C. Moser, H. K. Johansen, Z. Song, H. P. Hougen, J. Rygaard et al., Chronic Pseudomonas aeruginosa lung infection is more severe in Th2 responding BALB/c mice compared to Th1 responding C3H/ HeN mice, APMIS, vol.105, pp.838-880, 1997.

C. Moser, H. P. Hougen, Z. Song, J. Rygaard, A. Kharazmi et al., Early immune response in susceptible and resistant mice strains with chronic Pseudomonas aeruginosa lung infection determines the type of T-helper cell response, APMIS, vol.107, pp.1093-100, 1999.

C. Moser, S. Kjaergaard, T. Pressler, A. Kharazmi, C. Koch et al., The immune response to chronic Pseudomonas aeruginosa lung infection in cystic fibrosis patients is predominantly of the Th2 type, APMIS, vol.108, pp.329-364, 2000.

C. Moser, P. O. Jensen, and T. Pressler, Serum concentrations of GM-CSF and G-CSF correlate with the Th1/Th2 cytokine response in cystic fibrosis patients with chronic Pseudomonas aeruginosa lung infection, APMIS, vol.113, pp.400-409, 2005.

D. Hartl, M. Griese, and M. Kappler, Pulmonary T(H)2 response in Pseudomonas aeruginosa-infected patients with cystic fibrosis, J Allergy Clin Immunol, vol.117, pp.204-215, 2006.

B. S. Murphy, H. M. Bush, and V. Sundareshan, Characterization of macrophage activation states in patients with cystic fibrosis, J Cyst Fibrosis, vol.9, pp.314-336, 2010.

G. T. Macfarlane, H. Steed, and S. Macfarlane, Bacterial metabolism and health-related effects of galacto-oligosaccharides and other prebiotics, J Appl Microbiol, vol.104, pp.305-349, 2008.

M. Roberfroid, G. R. Gibson, and L. Hoyles, Prebiotic effects: metabolic and health benefits, Br J Nutr, vol.104, issue.2, pp.1-63, 2010.

C. Y. Li, H. C. Lin, C. H. Lai, J. J. Lu, S. F. Wu et al., Immunomodulatory effects of lactobacillus and Bifidobacterium on both murine and human mitogen-activated T cells, Int Arch Allergy Immunol, vol.156, pp.128-164, 2011.

H. M. Zhao, X. Y. Huang, and Z. Q. Zuo, Probiotics increase T regulatory cells and reduce severity of experimental colitis in mice, World J Gastroenterol, vol.19, pp.742-751, 2013.

K. Meijer, P. De-vos, and M. G. Priebe, Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health?, Curr Opin Clin Nutr Metab Care, vol.13, pp.715-736, 2010.

P. M. Smith, M. R. Howitt, and N. Panikov, The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science, vol.341, pp.569-73, 2013.

P. G. Reeves, F. H. Nielsen, and G. C. Fahey, AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet, J Nutr, vol.123, pp.1939-51, 1993.

L. Geurts, V. Lazarevic, and M. Derrien, Altered gut microbiota and endocannabinoid system tone in obese and diabetic leptin-resistant mice: impact on apelin regulation in adipose tissue, Front Microbiol, vol.2, p.149, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00617647

M. Rajilic-stojanovic, H. G. Heilig, and D. Molenaar, Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults, Environ Microbiol, vol.11, pp.1736-51, 2009.

J. Jalanka-tuovinen, A. Salonen, and J. Nikkila, Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms, PLoS One, vol.6, p.23035, 2011.

L. Lahti, L. L. Elo, T. Aittokallio, and S. Kaski, Probabilistic analysis of probe reliability in differential gene expression studies with short oligonucleotide arrays, IEEE/ACM Trans Comput Biol Bioinform, vol.8, pp.217-242, 2011.

M. Pierre, M. O. Husson, and B. R. Le, Omega-3 polyunsaturated fatty acids improve host response in chronic Pseudomonas aeruginosa lung infection in mice, Am J Physiol Lung Cell Mol Physiol, vol.292, pp.1422-1453, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00166087

G. Telford, D. Wheeler, and P. Williams, The Pseudomonas aeruginosa quorum-sensing signal molecule N-(3-oxododecanoyl)-L-homoserine lactone has immunomodulatory activity, Infect Immun, vol.66, pp.36-42, 1998.

A. J. Ritchie, A. Jansson, J. Stallberg, P. Nilsson, P. Lysaght et al., The Pseudomonas aeruginosa quorum-sensing molecule N-3-(oxododecanoyl)-L-homoserine lactone inhibits T-cell differentiation and cytokine production by a mechanism involving an early step in T-cell activation, Infect Immun, vol.73, pp.1648-55, 2005.

D. Ghadimi, R. Fölster-holst, M. De-vrese, P. Winkler, K. J. Heller et al., Effects of probiotic bacteria and their genomic DNA on TH1/TH2-cytokine production by peripheral blood mononuclear cells (PBMCs) of healthy and allergic subjects, Immunobiology, vol.213, pp.677-92, 2008.

B. Schouten, B. C. Van-esch, and G. A. Hofman, Cow milk allergy symptoms are reduced in mice fed dietary synbiotics during oral sensitization with whey, J Nutr, vol.139, pp.1398-403, 2009.

A. Rueda, M. Manas, A. Valverde, J. I. Fernandez, J. A. Naranjo et al., Conjugated bile acids and intestinal flora during the preruminant stage in goat. Influence of a lamb milk replacer, Arch Physiol Biochem, vol.104, pp.246-251, 1996.

S. Devkota, Y. Wang, and M. W. Musch, Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/-mice, Nature, vol.487, pp.104-112, 2012.

K. Atarashi, T. Tanoue, and K. Oshima, Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota, Nature, vol.500, pp.232-238, 2013.

H. W. Wollenweber, E. T. Rietschel, T. Hofstad, A. Weintraub, and A. A. Lindberg, Nature, type of linkage, quantity, and absolute configuration of (3-hydroxy) fatty acids in lipopolysaccharides from Bacteroides fragilis NCTC 9343 and related strains, J Bacteriol, vol.144, pp.898-903, 1980.

B. Ruiz-perez, D. R. Chung, and A. H. Sharpe, Modulation of surgical fibrosis by microbial zwitterionic polysaccharides, Proc Natl Acad Sci U S A, vol.102, pp.16753-16761, 2005.

J. L. Round and S. K. Mazmanian, Inducible Foxp31 regulatory T-cell development by a commensal bacterium of the intestinal microbiota, Proc Natl Acad Sci U S A, vol.107, pp.12204-12213, 2010.

T. Sato, K. Matsumoto, and T. Okumura, Isolation of lactate-utilizing butyrate-producing bacteria from human feces and in vivo administration of Anaerostipes caccae strain L2 and galacto-oligosaccharides in a rat model, FEMS Microbiol Ecol, vol.66, pp.528-564, 2008.

K. P. Scott, J. C. Martin, S. H. Duncan, and H. J. Flint, Prebiotic stimulation of human colonic butyrate-producing bacteria and bifidobacteria, in vitro, FEMS Microbiol Ecol, vol.86, pp.1-11, 2013.

J. Yang, I. Martínez, J. Walter, A. Keshavarzian, and D. J. Rose, In vitro characterization of the impact of selected dietary fibers on fecal microbiota composition and short chain fatty acid production, Anaerobe, vol.23, pp.74-81, 2013.

D. R. Donohoe, N. Garge, and X. Zhang, The microbiome and butyrate regulate energy metabolism and autophagy in the mamamlian colon, Cell Metab, vol.13, pp.517-543, 2011.

L. N. Nguyen, L. C. Lopes, R. J. Cordero, and J. D. Nosanchuk, Sodium butyrate inhibits pathogenic yeast growth and enhances the functions of macrophages, J Antimicrob Chemother, vol.66, pp.2573-80, 2011.

T. Eiwegger, B. Stahl, and J. Schmitt, Human milk-derived oligosaccharides and plant-derived oligosaccharides stimulate cytokine production of cord blood T-cells in vitro, Pediatr Res, vol.56, pp.536-576, 2004.

T. Eiwegger, B. Stahl, and P. Haidl, Prebiotic oligosaccharides: in vitro evidence for gastrointestinal epithelial transfer and immunomodulatory properties, Pediatr Allergy Immunol, vol.21, pp.1179-88, 2010.

S. Obermeier, S. Rudloff, G. Pohlentz, M. J. Lentze, and C. Kunz, Secretion of 13C-labelled oligosaccharides into human milk and infant's urine after an oral [13C]galactose load, Isotopes Environ Health Stud, vol.35, pp.119-144, 1999.

S. De-kivit, A. D. Kraneveld, J. Garssen, and L. E. Willemsen, Glycan recognition at the interface of the intestinal immune system: target for immune modulation via dietary components, Eur J Pharmacol, vol.668, issue.1, pp.124-156, 2011.

B. Van't-land, M. Schijf, and B. C. Van-esch, Regulatory T-cells have a1 prominent role in the immune modulated vaccine response by specific oligosaccharides, Vaccine, vol.28, pp.5711-5718, 2010.

B. Schouten, B. C. Van-esch, and G. A. Hofman, Oligosaccharide-induced whey-specific CD25(+) regulatory T-cells are involved in the suppression of cow milk allergy in mice, J Nutr, vol.140, pp.835-876, 2010.

A. P. Vos, B. C. Van-esch, and B. Stahl, Dietary supplementation with specific oligosaccharide mixtures decreases parameters of allergic asthma in mice, Int Immunopharmacol, vol.7, pp.1582-1589, 2007.

E. Van-hoffen, B. Ruiter, and J. Faber, A specific mixture of short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides induces a beneficial immunoglobulin profile in infants at high risk for allergy, Allergy, vol.64, pp.484-491, 2009.

M. A. Schijf, D. Kruijsen, and J. Bastiaans, specific dietary oligosaccharides increase Th1 responses in a mouse respiratory syncytial virus infection model, J Virol, vol.86, pp.11472-82, 2012.

N. Maeda, R. Nakamura, and Y. Hirose, Oral administration of heatkilled Lactobacillus plantarum L-137 enhances protection against influenza virus infection by stimulation of type I interferon production in mice, Int Immunopharmacol, vol.9, pp.1122-1127, 2009.

M. Kawase, F. He, A. Kubota, K. Yoda, K. Miyazawa et al., Heatkilled Lactobacillus gasseri TMC0356 protects mice against influenza virus infection by stimulating gut and respiratory immune responses, FEMS Immunol Med Microbiol, vol.64, pp.280-288, 2012.

A. Tanaka, M. Seki, and S. Yamahira, Lactobacillus pentosus strain b240 suppresses pneumonia induced by Streptococcus pneumoniae in mice, Lett Appl Microbiol, vol.53, pp.35-43, 2011.

E. O. Vintini and M. S. Medina, Host immunity in the protective response to nasal immunization with a pneumococcal antigen associated to live and heat-killed Lactobacillus casei, BMC Immunol, vol.12, pp.46-60, 2011.

, pAOS Improve the Outcome of P. aeruginosa Infection, vol.2015, p.165