H. Rabah, R. Do-carmo, F. L. , and J. G. , Dairy Propionibacteria: versatile, Probiotics. Microorganisms, vol.5, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01524323

F. J. Cousin, D. D. Mater, B. Foligne, and J. G. , Dairy propionibacteria as human probiotics: A review of recent evidence, Dairy Sci Technol, vol.91, pp.1-26, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00868601

G. Mogensen, S. Salminen, J. O'brien, A. Ouwenhand, W. Holzapfel et al., Inventory of microoganisms with a documented history of use in food, pp.10-19, 2002.

F. J. Cousin, B. Foligné, S. M. Deutsch, S. Massart, S. Parayre et al., Assessment of the probiotic potential of a dairy product fermented by Propionibacterium freudenreichii in piglets, J Agric Food Chem, vol.60, pp.7917-7944, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01209320

B. Foligné, S. M. Deutsch, J. Breton, F. J. Cousin, J. Dewulf et al., Promising immunomodulatory effects of selected strains of dairy propionibacteria as evidenced in vitro and in vivo, Appl Environ Microbiol, vol.76, pp.8259-64, 2010.

A. Oksaharju, T. Kooistra, R. Kleemann, W. Van-duyvenvoorde, M. Miettinen et al., Effects of probiotic Lactobacillus rhamnosus GG and Propionibacterium freudenreichii ssp. shermanii JS supplementation on intestinal and systemic markers of inflammation in ApoE*3Leiden mice consuming a high-fat diet, Br J Nutr, vol.110, pp.77-85, 2013.

K. Kajander, E. Myllyluoma, M. Rajili?-stojanovi?, S. Kyrönpalo, M. Rasmussen et al., Clinical trial: multispecies probiotic supplementation alleviates the symptoms of irritable bowel syndrome and stabilizes intestinal microbiota, Aliment Pharmacol Ther, vol.27, pp.48-57, 2008.

C. Plé, R. Richoux, J. Jardin, M. Nurdin, V. Briard-bion et al., Single-strain starter experimental cheese reveals anti-inflammatory effect of Propionibacterium freudenreichii CIRM BIA 129 in TNBScolitis model, J Funct Foods, vol.18, pp.575-85, 2015.

C. Plé, J. Breton, R. Richoux, M. Nurdin, S. M. Deutsch et al., Combining selected immunomodulatory Propionibacterium freudenreichii and Lactobacillus delbrueckii strains: reverse engineering development of an anti-inflammatory cheese, Mol Nutr Food Res, vol.60, pp.935-983, 2016.

L. Maréchal, C. Peton, V. Plé, C. Vroland, C. Jardin et al., Surface proteins of Propionibacterium freudenreichii are involved in its antiinflammatory properties, J Proteomics, vol.113, pp.447-61, 2015.

F. L. Do-carmo, H. Rabah, S. Huang, F. Gaucher, M. Deplanche et al., Propionibacterium freudenreichii Surface Protein SlpB Is Involved in Adhesion to Intestinal HT-29 Cells, Front Microbiol, vol.8, p.1033, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01824996

F. L. Do-carmo, W. M. Silva, G. C. Tavares, I. C. Ibraim, B. F. Cordeiro et al., Mutation of the surface layer protein SlpB has pleiotropic effects in the probiotic Propionibacterium freudenreichii CIRM-BIA 129, Front Microbiol, vol.9, p.1807, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01857987

S. M. Deutsch, M. Mariadassou, P. Nicolas, S. Parayre, L. Guellec et al., Identification of proteins involved in the anti-inflammatory properties of Propionibacterium freudenreichii by means of a multi-strain study, Sci Rep, vol.7, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01510019

F. Do-carmo, H. Rabah, F. Cordeiro, B. , D. Silva et al., Applications of Probiotic Bacteria and Dairy Foods in Health. Current Research in Microbiology. Open Access eBooks 919 North Market Street Suite 425 Wilmington, DE, pp.1-33, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01583804

S. T. Sonis, The pathobiology of mucositis, Nat Rev Cancer, vol.4, pp.277-84, 2004.

M. M. Antunes, P. C. Leocádio, L. G. Teixeira, A. J. Leonel, D. C. Cara et al., Pretreatment With L-Citrulline Positively Affects the Mucosal Architecture and Permeability of the Small Intestine in a Murine Mucositis Model, JPEN J Parenter Enteral Nutr, vol.40, pp.279-86, 2016.

C. T. Chang, T. Y. Ho, H. Lin, J. A. Liang, H. C. Huang et al., 5-Fluorouracil induced intestinal mucositis via nuclear factor-?B activation by transcriptomic analysis and in vivo bioluminescence imaging, PLoS One, vol.7, 2012.

A. M. Stringer, Interaction between host cells and microbes in chemotherapy-induced mucositis, Nutrients, vol.5, pp.1488-99, 2013.

R. D. Carvalho, F. L. Do-carmo, A. De-oliveira-junior, P. Langella, J. M. Chatel et al.,

M. S. Azevedo, Use of wild type or recombinant Lactic Acid Bacteria as an alternative treatment for gastrointestinal inflammatory diseases: A focus on Inflammatory Bowel Diseases and Mucositis, Front Microbiol, vol.8, p.800, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01602781

E. Cereda, M. Caraccia, and R. Caccialanza, Probiotics and mucositis, Curr Opin Clin Nutr Metab Care, vol.21, pp.399-404, 2018.

J. M. Bowen, R. J. Gibson, J. K. Coller, N. Blijlevens, P. Bossi et al., and Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society of Oral Oncology (MASCC/ISOO). Systematic review of agents for the management of cancer treatment-related gastrointestinal mucositis and clinical practice guidelines, Support Care Cancer, vol.27, pp.4011-4033, 2019.

R. V. Lalla, J. Bowen, A. Barasch, L. Elting, J. Epstein et al., Guidelines Leadership Group of the Multinational Association of Supportive Care in Cancer and International Society of Oral Oncology (MASCC/ISOO). MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy, Cancer, vol.120, pp.1453-61, 2014.

F. L. Do-carmo, H. Rabah, D. Oliveira-carvalho, R. D. Gaucher, F. Cordeiro et al., Extractable Bacterial Surface Proteins in Probiotic-Host Interaction, Front Microbiol, vol.9, p.645, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01763214

N. Colliou, Y. Ge, B. Sahay, M. Gong, M. Zadeh et al., Commensal Propionibacterium strain UF1 mitigates intestinal inflammation via Th17 cell regulation, J Clin Invest, vol.127, pp.3970-86, 2017.

E. Gerbino, P. Carasi, P. Mobili, M. A. Serradell, and A. Gómez-zavaglia, Role of S-layer proteins in bacteria. World www.oncotarget, J Microbiol Biotechnol, vol.31, pp.1877-87, 2015.

U. B. Sleytr, B. Schuster, E. M. Egelseer, and D. Pum, S-layers: principles and applications, FEMS Microbiol Rev, vol.38, pp.823-64, 2014.

J. Preising, P. D. Gleinser, M. Wei, H. Blum, S. Eikmanns et al., Selection of bifidobacteria based on adhesion and anti-inflammatory capacity in vitro for amelioration of murine colitis, Appl Environ Microbiol, vol.76, pp.3048-51, 2010.

R. Wang, L. Jiang, M. Zhang, L. Zhao, Y. Hao et al., The Adhesion of Lactobacillus salivarius REN to a Human Intestinal Epithelial Cell Line Requires S-layer, Proteins. Sci Rep, vol.7, p.44029, 2017.

B. R. Johnson and T. R. Klaenhammer, AcmB Is an S-Layer-Associated ?-N-Acetylglucosaminidase and Functional Autolysin in Lactobacillus acidophilus NCFM, Appl Environ Microbiol, vol.82, pp.5687-97, 2016.

J. P. Hymes, B. R. Johnson, R. Barrangou, and T. R. Klaenhammer, Functional Analysis of an S-Layer-Associated Fibronectin-Binding Protein in Lactobacillus acidophilus NCFM

, Appl Environ Microbiol, vol.82, pp.2676-85, 2016.

H. Rabah, O. Ménard, F. Gaucher, F. L. Do-carmo, D. Dupont et al., Cheese matrix protects the immunomodulatory surface protein SlpB of Propionibacterium freudenreichii during in vitro digestion, Food Res Int, vol.106, pp.712-733, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01707548

T. L. Denning, N. A. Campbell, F. Song, R. P. Garofalo, G. R. Klimpel et al., Expression of IL-10 receptors on epithelial cells from the murine small and large intestine, Int Immunol, vol.12, pp.133-142, 2000.

A. Jarry, C. Bossard, C. Bou-hanna, D. Masson, E. Espaze et al., Mucosal IL-10 and TGF-beta play crucial roles in preventing LPS-driven, IFN-gammamediated epithelial damage in human colon explants, J Clin Invest, vol.118, pp.1132-1174, 2008.

T. Kucharzik, . Hudson, A. Lügering, J. A. Abbas, M. Bettini et al., Acute induction of human IL-8 production by intestinal epithelium triggers neutrophil infiltration without mucosal injury, Gut, vol.54, pp.1565-72, 2005.

M. Singer and P. J. Sansonetti, IL-8 is a key chemokine regulating neutrophil recruitment in a new mouse model of Shigellainduced colitis, J Immunol, vol.173, pp.4197-206, 2004.

A. P. Bai, Q. Ouyang, W. Zhang, C. H. Wang, and S. F. Li, Probiotics inhibit TNF-alpha-induced interleukin-8 secretion of HT29 cells, World J Gastroenterol, vol.10, pp.455-57, 2004.

R. K. Duary, V. K. Batish, and S. Grover, Immunomodulatory activity of two potential probiotic strains in LPS-stimulated HT-29 cells, Genes Nutr, vol.9, p.398, 2014.

S. Ménard, C. Candalh, J. C. Bambou, K. Terpend, N. Cerf-bensussan et al., Lactic acid bacteria secrete metabolites retaining anti-inflammatory properties after intestinal transport, Gut, vol.53, pp.821-849, 2004.

V. Taverniti, M. Stuknyte, M. Minuzzo, S. Arioli, D. Noni et al.,

, Appl Environ Microbiol, vol.79, pp.1221-1252, 2013.

P. Li, Q. Yu, X. Ye, Z. Wang, and Q. Yang, Lactobacillus S-layer protein inhibition of Salmonella-induced reorganization of the cytoskeleton and activation of MAPK signalling pathways in Caco-2 cells, Microbiology, vol.157, pp.2639-2685, 2011.

S. R. Konstantinov, H. Smidt, W. M. De-vos, S. C. Bruijns, S. K. Singh et al., S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions, Proc Natl Acad Sci U S A, vol.105, pp.19474-79, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01454074

Y. L. Lightfoot, K. Selle, T. Yang, Y. J. Goh, B. Sahay et al., SIGNR3-dependent immune regulation by Lactobacillus acidophilus surface layer protein A in colitis, EMBO J, vol.34, pp.881-95, 2015.

B. Johnson, K. Selle, S. O'flaherty, Y. J. Goh, and T. Klaenhammer, Identification of extracellular surface-layer associated proteins in Lactobacillus acidophilus, NCFM. Microbiology, vol.159, pp.2269-82, 2013.

B. R. Johnson, S. O'flaherty, Y. J. Goh, I. Carroll, R. Barrangou et al., The S-layer Associated Serine Protease Homolog PrtX Impacts Cell Surface-Mediated Microbe-Host Interactions of Lactobacillus acidophilus NCFM, Front Microbiol, vol.8, p.1185, 2017.

J. Rong, H. Zheng, M. Liu, X. Hu, T. Wang et al., Probiotic and anti-inflammatory attributes of an isolate Lactobacillus helveticus NS8 from Mongolian fermented koumiss, BMC Microbiol, vol.15, 0196.

K. Uroi?, J. Novak, U. Hynönen, T. E. Pietilä, L. Pavunc et al., The role of S-layer in adhesive and immunomodulating properties of probiotic starter culture Lactobacillus brevis D6 isolated from artisanal smoked fresh cheese, Lebenson Wiss Technol, vol.69, pp.623-655, 2016.

S. Mukherjee, S. Karmakar, and S. P. Babu, TLR2 and TLR4 mediated host immune responses in major infectious diseases: a review, Braz J Infect Dis, vol.20, pp.193-204, 2016.

T. H. Mogensen, Pathogen recognition and inflammatory signaling in innate immune defenses, Clin Microbiol Rev, vol.22, pp.240-73, 2009.

R. Paolillo, R. Carratelli, C. Sorrentino, S. Mazzola, N. Rizzo et al., Immunomodulatory effects of Lactobacillus plantarum on human colon cancer cells, Int Immunopharmacol, vol.9, pp.1265-71, 2009.

Y. C. Lu, W. C. Yeh, and P. S. Ohashi, LPS/TLR4 signal transduction pathway, Cytokine, vol.42, pp.145-51, 2008.

V. Pinto, M. G. , R. Gómez, M. Seifert, S. Watzl et al., Lactobacilli stimulate the innate immune response and modulate the TLR expression of HT29 intestinal epithelial cells in vitro, Int J Food Microbiol, vol.133, pp.86-93, 2009.

M. Cammarota, D. Rosa, M. Stellavato, A. Lamberti, M. Marzaioli et al., In vitro evaluation of Lactobacillus plantarum DSMZ 12028 as a probiotic: emphasis on innate immunity, Int J Food Microbiol, vol.135, pp.90-98, 2009.

K. Gao, C. Wang, L. Liu, X. Dou, J. Liu et al., Immunomodulation and signaling mechanism of Lactobacillus rhamnosus GG and its components on porcine intestinal epithelial cells stimulated by lipopolysaccharide, J Microbiol Immunol Infect, vol.50, pp.700-713, 2017.

J. M. Bowen, A. M. Stringer, R. J. Gibson, A. S. Yeoh, S. Hannam et al., VSL#3 probiotic treatment reduces chemotherapy-induced diarrhea and weight loss, Cancer Biol Ther, vol.6, pp.1449-54, 2007.

S. Kato, N. Hamouda, Y. Kano, Y. Oikawa, Y. Tanaka et al., Probiotic Bifidobacterium bifidum G9-1 attenuates 5-fluorouracilinduced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses, Clin Exp Pharmacol Physiol, vol.44, pp.1017-1042, 2017.

L. M. Trindade, V. D. Martins, N. M. Rodrigues, E. L. Souza, F. S. Martins et al., Oral administration of Simbioflora® (synbiotic) attenuates intestinal damage in a mouse model of 5-fluorouracil-induced mucositis, Benef Microbes, vol.9, pp.477-86, 2018.

R. D. Carvalho, N. Breyner, Z. Menezes-garcia, N. M. Rodrigues, L. Lemos et al., Secretion of biologically active pancreatitis-associated protein I (PAP) by genetically modified dairy Lactococcus lactis NZ9000 in the prevention of intestinal mucositis, Microb Cell Fact, vol.16, p.27, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607391

N. Y. Lycke and M. Bemark, The regulation of gut mucosal IgA B-cell responses: recent developments, Mucosal Immunol, vol.10, pp.1361-74, 2017.

D. L. Schmucker, R. L. Owen, R. Outenreath, and K. Thoreux, Basis for the age-related decline in intestinal mucosal immunity, Clin Dev Immunol, vol.10, pp.167-72, 2003.

S. Sakaguchi, K. Wing, Y. Onishi, P. Prieto-martin, and T. Yamaguchi, Regulatory T cells: how do they suppress immune responses?, Int Immunol, vol.21, pp.1105-1116, 2009.

J. D. Fontenot, M. A. Gavin, and A. Y. Rudensky, Foxp3 programs the development and function of CD4+CD25+ regulatory T cells, Nat Immunol, vol.4, pp.330-366, 2003.

Y. Wei, C. Lu, J. Chen, G. Cui, L. Wang et al., High salt diet stimulates gut Th17 response and exacerbates TNBSinduced colitis in mice, Oncotarget, vol.8, pp.70-82, 2017.

I. Monteleone, I. Marafini, V. Dinallo, D. Fusco, D. Troncone et al., Sodium chlorideenriched Diet Enhanced Inflammatory Cytokine Production and Exacerbated Experimental Colitis in Mice, J Crohns Colitis, vol.11, pp.237-282, 2017.

N. Powell, M. M. Walker, and N. J. Talley, The mucosal immune system: master regulator of bidirectional gut-brain communications, Nat Rev Gastroenterol Hepatol, vol.14, pp.143-59, 2017.

K. S. Voo, Y. H. Wang, F. R. Santori, C. Boggiano, Y. H. Wang et al., Identification of IL-17-producing FOXP3+ regulatory T cells in humans, Proc Natl Acad Sci U S A, vol.106, pp.4793-98, 2009.

M. K. Jung, J. E. Kwak, and E. C. Shin, IL-17A-Producing Foxp3+ Regulatory T Cells and Human Diseases, Immune Netw. www.oncotarget.com, vol.17, pp.276-86, 2017.

P. A. De-barros, R. Andrade, M. E. De-vasconcelos-generoso, S. , M. Miranda et al., Conjugated linoleic acid prevents damage caused by intestinal mucositis induced by 5-fluorouracil in an experimental model, Biomed Pharmacother, vol.103, pp.1567-76, 2018.

F. Galdino, M. Andrade, P. De-barros, S. V. Generoso, J. I. Alvarez-leite et al., Pretreatment and treatment with fructo-oligosaccharides attenuate intestinal mucositis induced by 5-FU in mice, J Funct Foods, vol.49, pp.485-92, 2018.

H. R. Wardill, R. J. Gibson, R. M. Logan, and J. M. Bowen, TLR4/ PKC-mediated tight junction modulation: a clinical marker of chemotherapy-induced gut toxicity?, Int J Cancer, vol.135, pp.2483-92, 2014.

D. Corridoni, L. Pastorelli, B. Mattioli, S. Locovei, D. Ishikawa et al., Probiotic bacteria regulate intestinal epithelial permeability in experimental ileitis by a TNF-dependent mechanism, PLoS One, vol.7, 2012.

R. Mennigen, K. Nolte, E. Rijcken, M. Utech, B. Loeffler et al., Probiotic mixture VSL#3 protects the epithelial barrier by maintaining tight junction protein expression and preventing apoptosis in a murine model of colitis, Am J Physiol Gastrointest Liver Physiol, vol.296, pp.1140-1189, 2009.

P. Henderson, J. E. Van-limbergen, J. Schwarze, and D. C. Wilson, Function of the intestinal epithelium and its dysregulation in inflammatory bowel disease, Inflamm Bowel Dis, vol.17, pp.382-95, 2011.

H. Mi, Y. Dong, B. Zhang, H. Wang, C. C. Peter et al., Bifidobacterium Infantis Ameliorates Chemotherapy-Induced Intestinal Mucositis Via Regulating T Cell Immunity in Colorectal Cancer Rats, Cell Physiol Biochem, vol.42, pp.2330-2371, 2017.

P. F. Justino, L. F. Melo, A. F. Nogueira, C. M. Morais, W. O. Mendes et al., Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice, Cancer Chemother Pharmacol, vol.75, pp.559-67, 2015.

B. Foligne, S. Nutten, C. Grangette, V. Dennin, D. Goudercourt et al., Correlation between in vitro and in vivo immunomodulatory properties of lactic acid bacteria, World J Gastroenterol, vol.13, pp.236-279, 2007.

C. V. Suschek, O. Schnorr, and V. Kolb-bachofen, The role of iNOS in chronic inflammatory processes in vivo: is it damagepromoting, protective, or active at all?, Curr Mol Med, vol.4, pp.763-75, 2004.

D. Miljkovic and V. Trajkovic, Inducible nitric oxide synthase activation by interleukin-17, Cytokine Growth Factor Rev, vol.15, pp.21-32, 2004.

J. H. Niess, F. Leithäuser, G. Adler, and J. Reimann, Commensal gut flora drives the expansion of proinflammatory CD4 T cells in the colonic lamina propria under normal and inflammatory conditions, J Immunol, vol.180, pp.559-68, 2008.

A. C. Malik, G. W. Reinbold, and E. R. Vedamuthu, An evaluation of the taxonomy of Propionibacterium, Can J Microbiol, vol.14, pp.1185-91, 1968.

B. R. Macpherson and C. J. Pfeiffer, Experimental production of diffuse colitis in rats, Digestion, vol.17, pp.135-50, 1978.

S. Rocha, C. Gomes-santos, A. C. , G. Moreira, T. De-azevedo et al., Local and systemic immune mechanisms underlying the anti-colitis effects of the dairy bacterium Lactobacillus delbrueckii, PLoS One, vol.9, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204288

R. M. Rezende, R. P. Oliveira, S. R. Medeiros, A. C. Gomes-santos, A. C. Alves et al., Hsp65-producing Lactococcus lactis prevents experimental autoimmune encephalomyelitis in mice by inducing CD4+LAP+ regulatory T cells, J Autoimmun, vol.40, pp.45-57, 2013.

M. C. Canesso, L. Lemos, T. C. Neves, F. M. Marim, T. B. Castro et al., The cytosolic sensor STING is required for intestinal homeostasis and control of inflammation, Mucosal Immunol, vol.11, pp.820-854, 2018.

J. S. Oliveira, K. Costa, L. B. Acurcio, S. H. Sandes, G. D. Cassali et al., In vitro and in vivo evaluation of two potential probiotic lactobacilli isolated from cocoa fermentation (Theobroma cacao L.), J Funct Foods, vol.47, pp.184-91, 2018.

A. Giulietti, L. Overbergh, D. Valckx, B. Decallonne, R. Bouillon et al., An overview of real-time quantitative PCR: applications to quantify cytokine gene expression, Methods, vol.25, pp.386-401, 2001.

G. Q. Hu, P. X. Song, N. Li, W. Chen, Q. Q. Lei et al., AIM2 contributes to the maintenance of intestinal integrity via Akt and protects against Salmonella mucosal infection, Mucosal Immunol, vol.9, pp.1330-1369, 2016.

R. Tokumasu, K. Yamaga, Y. Yamazaki, H. Murota, K. Suzuki et al., Dosedependent role of claudin-1 in vivo in orchestrating features of atopic dermatitis, Proc Natl Acad Sci U S A, vol.113, pp.4061-68, 2016.

M. Wlodarska, B. Willing, K. M. Keeney, A. Menendez, K. S. Bergstrom et al., Antibiotic treatment alters the colonic mucus layer and predisposes the host to exacerbated Citrobacter rodentiuminduced colitis, Infect Immun, vol.79, pp.1536-1581, 2011.

J. Hellemans, G. Mortier, A. De-paepe, F. Speleman, and J. Vandesompele, qBase relative quantification framework and software for management and automated analysis of real-time quantitative PCR data, Genome Biol, vol.8, p.19, 2007.

C. Carrasco-pozo, P. Morales, and M. Gotteland, Polyphenols protect the epithelial barrier function of Caco-2 cells exposed to indomethacin through the modulation of occludin and zonula occludens-1 expression, J Agric Food Chem, vol.61, pp.5291-97, 2013.