei B21060 and L. casei subsp. casei DG at day 3 of culture. In contrast, at the same time point, the number of L. paracasei subsp. paracasei strain F19 increased of nearly 50 times. The amount of d,l-lactate produced by lactobacilli was evaluated in 72-h culture supernatants by spectrophotometry using a commercially available kit (Raisio, Rome, Italy).

To examine the effect of factors secreted by L. paracasei subsp. paracasei B21060 on T-cell function, L. paracasei subsp. paracasei B21060 (106 cells/ml) was resuspended in RPMI 1640 containing no antibiotics and incubated for 72 h at 37°C. The bacterial culture supernatant was then recovered by centrifugation at 1,000 × g for 15 min, filtered through a 0.22-μm-pore-size syringe-driven filter (conditioned medium), and either kept at −80°C until used or boiled for 20 min at 100°C. T cells were cultured in RPMI plus 10% fetal bovine serum containing 0, 12.5, 25, and 50% (vol/vol) freshly prepared or boiled conditioned medium in the presence of anti-CD3/CD2/CD28 beads. In addition, cultures of activated CD4+ T cells were added with heat-killed L. paracasei strain B21060 (106 cells/ml) that was prepared by boiling the probiotics for 20 min at 100°C. To ascertain the efficiency of such a treatment, an aliquot of the heat-treated probiotics was resuspended in phosphate-buffered saline, and the viability was determined as indicated above. According to this procedure, cells were considered dead only if they were stained with PI but were SYBR-green negative. To examine the effect of lactate on T-cell growth, graded doses of exogenous d,l-lactate (5 to 50 μg/ml; Sigma-Aldrich) were added to cultures of activated CD4+ T cells. To track the proliferation, CD4+ or CD8+ T cells were incubated in 0.2 μM carboxyfluorescein diacetate succinimidyl ester (CFSE; Invitrogen, Milan Italy) at 37°C for 30 min. After 3 days culture, fluorescence was detected, and the proportion of cells undergoing divisions was determined.

To evaluate the effect of lactobacilli on cell survival, CD4+ T cells were cultured as described above, and the fraction of annexin V (AV)- and PI-positive cells was evaluated by flow cytometry using a commercially available kit (Beckman Coulter, Milan, Italy).

Cell phenotype analysis.Anti-CD4 fluorescein isothiocyanate and anti-CD8 APC (both from Beckman Coulter), anti-Foxp3 APC (Società Italiana Chimici, Florence, Italy), and control isotype antibodies were used for analysis of relative antigens according to the manufacturer' instructions.

Cytokine assays.Gamma interferon (IFN-γ), interleukin-4 (IL-4), IL-5, and IL-10 were analyzed in cell culture supernatants by flow cytometry using a commercially available kit (Bender, Vienna, Austria).

pH measurements.Cells were incubated in RPMI with 2 μM 2′,7′-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein, acetoxymethyl ester (BCECF-AM; Sigma-Aldrich) at 37°C for 30 min as previously described (24). The mean intensity green and red fluorescence levels were then detected by flow cytometry, and the pH was calculated by using a standard curve with potassium phosphate buffers (pH ranging from 6 to 8). Experiments were performed in the presence of 10 μM nigericin (Sigma-Aldrich).

Western blotting.MCT-1 was analyzed in total extracts of CD4+ T cells using goat anti-human MCT-1 antibody (final dilution, 1:3,000; ABCAM plc, Cambridge, United Kingdom), followed by horseradish peroxidase-conjugated rabbit anti-goat immunoglobulin G (final dilution, 1:30,000; Dako SpA, Milan, Italy). The reaction was detected with a sensitive enhanced chemiluminescence kit (West DURA; Pierce, Rockford, IL). After the analysis of MCT-1, blots were stripped and incubated with mouse anti-human β-actin antibody (final dilution, 1:5,000; Sigma-Aldrich), followed by goat anti-mouse antibody conjugated to horseradish peroxidase (1:30,000 dilution) and detection by chemiluminescence.

Statistics.Two-way analysis of variance, followed by pairwise multiple comparison procedures (the Student-Newman-Keuls method) and the paired Student t test, was performed. The data are expressed as means ± the standard errors of the mean (SEM).

RESULTS

L. paracasei subsp. paracasei B21060 inhibits blood CD4+ T-cell proliferation.Culture of CD4+ T lymphocytes with anti-CD3/CD2/CD28 resulted in a significant increase in the percentage of proliferating cells (Fig. 1A, P < 0.001). The addition of L. paracasei subsp. paracasei B21060 (106 cells/ml) to such cultures significantly inhibited the cell growth (P < 0.01). In contrast, equivalent concentrations of L. paracasei subsp. paracasei F19 or L. casei subsp. casei DG did not inhibit cell proliferation (Fig. 1A). To exclude that the antimitogenic effect of L. paracasei subsp. paracasei B21060 was secondary to changes in cell viability, we evaluated the fraction of AV- and/or PI-positive cells in the same cultures. Figure 1B shows that L. paracasei subsp. paracasei B21060 did not augment the percentage of AV- and/or PI-positive cells.

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

L. paracasei subsp. paracasei B21060 suppresses the growth of activated blood CD4+ T cells. (A) CFSE-labeled CD4+ T cells (5 × 105 cells/ml) were stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml) or L. paracasei subsp. paracasei F19 (F19) (106 cells/ml) or L. casei subsp. casei DG (106 cells/ml). After 3 days, the percentage of proliferating cells was evaluated by flow cytometry. The data indicate mean ± the SEM of six different experiments. (B) Effect of L. paracasei subsp. paracasei B21060 on CD4+ T-cell death. Cells were cultured as indicated for panel A, and the percentage of cell death was assessed by fluorescence-activated cell sorting analysis of AV- and/or PI-positive cells. The data indicate means ± the SEM of three separate experiments.

We also established the optimal concentration at which L. paracasei subsp. paracasei B21060 inhibits CD4+ T-cell growth. At the initial concentrations of 104 or 105 cells/ml, L. paracasei subsp. paracasei B21060 reduced the percentage of proliferating CD4+ T cells (21 ± 3% in the absence of L. paracasei subsp. paracasei B21060 versus 16 ± 2 and 14 ± 3% in the presence of 104 or 105 cells/ml, respectively), but such effects were not significant. A marked bacterial overgrowth and complete CD4+ T-cell death were seen in cultures added with 107 cells/ml (not shown). Therefore, in the subsequent experiments L. paracasei subsp. paracasei B21060 was used at 106 cells/ml.

L. paracasei subsp. paracasei B21060 alters neither the percentage Foxp3+ T cells nor the synthesis of cytokines.Treg are a subpopulation of CD4+ CD25+ T lymphocytes that specifically express the forkhead transcription factor forkhead winged helix transcription factor gene (Foxp3) and are highly specialized for the suppression of proliferation of effector T cells (39). In addition to naturally occurring Treg that are produced by the thymus, Treg can arise in the periphery upon conversion of CD4+ CD25− T cells into Foxp3+ CD4+ CD25+ cells in response to a variety of stimuli, including bacterial products (9). Therefore, we examined whether the antiproliferative effect of L. paracasei subsp. paracasei B21060 was associated with changes in the percentage of Foxp3-positive cells. Culture of CD4+ T cells with anti-CD3/CD2/CD28 significantly augmented the fraction of Foxp-3-positive cells (Fig. 2A, P < 0.01). However, this percentage was not increased further by L. paracasei subsp. paracasei B21060, thus arguing against the hypothesis that the antimitogenic effect of L. paracasei subsp. paracasei B21060 is due to an expansion of Treg. This was substantiated further by the demonstration that L. paracasei strain B21060 exerted antimitogenic effects also in cultures of CD8+ T cells that do not contain Treg (Fig. 2B).

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

(A) Percentage of Foxp3+ cells induced from blood CD4+ T cells after culture with medium alone (Unst) or anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml), L. paracasei subsp. paracasei F19 (106 cells/ml), or L. casei subsp. casei DG (106 cells/ml). After 3 day culture, Foxp3 was analyzed by flow cytometry. The data indicate means ± the SEM of four separate experiments. (B) L. paracasei subsp. paracasei B21060 inhibits the proliferation of activated blood CD8+ T cells. CFSE-labeled CD8+ T cells (5 × 105 cells/ml) were either left unstimulated (Unst) or stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml). After 3 days, the percentage of proliferating cells was evaluated by flow cytometry. The data indicate means ± the SEM of three different experiments.

Subsequently, we examined whether L. paracasei subsp. paracasei B21060 modulates the production of CD4+ T cell-derived cytokines. Culture of cells with anti-CD3/CD2/CD28 significantly enhanced the synthesis of IFN-γ (Fig. 3A, P < 0.001). However, no further increase was seen when these cell cultures were added with any lactobacilli or Lactobacillus subsp. (Fig. 3A). Similarly, probiotics were not able to alter the secretion of IL-4 and IL-5 (Fig. 3B). Since some probiotics enhance IL-10 synthesis and IL-10 exerts antiproliferative effects on CD4+ T cells (7), we also measured IL-10. Activation of cells caused a significant increase in IL-10, but such a synthesis was not modified by probiotics (Fig. 3C).

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

CD4+ T cells (5 × 105 cells/ml) were either left unstimulated (Unst) or stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml), L. paracasei subsp. paracasei F19 (106 cells/ml), or L. casei subsp. casei DG (106 cells/ml). After 3 days, the cell-free culture supernatants were collected and analyzed for the IFN-γ (A), IL-4 and IL-5 (B), and IL-10 (C) contents by flow cytometry. The data are expressed as pg/ml and indicate the means ± the SEM of five separate experiments.

L. paracasei subsp. paracasei B21060 inhibits the expression of monocarboxylate transporter MCT-1.MCT-1, a membrane protein of the MCT family, is involved in the proton-linked transport of lactate, pyruvate, and other monocarboxylates across the plasma membrane (6). Inhibition of MCT-1 in activated T cells blocks the proliferation but not cytokine production (24), thus showing striking similarities with the effect of L. paracasei subsp. paracasei B21060 on CD4+ T-cell activation. Therefore, we evaluated the effect of L. paracasei subsp. paracasei B21060 on MCT-1. Western blotting of total extracts revealed that activation of CD4+ T cells augmented the expression of MCT-1. This upregulation was evident at 24 h, reached the maximal induction at 48 h, and then declined (Fig. 4). L. paracasei subsp. paracasei B21060 inhibited MCT-1 expression, and this was evident at each time point (Fig. 4A).

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

L. paracasei subsp. paracasei B21060 inhibits the expression of MCT-1 and reduces the intracellular pH in activated CD4+ T cells. CD4+ T cells were either left unstimulated (Unst) or stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml). Total extracts were analyzed for MCT-1 by Western blotting. One of three separate experiments is shown. (B) Cells were cultured as indicated for panel A, and the intracellular pH was assessed by flow cytometry. The data indicate means ± the SEM of three different experiments. (C) Representative CFSE dilution profiles of CD4+ T cells stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of the specified doses of exogenous d,l-lactate for 72 h. The results for one of two representative experiments is shown.

The inhibition of MCT-1 is followed by an intracellular accumulation of lactate and hence reduction of the intracellular pH (26). Indeed, L. paracasei subsp. paracasei B21060 significantly reduced the intracellular pH (Fig. 4B). Since lactic acid can modulate T-cell growth, we also evaluated the level of d,l-lactate in culture supernatants of Lactobacilli. Notably, after 72 h culture, the production of d,l-lactate by L. paracasei subsp. paracasei B21060 was significantly greater (354 ± 14 μg/ml) than that produced by L. casei subsp. casei DG (254 ± 22 μg/ml) and by L. paracasei subsp. paracasei F19 (165 ± 5 μg/ml) (P < 0.05 and P < 0.01, respectively). In addition, we showed that the addition of 10 and 20 μg of exogenous d,l-lactate/ml to CD4+ T-cell cultures markedly inhibited cell growth (Fig. 4C), whereas no inhibitory effect was seen when d,l-lactate was used at 5 μg/ml (not shown). Moreover, incubation of CD4+ T cells with concentrations of d,l-lactate greater than 50 μg/ml resulted in massive cell death (results not shown). Overall, these findings suggest that the antimitogenic effect of L. paracasei subsp. paracasei B21060 may be in part mediated by lactate.

The L. paracasei subsp. paracasei B21060-induced inhibition of CD4+ T-cell proliferation is mediated by thermostable secreted factor(s).To assess whether the effect of L. paracasei subsp. paracasei B21060 on CD4+ T-cell proliferation requires a viable bacterium, CFSE-labeled CD4+ T cells were cultured with anti-CD3/CD2/CD28 in the presence or absence of live or heat-killed L. paracasei subsp. paracasei B21060 (Fig. 5A). Figure 5B shows that heat-killed L. paracasei subsp. paracasei B21060 did not inhibit CD4+ T-cell proliferation. Then, we evaluated whether the antiproliferative effect of L. paracasei subsp. paracasei B21060 was mediated by soluble factor(s). To begin to address this issue, we used L. paracasei subsp. paracasei B21060-derived conditioned medium. To evaluate whether the effect of such conditioned medium was due to thermolabile molecules, cell cultures were also treated with boiled conditioned medium. Both the freshly prepared and the boiled conditioned media dose dependently inhibited the growth of activated CD4+ T cells (Fig. 5).

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

(A) Representative dot plots of SYBR green (SYBR) and PI of L. paracasei subsp. paracasei B21060. The lactobacilli were stained before (L. paracasei subsp. paracasei B21060 [P]) or after heating at 100°C for 20 min (heat-killed L. paracasei subsp. paracasei B21060 [HKP]). Numbers indicate the percentage of bacteria in the designated gates. Viable bacteria are PI negative and SYBR positive, whereas dead bacteria are PI positive and SYBR negative. Double positivity indicates bacteria with a slightly damaged membrane but still alive. One of three representative experiments is shown. (B) Viable but not heat-killed L. paracasei subsp. paracasei B21060 inhibits the proliferation of activated blood CD4+ T cells. CFSE-labeled CD4+ T cells (5 × 105 cells/ml) were stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of live L. paracasei subsp. paracasei B21060 (P) (106 cells/ml) or heat-killed L. paracasei subsp. paracasei B21060 (106 cells/ml). After 3 days, the percentage of proliferating cells was evaluated by flow cytometry. The data indicate means ± the SEM of three different experiments. (C) Freshly prepared and boiled conditioned media of L. paracasei subsp. paracasei B21060 inhibit the growth of activated blood CD4+ T cells. CFSE-labeled CD4+ T cells (5 × 105 cells/ml) were stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of either freshly prepared or boiled conditioned medium at the indicated dilutions. After 3 days, the percentage of proliferating cells was evaluated by flow cytometry. The data indicate means ± SEM of 3 different experiments.

L. paracasei subsp. paracasei B21060 suppresses the proliferation of PP and intestinal LP lymphocytes.PP is one of the major sites where the intestinal immune system interacts and responds to luminal antigens (19). Therefore, we examined whether L. paracasei subsp. paracasei B21060 modulates the response of freshly isolated PP lymphocytes. In these experiments, we used unfractionated PP mononuclear cells (PPMC) since the number of cells we isolated from small endoscopic biopsies was not sufficient to purify CD4+ T cells. Activation of PPMC with anti-CD3/CD2/CD28 enhanced the percentage of proliferating cells, and this effect was inhibited by L. paracasei subsp. paracasei B21060 (Fig. 6A). Activated cells produced higher levels of IFN-γ than unstimulated cells (P < 0.01), but such a synthesis was not significantly modified by L. paracasei subsp. paracasei B21060 (Fig. 6B). In contrast, the synthesis of IL-4 and IL-5 was not enhanced by activation of cells, and this was evident regardless of whether cells were left untreated or treated with L. paracasei subsp. paracasei B21060 (Fig. 5C). The production of IL-10 was augmented by the activation of PPMC, but again this synthesis was not increased by L. paracasei subsp. paracasei B21060 (Fig. 6D).

FIG. 6.
  • Open in new tab
  • Download powerpoint
FIG. 6.

L. paracasei strain B21060 suppresses the growth of activated PPMC without affecting the synthesis of cytokines. PPMC (5 × 105 cells/ml) were stimulated with anti-CD3/CD2/CD28 (ACT) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml). After 3 days, the percentage of CFSE-labeled proliferating cells (A) and the IFN-γ (B), IL-4 and IL-5 (C), and IL-10 (D) contents in the culture supernatants were evaluated by flow cytometry. The data indicate the means ± the SEM of four separate experiments.

Since intestinal LP contains a large population of effector-memory T cells that exhibit marked phenotypic and functional differences with peripheral blood and PP cells (19), we also examined whether L. paracasei subsp. paracasei B21060 affects the proliferation and cytokine production of LP CD4+ T lymphocytes. Initially, we evaluated the effects of L. paracasei subsp. paracasei B21060 on the proliferation and cytokine response of normal intestinal LP CD4+ cells activated with anti-CD3/CD2/CD28. L. paracasei subsp. paracasei B21060 significantly inhibited the proliferation of activated LP CD4+ T cells (Fig. 7A, P < 0.05) but did not alter the production of any cytokine (Fig. 7B to D). In addition, the significant increase in the percentage of Foxp3+ cells induced by anti-CD3/CD2/CD28 (P < 0.05) was not modified by L. paracasei subsp. paracasei B21060 (Fig. 7E). Subsequently, we assessed the antimitogenic effect of L. paracasei subsp. paracasei B21060 on LP CD4+ T cells isolated from IBD patients. As shown in Fig. 8, the proliferation of CD4+ T cells isolated from the guts of three patients with CD and one patient with UC was significantly inhibited by L. paracasei subsp. paracasei B21060 (P < 0.05).

FIG. 7.
  • Open in new tab
  • Download powerpoint
FIG. 7.

L. paracasei strain B21060 suppresses the growth of normal intestinal LP CD4+ T cells without affecting the cytokine synthesis. CFSE-labeled CD4+ T cells (5 × 105 cells/ml) were stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml). After 3 days, the percentage of proliferating cells (A); the IFN-γ (B), IL-4 and IL-5, (C), and IL-10 (D) contents in the culture supernatants; and the percentage of Foxp-3-positive cells were evaluated by flow cytometry. The data indicate means ± the SEM of five separate experiments.

FIG. 8.
  • Open in new tab
  • Download powerpoint
FIG. 8.

L. paracasei subsp. paracasei B21060 suppresses the growth of IBD intestinal LP CD4+ T cells. CFSE-labeled CD4+ T cells (5 × 105 cells/ml), isolated from three patients with CD and one patient with UC, were stimulated with anti-CD3/CD2/CD28 (Act) in the presence or absence of L. paracasei subsp. paracasei B21060 (P) (106 cells/ml). After 3 days, the percentage of proliferating cells was evaluated by flow cytometry.

DISCUSSION

This study was undertaken to examine whether different strains and subspecies of lactobacilli modulate the functions of CD4+ T cells. We show that L. paracasei subsp. paracasei B21060 directly inhibits the in vitro proliferation of human CD4+ T cells, a finding consistent with the demonstration that T cells are able to directly respond to bacteria (17, 20). Our study also reveals that the L. paracasei subsp. paracasei B21060-mediated antimitogenic effect is specific, since two other different lactobacillus strains or Lactobacillus subsp., namely, L. paracasei subsp. paracasei F19 and L. casei subsp. casei DG, did not suppress CD4+ T-cell growth. This effect would not seem to rely simply on a different propagation of the lactobacillus strains or Lactobacillus subsp. in our system, since the number of L. paracasei subsp. paracasei B21060 was similar to that of L. casei subsp. casei DG at any time point during the culture. Therefore, our data further support earlier observations that different probiotics strains and/or subspecies may have distinct regulatory properties and exert variable effects on the course of inflammatory diseases (32, 34). It is also known that lactobacilli or Lactobacillus subsp. may differently modulate specific immune responses depending on the cell system considered. For example, lactobacilli are powerful inducers of Th1-type cytokines, such as IL-12 and TNF-α, in blood cells, whereas they downregulate TNF-α when used in ex vivo organ cultures of IBD mucosal explants (2, 15, 21). While our study was under way, Sturm et al. (36) demonstrated that a conditioned medium of Escherichia coli strain Nissle 1917 inhibited the growth of blood and mucosal T cells and that such an effect was associated with decreased synthesis of IFN-γ and high IL-10 secretion. This later finding fits well with the data of other studies that have linked the beneficial effect of probiotics with the induction of IL-10 in experimental models of IBD (7). However, no increase in IL-10 was seen in our cell cultures with L. paracasei subsp. paracasei B21060. Moreover, L. paracasei subsp. paracasei B21060 did not alter the percentage of Foxp3+ cells, suggesting that the antiproliferative effect of L. paracasei subsp. paracasei B21060 does not rely on the expansion of counter-regulatory mechanisms.

A surprising finding of our study is that the CD4+ T-cell growth arrest induced by L. paracasei subsp. paracasei B21060 was associated with no change in the production of Th1 or Th2 cytokines. These immunomodulatory effects of L. paracasei subsp. paracasei B21060 resemble those exerted by inhibitors of the monocarboxylate transporter MCT-1, given that such compounds suppress T-cell growth without affecting cytokine production (24). Therefore, we extended our analysis by examining the effect of L. paracasei subsp. paracasei B21060 on MCT-1. By Western blotting we show that L. paracasei subsp. paracasei B21060 inhibits the expression of MCT-1 and consequently reduces the intracellular pH in CD4+ T cells. Although, no functional experiment was carried out to mechanistically link the downregulation of MCT-1 and the reduction of intracellular pH with the cell growth arrest, it is noteworthy that the intracellular pH is crucial in controlling cell growth and that acidification of the intracellular compartment leads to cell growth arrest (33). In line with this, we show that the amount of d,l-lactate made by L. paracasei subsp. paracasei B21060 was greater than that produced by the other two lactobacillus strains and that the addition of exogenous lactate to CD4+ T-cell cultures caused a marked inhibition of CD4+ T-cell proliferation.

While the definition of probiotics is one of live microorganisms, recent studies have demonstrated that bacterial DNA could be responsible for some beneficial effects of probiotics (29). However, our data indicate that the L. paracasei subsp. paracasei B21060- immunomodulatory effects require the presence of a viable bacterium, as heat-killed L. paracasei subsp. paracasei B21060 did not inhibit the growth of CD4+ T cells. The results presented here suggest that the L. paracasei subsp. paracasei B21060-induced antiproliferative effect is mediated by secretion of lactic acid.

In conclusion, our study shows that L. paracasei subsp. paracasei B21060 blocks the proliferation of T lymphocytes, and this occurs in cultures of CD4+ T cells isolated from the normal and inflamed intestinal LP, as well as in cultures of PP cells. It is thus tempting to speculate that L. paracasei subsp. paracasei B21060 might affect both the inductive and the effector phases of the human mucosal T-cell response. However, additional in vivo studies will be necessary to ascertain whether results from these in vitro experiments can be translated to the clinical practice and examine whether L. paracasei subsp. paracasei B21060 can interfere with T-cell-driven immune responses.

ACKNOWLEDGMENTS

This study received support from the Fondazione Umberto di Mario, Rome, Italy; the Ministero dell' Istruzione, dell' Università e della Ricerca (no. 2004065777-004); and the Ministero della Salute (no. 6AC/F7).

FOOTNOTES

    • Received 25 July 2006.
    • Returned for modification 5 November 2006.
    • Accepted 9 January 2007.
  • Copyright © 2007 American Society for Microbiology

REFERENCES

  1. 1.↵
    Bibiloni, R., R. N. Fedorak, G. W. Tannock, K. L. Madsen, P. Gionchetti, M. Campieri, C. De Simone, and R. B. Sartor. 2005. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am. J. Gastroenterol.100:1539-1546.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    Borruel, N., M. Carol, F.Casellas, M. Antolin, F. de Lara, E. Espin, J. Naval, F. Guarner, and J. R. Malagelada. 2002. Increased mucosal tumour necrosis factor alpha production in Crohn's disease can be downregulated ex vivo by probiotic bacteria. Gut51:659-664.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    Caramalho, I., T. Lopes-Carvalho, D. Ostler, S. Zelenay, M. Haury, and J. Demengeot. 2003. Regulatory T cells selectively express Toll-like receptors and are activated by lipopolysaccharide. J. Exp. Med.197:403-411.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    Cong, Y., S. L. Brandwein, R. P. McCabe, A. Lazenby, E. H. Birkenmeier, J. P. Sundberg, and C. O. Elson. 1998. CD4+ T cells reactive to enteric bacterial antigens in spontaneously colitic C3H/HeJBir mice: increased T helper cell type 1 response and ability to transfer disease. J. Exp. Med.187:855-864.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    Crellin, N. K., R. V. Garcia, O. Hadisfar, S. E. Allan, T. S. Steiner, and M. K. Levings. 2005. Human CD4+ T cells express TLR5 and its ligand flagellin enhances the suppressive capacity and expression of FOXP3 in CD4+ CD25+ T regulatory cells. J. Immunol.175:8051-8059.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    Daberkow, R. L., B. R. White, R. A. Cederberg, J. B. Griffin, and J. Zempleni. 2003. Monocarboxylate transporter 1 mediates biotin uptake in human peripheral blood mononuclear cells. J. Nutr.133:2703-2706.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Di Giacinto, C., M. Marinaro, M. Sanchez, W. Strober, and M. Boirivant. 2005. Probiotics ameliorate recurrent Th1-mediated murine colitis by inducing IL-10- and IL-10-dependent TGF-beta-bearing regulatory cells. J. Immunol.174:3237-3246.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    Duchmann, R., I. Kaiser, E. Hermann, W. Mayet, K. Ewe, and K. H. Meyer zum Buschenfelde. 1995 Tolerance exists toward resident intestinal flora but is broken in active inflammatory bowel disease (IBD). Clin. Exp. Immunol.102:448-455.
    OpenUrlPubMedWeb of Science
  9. 9.↵
    Fantini, M. C., C. Becker, G. Monteleone, F. Pallone, P. R. Galle, and M. F. Neurath. 2004. Cutting edge: TGF-beta induces a regulatory phenotype in CD4+ CD25− T cells through Foxp3 induction and downregulation of Smad7. J. Immunol.172:5149-5153.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    Gionchetti, P., F. Rizzello, A. Venturi, P. Brigidi, D. Matteuzzi, G. Bazzocchi, G. Poggioli, M. Miglioli, and M. Campieri. 2000. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology119:305-309.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.
    Gionchetti, P., F. Rizzello, U. Helwig, A. Venturi, K. M. Lammers, P. Brigidi, B. Vitali, G. Poggioli, M. Miglioli, and M. Campieri. 2003. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology124:1202-1209.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    Grabig, A., D. Paclik, C. Guzy, A. Dankof, D. C. Baumgart, J. Erckenbrecht, B. Raupach, U. Sonnenborn, J. Eckert, R. R. Schumann, B. Wiedenmann, A. U. Dignass, and A. Sturm. 2006. Escherichia coli strain Nissle 1917 ameliorates experimental colitis via Toll-like receptor 2- and Toll-like receptor 4-dependent pathways. Infect. Immun.74:4075-4082.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    Gregori, G., S. Citterio, A. Ghiani, M. Labra, S. Sgorbati, S. Brown, and M. Denis. 2001. Resolution of viable and membrane-compromised bacteria in freshwater and marine waters based on analytical flow cytometry and nucleic acid double staining. Appl. Environ. Microbiol.67:4662-4670.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    Heilig, H. G., E. G. Zoetendal, E. E. Vaughan, P. Marteau, A. D. Akkermans, and W. M. de Vos. 2002. Molecular diversity of Lactobacillus spp. and other lactic acid bacteria in the human intestine as determined by specific amplification of 16S ribosomal DNA. Appl. Environ. Microbiol.68:114-123.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    Hessle, C., L. A. Hanson, and A. E. Wold. 1999. Lactobacilli from human gastrointestinal mucosa are strong stimulators of IL-12 production. Clin. Exp. Immunol.116:276-282.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Iwasaki, A., and R. Medzhitov. 2004. Toll-like receptor control of the adaptive immune responses. Nat. Immunol.5:987-995.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    Komai-Koma, M., L. Jones, G. S. Ogg, D. Xu, and F. Y. Liew. 2004. TLR2 is expressed on activated T cells as a costimulatory receptor. Proc. Natl. Acad. Sci. USA101:3029-3034.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    Maassen, C. B., C. van Holten-Neelen, F. Balk, M. J. den Bak-Glashouwer, R. J. Leer, J. D. Laman, W. J. Boersma, and E. Claassen. 2000. Strain-dependent induction of cytokine profiles in the gut by orally administered Lactobacillus strains. Vaccine18:2613-2623.
    OpenUrlCrossRefPubMedWeb of Science
  19. 19.↵
    Macdonald, T. T., and G. Monteleone. 2005. Immunity, inflammation, and allergy in the gut. Science307:1920-1925.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Mansson, A., M. Adner, and L. O. Cardell. 2006. Toll-like receptors in cellular subsets of human tonsil T cells: altered expression during recurrent tonsillitis. Respir. Res.7:36.
    OpenUrlCrossRefPubMed
  21. 21.↵
    Miettinen, M., J. Vuopio-Varkila, and K. Varkila. 1996. Production of human tumor necrosis factor alpha, interleukin-6, and interleukin-10 is induced by lactic acid bacteria. Infect. Immun.64:5403-5405.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    Monteleone, G., J. Holloway, V. M. Salvati, S. L. Pender, P. D. Fairclough, N. Croft, and T. T. MacDonald. 2003. Activated STAT4 and a functional role for IL-12 in human Peyer's patches. J. Immunol.170:300-307.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    Morelli, L., D. Zonenschain, M. L. Callegari, E. Grossi, F. Malsano, and M. Fusillo. 2003. Assessment of a new synbiotic preparation in healthy volunteers: survival, persistence of probiotic strains and its effect on the indigenous flora. Nutr. J.2:11.
    OpenUrlCrossRefPubMed
  24. 24.↵
    Murray, C. M., R. Hutchinson, J. R. Bantick, G. P. Belfield, A. D. Benjamin, D. Brazma, R. V. Bundick, I. D. Cook, R. I. Craggs, S. Edwards, L. R. Evans, R. Harrison, E. Holness, A. P. Jackson, C. G. Jackson, L. P. Kingston, M. W. Perry, A. R. Ross, P. A. Rugman, S. S. Sidhu, M. Sullivan, D. A. Taylor-Fishwick, P. C. Walker, Y. M. Whitehead, D. J. Wilkinson, A. Wright, and D. K. Donald. 2005. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat. Chem. Biol.1:371-376.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    Nagata, S., C. McKenzie, S. L. Pender, M. Bajaj-Elliott, P. D. Fairclough, J. A. Walker-Smith, G. Monteleone, and T. T. MacDonald. 2000. Human Peyer's patch T cells are sensitized to dietary antigen and display a Th cell type 1 cytokine profile. J. Immunol.165:5315-5321.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    Nilsson, C., K. Kagedal, U. Johansson, and K. Ollinger. 2003. Analysis of cytosolic and lysosomal pH in apoptotic cells by flow cytometry. Methods Cell Sci.25:185-194.
    OpenUrlCrossRefPubMed
  27. 27.↵
    Pasare, C., and R. Medzhitov. 2005. Toll-like receptors: linking innate and adaptive immunity. Adv. Exp. Med. Biol.560:11-18.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    Pickard, K. M., A. R. Bremner, J. N. Gordon, and T. T. MacDonald. 2004. Microbial-gut interactions in health and disease. Immune responses. Best. Pract. Res. Clin. Gastroenterol.18:271-285.
    OpenUrlCrossRef
  29. 29.↵
    Rachmilewitz, D., K. Katakura, F. Karmeli, T. Hayashi, C. Reinus, B. Rudensky, S. Akira, K. Takeda, J. Lee, K. Takabayashi, and E. Raz. 2004. Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology126:520-528.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Rousseaux, C., X. Thuru, A. Gelot, N. Barnich, C. Neut, L. Dubuquoy, C. Dubuquoy, E. Merour, K. Geboes, M. Chamaillard, A. Ouwehand, G. Leyer, D. Carcano, J. F. Colombel, D. Ardid, and P. Desreumaux. 2007. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat. Med.15:35-37.
    OpenUrl
  31. 31.↵
    Rutgeerts, P., K. Goboes, M. Peeters, M. Hiele, F. Penninckx, R. Aerts, R. Kerremans, and G. Vantrappen. 1991. Effect of faecal stream diversion on recurrence of Crohn's disease in the neoterminal ileum. Lancet338:771-774.
    OpenUrlCrossRefPubMedWeb of Science
  32. 32.↵
    Sartor, R. B. 2004. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology126:1620-1633.
    OpenUrlCrossRefPubMedWeb of Science
  33. 33.↵
    Schreiber, R. 2005. Ca2+ signaling, intracellular pH, and cell volume in cell proliferation. J. Membr. Biol.205:129-137.
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    Shanahan, F. 2000. Probiotics and inflammatory bowel disease: is there a scientific rationale? Inflamm. Bowel. Dis.6:107-115.
    OpenUrlCrossRefPubMedWeb of Science
  35. 35.↵
    Strober, W., I. J. Fuss, and R. S. Blumberg. 2002. The immunology of mucosal models of inflammation. Annu. Rev. Immunol.20:495-549.
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.↵
    Sturm, A., K. Rilling, D. C. Baumgart, K. Gargas, T. Abou-Ghazale, B. Raupach, J. Eckert, R. R. Schumann, C. Enders, U. Sonnenborn, B. Wiedenmann, and A. U. Dignass. 2005. Escherichia coli Nissle 1917 distinctively modulates T-cell cycling and expansion via Toll-like receptor 2 signaling. Infect. Immun.73:1452-1465.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    Sutmuller, R. P., M. H. den Brok, M. Kramer, E. J. Bennink, L. W. Toonen, B. J. Kullberg, L. A. Joosten, S. Akira, M. G. Netea, and G. J. Adema. 2006. Toll-like receptor 2 controls expansion and function of regulatory T cells. J. Clin. Investig.116:485-494.
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    Thompson-Chagoyan, O. C., J. Maldonado, and A. Gil. 2005. Aetiology of inflammatory bowel disease (IBD): role of intestinal microbiota and gut-associated lymphoid tissue immune response. Clin. Nutr.24:339-352.
    OpenUrlCrossRefPubMedWeb of Science
  39. 39.↵
    Ziegler, S. F. 2006. FOXP3: of mice and men. Annu. Rev. Immunol.24:209-226.
    OpenUrlCrossRefPubMedWeb of Science
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
Lactobacillus paracasei subsp. paracasei B21060 Suppresses Human T-Cell Proliferation
Ilaria Peluso, Daniele Fina, Roberta Caruso, Carmine Stolfi, Flavio Caprioli, Massimo Claudio Fantini, Giorgio Caspani, Enzo Grossi, Laura Di Iorio, Francesco Maria Paone, Francesco Pallone, Giovanni Monteleone
Infection and Immunity Mar 2007, 75 (4) 1730-1737; DOI: 10.1128/IAI.01172-06

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Infection and Immunity article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Lactobacillus paracasei subsp. paracasei B21060 Suppresses Human T-Cell Proliferation
(Your Name) has forwarded a page to you from Infection and Immunity
(Your Name) thought you would be interested in this article in Infection and Immunity.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Lactobacillus paracasei subsp. paracasei B21060 Suppresses Human T-Cell Proliferation
Ilaria Peluso, Daniele Fina, Roberta Caruso, Carmine Stolfi, Flavio Caprioli, Massimo Claudio Fantini, Giorgio Caspani, Enzo Grossi, Laura Di Iorio, Francesco Maria Paone, Francesco Pallone, Giovanni Monteleone
Infection and Immunity Mar 2007, 75 (4) 1730-1737; DOI: 10.1128/IAI.01172-06
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

CD4-Positive T-Lymphocytes
cell proliferation
Lactobacillus
probiotics

Related Articles

Cited By...

About

  • About IAI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #IAIjournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0019-9567; Online ISSN: 1098-5522