Local and systemic immune mechanisms underlying the anti-colitis effects of the dairy bacterium Lactobacillus delbrueckii

Clarissa Santos Rocha, Ana Cristina Gomes-Santos, Thais Garcias Moreira, Marcela de Azevedo, Tessalia Diniz Luerce, Mahendra Mariadassou, Ana Paula Longaray Delamare, Philippe Langella, Emmanuelle Maguin, Vasco Azevedo, Ana Maria Caetano de Faria, Anderson Miyoshi, Maarten van de Guchte, Clarissa Santos Rocha, Ana Cristina Gomes-Santos, Thais Garcias Moreira, Marcela de Azevedo, Tessalia Diniz Luerce, Mahendra Mariadassou, Ana Paula Longaray Delamare, Philippe Langella, Emmanuelle Maguin, Vasco Azevedo, Ana Maria Caetano de Faria, Anderson Miyoshi, Maarten van de Guchte

Abstract

Several probiotic bacteria have been proposed for treatment or prevention of inflammatory bowel diseases (IBD), showing a protective effect in animal models of experimental colitis and for some of them also in human clinical trials. While most of these probiotic bacteria are isolated from the digestive tract, we recently reported that a Lactobacillus strain isolated from cheese, L. delbrueckii subsp. lactis CNRZ327 (Lb CNRZ327), also possesses anti-inflammatory effects in vitro and in vivo, demonstrating that common dairy bacteria may be useful in the treatment or prevention of IBD. Here, we studied the mechanisms underlying the protective effects of Lb CNRZ327 in vivo, in a mouse dextran sodium sulfate (DSS) colitis model. During colitis, Lb CNRZ327 modulated the production of TGF-β, IL-6, and IL-12 in colonic tissue and of TGF-β and IL-6 in the spleen, and caused an expansion of CD4+Foxp3+ regulatory T cells in the cecal lymph nodes. Moreover, a strong tendency to CD4+Foxp3+ expansion was also observed in the spleen. The results of this study for the first time show that orally administered dairy lactobacilli can not only modulate mucosal but also systemic immune responses and constitute an effective treatment of IBD.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Effect of Lb CNRZ327 on…
Figure 1. Effect of Lb CNRZ327 on body weight and DAI in DSS-induced colitis.
A. Body weight change from day 1 to day 8. Each bar represents the average body weight change ± SEM of the 5 mice in the group (one experiment, see text for further details) as a percentage of their initial weights. B. Disease activity index (DAI) on day 8. Bars represent the mean ± SEM calculated across three independent experiments, with 5 to 10 mice per group in each experiment (combined sample sizes: 17 to 20 mice per treatment). * = p<0.05; ** = p<0.01; *** = p<0.001.
Figure 2. Colon histology after DSS-induced colitis.
Figure 2. Colon histology after DSS-induced colitis.
A. Descending colon of the negative control group (without bacteria or DSS). B. Descending colon of the positive group control (only DSS). C. Descending colon of the experimental group (DSS plus Lb CNRZ327). A, B, and C each show representative results from three independent experiments. D. Histological score of the groups described above. Bars represent the mean ± SEM calculated across three independent experiments, with 4 or 5 mice per group in each experiment (combined sample sizes: 13 to 14 mice per treatment). *** = p<0.001.
Figure 3. Secretory IgA levels after DSS-induced…
Figure 3. Secretory IgA levels after DSS-induced colitis.
Feces were collected and total sIgA was measured by ELISA. PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327. 100 (the average amount observed in the PBS control group) corresponds to the range of 61.5 to 126 pg/mL. Bars represent the mean ± SEM calculated across three independent experiments, with 3 to 10 mice per group in each experiment (combined sample sizes: 15 to 24 mice per treatment). ** = p

Figure 4. Cytokine production in colonic tissue…

Figure 4. Cytokine production in colonic tissue and spleen after DSS-induced colitis.

Cytokine concentrations in…

Figure 4. Cytokine production in colonic tissue and spleen after DSS-induced colitis.
Cytokine concentrations in homogenized tissues are presented as a percentage of the amount observed in the PBS control group (range from different experiments given in parentheses for each cytokine) which received the value of 100. Upper panel, colon: TGF-β (100 = 69 to 131.4 pg/mL), IL-4 (100 = 70.8 to 161.2 pg/mL), IL-6 (100 = 63.2 to 159.2 pg/mL), IL-10 (100 = 72.8 to 146.2 pg/mL), IL-12 (100 = 71.4 to 168.8 pg/mL), IL-17 (100 = 83.3 to 143.4 pg/mL). Lower panel, spleen: TGF-β (100 = 80.7 to 169.7 pg/mL), IL-6 (100 = 80.1 to 134 pg/mL), IL-10 (100 = 73.1 to 143.2 pg/mL), IL-12 (100 = 53.1 to 123 pg/mL), IL-17 (100 = 68.3 to 136.2 pg/mL). PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327.Bars represent the mean ± SEM calculated across two to six independent experiments, with 2 to 10 mice per group in each experiment (combined sample sizes: 9 to 33 mice per treatment (colon) and 5 to 12 mice per treatment (spleen)). * = p

Figure 5. T lymphocyte profiles in cecal…

Figure 5. T lymphocyte profiles in cecal lymph nodes and spleen after DSS-induced colitis.

T…

Figure 5. T lymphocyte profiles in cecal lymph nodes and spleen after DSS-induced colitis.
T cells were isolated from cecal lymph nodes (CLN, upper panel) and spleen (lower panel) at the end of the experiment, and the frequencies of CD4+CD69+ and CD4+Foxp3+ T cells, as percentage of CD4+ T cells, were assessed by flow cytometry. T-cell frequencies are presented relative to the average frequency observed in the PBS control group (range from different experiments given in parentheses for each T cell type) which received the value of 100. Upper panel, CLN: CD4+CD69+ (100 = 2.7 to 3.54), CD4+FoxP3+ (100 = 3.41 to 5.41). Lower panel, spleen: CD4+CD69+ (100 = 4.5 to 4.51), CD4+FoxP3+ (100 = 1.59 to 4.02). PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327. For CD4+CD69+ T cells, bars represent the mean ± SEM of one experiment, with 2 or 3 mice in the PBS control groups and 5 mice in each of the other groups. For CD4+FoxP3+ T cells, bars represent the mean ± SEM calculated across two independent experiments, with 3 to 5 mice per group in each experiment (combined sample sizes: 8 to 10 mice per treatment (colon) and 8 mice per treatment (spleen)). ** = p<0.01; *** = p<0.001; • = p = 0.053.

Figure 6. Correlation between relative frequencies of…

Figure 6. Correlation between relative frequencies of CD4+FoxP3 T cells in CLN and spleen.

Each…

Figure 6. Correlation between relative frequencies of CD4+FoxP3 T cells in CLN and spleen.
Each point represents one mouse, placed as a function of relative CD4+FoxP3 T cell frequencies in CLN and spleen (cf legend to Fig. 5). Black dots, PBS (control without bacteria or DSS); grey triangles, DSS (DSS only, without bacteria); light grey squares, 327+DSS (DSS plus Lb CNRZ327). The grey line describes the linear relationship between the frequencies of CD4+Foxp3+ cells in the spleen and in the colon (R2 = 0.415).
Similar articles
Cited by
References
    1. Neish AS (2009) Microbes in gastrointestinal health and disease. Gastroenterology 136: 65–80. - PMC - PubMed
    1. Hooper LV, Macpherson AJ (2010) Immune adaptations that maintain homeostasis with the intestinal microbiota. Nature reviews Immunology 10: 159–169. - PubMed
    1. Kamada N, Seo SU, Chen GY, Nunez G (2013) Role of the gut microbiota in immunity and inflammatory disease. Nature reviews Immunology 13: 321–335. - PubMed
    1. Faria AM, Weiner HL (2005) Oral tolerance. Immunological reviews 206: 232–259. - PMC - PubMed
    1. Strober W, Fuss I, Mannon P (2007) The fundamental basis of inflammatory bowel disease. J Clin Invest 117: 514–521. - PMC - PubMed
Show all 47 references
Publication types
MeSH terms
Related information
Grant support
This study received financial support from Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brasil (CNPq), Fundação de Amparo a Pesquisa do Estado de Minas Gerais, Brasil (FAPEMIG), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, Brasil (CAPES), and the EC EST project “LABHEALTH” (Marie Curie Contract MEST-2-CT-2004-514428). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Cytokine production in colonic tissue…
Figure 4. Cytokine production in colonic tissue and spleen after DSS-induced colitis.
Cytokine concentrations in homogenized tissues are presented as a percentage of the amount observed in the PBS control group (range from different experiments given in parentheses for each cytokine) which received the value of 100. Upper panel, colon: TGF-β (100 = 69 to 131.4 pg/mL), IL-4 (100 = 70.8 to 161.2 pg/mL), IL-6 (100 = 63.2 to 159.2 pg/mL), IL-10 (100 = 72.8 to 146.2 pg/mL), IL-12 (100 = 71.4 to 168.8 pg/mL), IL-17 (100 = 83.3 to 143.4 pg/mL). Lower panel, spleen: TGF-β (100 = 80.7 to 169.7 pg/mL), IL-6 (100 = 80.1 to 134 pg/mL), IL-10 (100 = 73.1 to 143.2 pg/mL), IL-12 (100 = 53.1 to 123 pg/mL), IL-17 (100 = 68.3 to 136.2 pg/mL). PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327.Bars represent the mean ± SEM calculated across two to six independent experiments, with 2 to 10 mice per group in each experiment (combined sample sizes: 9 to 33 mice per treatment (colon) and 5 to 12 mice per treatment (spleen)). * = p

Figure 5. T lymphocyte profiles in cecal…

Figure 5. T lymphocyte profiles in cecal lymph nodes and spleen after DSS-induced colitis.

T…

Figure 5. T lymphocyte profiles in cecal lymph nodes and spleen after DSS-induced colitis.
T cells were isolated from cecal lymph nodes (CLN, upper panel) and spleen (lower panel) at the end of the experiment, and the frequencies of CD4+CD69+ and CD4+Foxp3+ T cells, as percentage of CD4+ T cells, were assessed by flow cytometry. T-cell frequencies are presented relative to the average frequency observed in the PBS control group (range from different experiments given in parentheses for each T cell type) which received the value of 100. Upper panel, CLN: CD4+CD69+ (100 = 2.7 to 3.54), CD4+FoxP3+ (100 = 3.41 to 5.41). Lower panel, spleen: CD4+CD69+ (100 = 4.5 to 4.51), CD4+FoxP3+ (100 = 1.59 to 4.02). PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327. For CD4+CD69+ T cells, bars represent the mean ± SEM of one experiment, with 2 or 3 mice in the PBS control groups and 5 mice in each of the other groups. For CD4+FoxP3+ T cells, bars represent the mean ± SEM calculated across two independent experiments, with 3 to 5 mice per group in each experiment (combined sample sizes: 8 to 10 mice per treatment (colon) and 8 mice per treatment (spleen)). ** = p<0.01; *** = p<0.001; • = p = 0.053.

Figure 6. Correlation between relative frequencies of…

Figure 6. Correlation between relative frequencies of CD4+FoxP3 T cells in CLN and spleen.

Each…

Figure 6. Correlation between relative frequencies of CD4+FoxP3 T cells in CLN and spleen.
Each point represents one mouse, placed as a function of relative CD4+FoxP3 T cell frequencies in CLN and spleen (cf legend to Fig. 5). Black dots, PBS (control without bacteria or DSS); grey triangles, DSS (DSS only, without bacteria); light grey squares, 327+DSS (DSS plus Lb CNRZ327). The grey line describes the linear relationship between the frequencies of CD4+Foxp3+ cells in the spleen and in the colon (R2 = 0.415).
Figure 5. T lymphocyte profiles in cecal…
Figure 5. T lymphocyte profiles in cecal lymph nodes and spleen after DSS-induced colitis.
T cells were isolated from cecal lymph nodes (CLN, upper panel) and spleen (lower panel) at the end of the experiment, and the frequencies of CD4+CD69+ and CD4+Foxp3+ T cells, as percentage of CD4+ T cells, were assessed by flow cytometry. T-cell frequencies are presented relative to the average frequency observed in the PBS control group (range from different experiments given in parentheses for each T cell type) which received the value of 100. Upper panel, CLN: CD4+CD69+ (100 = 2.7 to 3.54), CD4+FoxP3+ (100 = 3.41 to 5.41). Lower panel, spleen: CD4+CD69+ (100 = 4.5 to 4.51), CD4+FoxP3+ (100 = 1.59 to 4.02). PBS, control without bacteria or DSS; DSS, DSS only, without bacteria; 327+DSS, DSS plus Lb CNRZ327. For CD4+CD69+ T cells, bars represent the mean ± SEM of one experiment, with 2 or 3 mice in the PBS control groups and 5 mice in each of the other groups. For CD4+FoxP3+ T cells, bars represent the mean ± SEM calculated across two independent experiments, with 3 to 5 mice per group in each experiment (combined sample sizes: 8 to 10 mice per treatment (colon) and 8 mice per treatment (spleen)). ** = p<0.01; *** = p<0.001; • = p = 0.053.
Figure 6. Correlation between relative frequencies of…
Figure 6. Correlation between relative frequencies of CD4+FoxP3 T cells in CLN and spleen.
Each point represents one mouse, placed as a function of relative CD4+FoxP3 T cell frequencies in CLN and spleen (cf legend to Fig. 5). Black dots, PBS (control without bacteria or DSS); grey triangles, DSS (DSS only, without bacteria); light grey squares, 327+DSS (DSS plus Lb CNRZ327). The grey line describes the linear relationship between the frequencies of CD4+Foxp3+ cells in the spleen and in the colon (R2 = 0.415).

References

    1. Neish AS (2009) Microbes in gastrointestinal health and disease. Gastroenterology 136: 65–80.
    1. Hooper LV, Macpherson AJ (2010) Immune adaptations that maintain homeostasis with the intestinal microbiota. Nature reviews Immunology 10: 159–169.
    1. Kamada N, Seo SU, Chen GY, Nunez G (2013) Role of the gut microbiota in immunity and inflammatory disease. Nature reviews Immunology 13: 321–335.
    1. Faria AM, Weiner HL (2005) Oral tolerance. Immunological reviews 206: 232–259.
    1. Strober W, Fuss I, Mannon P (2007) The fundamental basis of inflammatory bowel disease. J Clin Invest 117: 514–521.
    1. Bouma G, Strober W (2003) The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol 3: 521–533.
    1. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, et al. (2008) Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 105: 16731–16736.
    1. Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, et al. (2009) Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis 15: 1183–1189.
    1. Mileti E, Matteoli G, Iliev ID, Rescigno M (2009) Comparison of the immunomodulatory properties of three probiotic strains of Lactobacilli using complex culture systems: prediction for in vivo efficacy. PloS one 4: e7056.
    1. Meijer BJ, Dieleman LA (2011) Probiotics in the treatment of human inflammatory bowel diseases: update 2011. Journal of clinical gastroenterology 45 Suppl: S139–144.
    1. Ohland CL, Macnaughton WK (2010) Probiotic bacteria and intestinal epithelial barrier function. American journal of physiology Gastrointestinal and liver physiology 298: G807–819.
    1. Lievin V, Peiffer I, Hudault S, Rochat F, Brassart D, et al. (2000) Bifidobacterium strains from resident infant human gastrointestinal microflora exert antimicrobial activity. Gut 47: 646–652.
    1. de Moreno de Leblanc A, Del Carmen S, Zurita-Turk M, Santos Rocha C, van de Guchte M, et al. (2011) Importance of IL-10 modulation by probiotic microorganisms in gastrointestinal inflammatory diseases. ISRN gastroenterology 2011: 892971.
    1. Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M (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.
    1. Round JL, Mazmanian SK (2010) Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 107: 12204–12209.
    1. Santos Rocha C, Lakhdari O, Blottiere HM, Blugeon S, Sokol H, et al. (2012) Anti-inflammatory properties of dairy lactobacilli. Inflamm Bowel Dis 18: 657–666.
    1. Jobin C, Sartor RB (2000) The I kappa B/NF-kappa B system: a key determinant of mucosalinflammation and protection. American journal of physiology Cell physiology 278: C451–462.
    1. Murthy SN, Cooper HS, Shim H, Shah RS, Ibrahim SA, et al. (1993) Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin. Dig Dis Sci 38: 1722–1734.
    1. McCafferty DM, Sihota E, Muscara M, Wallace JL, Sharkey KA, et al. (2000) Spontaneously developing chronic colitis in IL-10/iNOS double-deficient mice. Am J Physiol Gastrointest Liver Physiol 279: G90–99.
    1. Maron R, Hancock WW, Slavin A, Hattori M, Kuchroo V, et al. (1999) Genetic susceptibility or resistance to autoimmune encephalomyelitis in MHC congenic mice is associated with differential production of pro- and anti-inflammatory cytokines. Int Immunol 11: 1573–1580.
    1. Pinheiro J, Bates D (2000) Mixed-effects models in S and S-PLUS. Berlin: Springer.
    1. Solomon L, Mansor S, Mallon P, Donnelly E, Hoper M, et al. (2010) The dextran sulphate sodium (DSS) model of colitis: an overview. Comp Clin Pathol 19: 235–239.
    1. O'Sullivan DJ (2001) Screening of intestinal microflora for effective probiotic bacteria. J Agric Food Chem 49: 1751–1760.
    1. Malin M, Suomalainen H, Saxelin M, Isolauri E (1996) Promotion of IgA immune response in patients with Crohn's disease by oral bacteriotherapy with Lactobacillus GG. Ann Nutr Metab 40: 137–145.
    1. Hamilton MJ, Sinnamon MJ, Lyng GD, Glickman JN, Wang X, et al. (2011) Essential role for mast cell tryptase in acute experimental colitis. Proc Natl Acad Sci U S A 108: 290–295.
    1. Melgar S, Karlsson A, Michaelsson E (2005) Acute colitis induced by dextran sulfate sodium progresses to chronicity in C57BL/6 but not in BALB/c mice: correlation between symptoms and inflammation. Am J Physiol Gastrointest Liver Physiol 288: G1328–1338.
    1. Thomas CM, Versalovic J (2010) Probiotics-host communication: Modulation of signaling pathways in the intestine. Gut microbes 1: 148–163.
    1. Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, et al. (1990) A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 98: 694–702.
    1. Laroui H, Ingersoll SA, Liu HC, Baker MT, Ayyadurai S, et al. (2012) Dextran sodium sulfate (DSS) induces colitis in mice by forming nano-lipocomplexes with medium-chain-length fatty acids in the colon. PloS one 7: e32084.
    1. Letterio JJ, Roberts AB (1998) Regulation of immune responses by TGF-beta. Annu Rev Immunol 16: 137–161.
    1. Fichtner-Feigl S, Fuss IJ, Young CA, Watanabe T, Geissler EK, et al. (2007) Induction of IL-13 triggers TGF-beta1-dependent tissue fibrosis in chronic 2,4,6-trinitrobenzene sulfonic acid colitis. J Immunol 178: 5859–5870.
    1. Pallone F, Monteleone G (1998) Interleukin 12 and Th1 responses in inflammatory bowel disease. Gut 43: 735–736.
    1. Brand S (2009) Crohn's disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn's disease. Gut 58: 1152–1167.
    1. Swain SL, Weinberg AD, English M, Huston G (1990) IL-4 directs the development of Th2-like helper effectors. Journal of immunology 145: 3796–3806.
    1. Mannon PJ, Fuss IJ, Mayer L, Elson CO, Sandborn WJ, et al. (2004) Anti-interleukin-12 antibody for active Crohn's disease. The New England journal of medicine 351: 2069–2079.
    1. Chung YW, Choi JH, Oh TY, Eun CS, Han DS (2008) Lactobacillus casei prevents the development of dextran sulphate sodium-induced colitis in Toll-like receptor 4 mutant mice. Clin Exp Immunol 151: 182–189.
    1. Philippe D, Heupel E, Blum-Sperisen S, Riedel CU (2011) Treatment with Bifidobacterium bifidum 17 partially protects mice from Th1-driven inflammation in a chemically induced model of colitis. International journal of food microbiology 149: 45–49.
    1. Kim SW, Kim HM, Yang KM, Kim SA, Kim SK, et al. (2010) Bifidobacterium lactis inhibits NF-kappaB in intestinal epithelial cells and prevents acute colitis and colitis-associated colon cancer in mice. Inflamm Bowel Dis 16: 1514–1525.
    1. Nishitani Y, Tanoue T, Yamada K, Ishida T, Yoshida M, et al. (2009) Lactococcus lactis subsp. cremoris FC alleviates symptoms of colitis induced by dextran sulfate sodium in mice. International immunopharmacology 9: 1444–1451.
    1. Diaz-Ropero MP, Martin R, Sierra S, Lara-Villoslada F, Rodriguez JM, et al. (2007) Two Lactobacillus strains, isolated from breast milk, differently modulate the immune response. Journal of applied microbiology 102: 337–343.
    1. Lavasani S, Dzhambazov B, Nouri M, Fak F, Buske S, et al. (2010) A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PloS one 5: e9009.
    1. Curotto de Lafaille MA, Lafaille JJ (2009) Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 30: 626–635.
    1. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330–336.
    1. Makita S, Kanai T, Oshima S, Uraushihara K, Totsuka T, et al. (2004) CD4+CD25bright T cells in human intestinal lamina propria as regulatory cells. J Immunol 173: 3119–3130.
    1. Maul J, Loddenkemper C, Mundt P, Berg E, Giese T, et al. (2005) Peripheral and intestinal regulatory CD4+ CD25(high) T cells in inflammatory bowel disease. Gastroenterology 128: 1868–1878.
    1. Macho Fernandez E, Valenti V, Rockel C, Hermann C, Pot B, et al. (2011) Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut 60: 1050–1059.
    1. Smelt MJ, de Haan BJ, Bron PA, van Swam I, Meijerink M, et al. (2012) L. plantarum, L. salivarius, and L. lactis attenuate Th2 responses and increase Treg frequencies in healthy mice in a strain dependent manner. PLoS One 7: e47244.

Source: PubMed

3
Sottoscrivi