E. Altermann, W. M. Russell, M. A. Azcarate-peril, and R. Barrangou, Complete genome sequence of the probiotic lactic acid bacterium Lactobacillus acidophilus NCFM, Proc. Natl. Acad. Sci, vol.102, pp.3906-3912, 2005.

H. An, F. P. Douillard, G. Wang, and Z. Zhai, Integrated Transcriptomic and Proteomic Analysis of the Bile Stress Response in a Centenarian-originated Probiotic Bifidobacterium longum BBMN68, Mol. Cell. Proteomics, vol.13, pp.2558-2572, 2014.

Y. Bao, K. F. Al, R. M. Chanyi, and S. Whiteside, Questions and challenges associated with studying the microbiome of the urinary tract, Ann. Transl. Med, vol.5, pp.33-33, 2017.

A. Bateman, M. Holden, and C. Yeats, The G5 domain: a potential N-acetylglucosamine recognition domain involved in biofilm formation, Bioinformatics, vol.21, pp.1301-1303, 2005.

Y. Belkaid and T. W. Han, Role of the Microbiota in Immunity and Inflammation, Cell, vol.157, pp.121-141, 2014.

M. Bermudez-brito, J. Plaza-díaz, S. Muñoz-quezada, and C. Gómez-llorente, Probiotic Mechanisms of Action, Ann. Nutr. Metab, vol.61, pp.160-174, 2012.

A. Bezkorovainy, Probiotics: determinants of survival and growth in the gut, Am. J. Clin. Nutr, vol.73, pp.399-405, 2001.

L. Bianchi, L. Laghi, V. Correani, and E. Schifano, A Combined Proteomics, Metabolomics and In Vivo Analysis Approach for the Characterization of Probiotics in Large-Scale Production, Biomolecules, vol.10, p.157, 2020.

D. J. Bibel, Elie Metchnikoff's Bacillus of Long Life, ASM News, vol.54, pp.661-665, 1988.

R. Bibiloni, R. N. Fedorak, G. W. Tannock, and K. L. Madsen, VSL#3 Probiotic-Mixture Induces Remission in Patients with Active Ulcerative Colitis, Am. J. Gastroenterol, vol.100, pp.1539-1546, 2005.

M. Bilen, J. Dufour, J. C. Lagier, and F. Cadoret, The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. Microbiome, vol.6, p.94, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01858939

L. Bo, J. Li, T. Tao, and Y. Bai, Probiotics for preventing ventilator-associated pneumonia, Cochrane Database Syst. Rev, 2014.

A. Bollmann, L. K. Epstein, and S. S. , Incubation of Environmental Samples in a Diffusion Chamber Increases the Diversity of Recovered Isolates, Appl. Environ. Microbiol, vol.73, pp.6386-6390, 2007.

A. Bolotin, B. Quinquis, P. Renault, and A. Sorokin, Complete sequence and comparative genome analysis of the dairy bacterium Streptococcus thermophilus, Nat. Biotechnol, vol.22, pp.1554-1558, 2004.

A. Bolotin, P. Wincker, S. Mauger, and O. Jaillon, The Complete Genome Sequence of the Lactic Acid Bacterium, Genome Res, pp.731-753, 2001.
URL : https://hal.archives-ouvertes.fr/hal-02681585

H. Brüssow, Phages of dairy bacteria, Annu. Rev. Microbiol, vol.55, pp.283-303, 2001.

J. Bu and Z. Wang, Cross-Talk between Gut Microbiota and Heart via the Routes of Metabolite and Immunity, Gastroenterol. Res. Pract, pp.1-8, 2018.

B. L. Buck, E. Altermann, T. Svingerud, and T. R. Klaenhammer, Functional Analysis of Putative Adhesion Factors in Lactobacillus acidophilus NCFM, Appl. Environ. Microbiol, vol.71, pp.8344-8351, 2005.

N. Buntin, T. Hongpattarakere, J. Ritari, and F. P. Douillard, An inducible operon is involved in inulin utilization in Lactobacillus plantarum strains, as revealed by comparative proteogenomics and metabolic profiling, Appl. Environ. Microbiol, 2017.

F. Calcinaro, S. Dionisi, M. Marinaro, and P. Candeloro, Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse, Diabetologia, vol.48, pp.1565-1575, 2005.

D. Cavanagh, C. M. Guinane, H. Neve, and A. Coffey, Phages of non-dairy lactococci: isolation and characterization of ?L47, a phage infecting the grass isolate Lactococcus lactis ssp. cremoris DPC6860, Front. Microbiol, vol.4, pp.1-15, 2014.

C. S. Chae, H. Kwon, J. Hwang, and J. Kim, Prophylactic Effect of Probiotics on the Development of Experimental Autoimmune Myasthenia Gravis, PLoS One, vol.7, p.52119, 2012.

S. Chaillou, -. Champomier, M. Cornet, C. Coq, and A. , The complete genome sequence of the meat-borne lactic acid bacterium Lactobacillus sakei, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02683108

G. Chauviere, M. Coconnier, S. Kerneis, and J. Fourniat, Adhesion of human Lactobacillus acidophilus strain LB to human enterocyte-like Caco-2 cells, J. Gen. Microbiol, vol.138, pp.1689-1696, 1992.

N. Colliou, Y. Ge, B. Sahay, and M. Gong, Commensal Propionibacterium strain UF1 mitigates intestinal inflammation via Th17 cell regulation, J. Clin. Invest, vol.127, pp.3970-3986, 2017.

B. Corcoran, C. Stanton, F. G. Ross, and R. , Life Under Stress: The Probiotic Stress Response and How it may be Manipulated, Curr. Pharm. Des, 2008.

P. D. Cotter, C. Hill, and R. P. Ross, Bacteriocins: developing innate immunity for food, Nat. Rev. Microbiol, vol.3, pp.777-788, 2005.

F. J. Cousin, S. Jouan-lanhouet, N. Théret, and C. Brenner, The probiotic Propionibacterium freudenreichii as a new adjuvant for TRAIL-based therapy in colorectal cancer, Oncotarget, vol.7, pp.7161-7178, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01404971

A. B. Crawley and R. Barrangou, Conserved Genome Organization and Core Transcriptome of the Lactobacillus acidophilus, Complex. Front. Microbiol, vol.9, p.1834, 2018.

M. Cronin, M. Ventura, F. Gf, and D. Van-sinderen, Progress in genomics, metabolism and biotechnology of bifidobacteria, Int. J. Food Microbiol, vol.149, pp.4-18, 2011.

P. R. David, B. Berger, F. Desiere, and D. Vilanova, The genome sequence of the probiotic intestinal bacterium Lactobacillus johnsonii NCC 533, Available at: www.ncbi.nlm.nih.govTaxonomy, vol.101, pp.2512-2517, 2004.

M. De-la-plaza, F. De-palencia, P. Peláez, and C. , Biochemical and molecular characterization of ?ketoisovalerate decarboxylase, an enzyme involved in the formation of aldehydes from amino acids by Lactococcus lactis, FEMS Microbiol. Lett, vol.238, pp.367-374, 2004.

A. De-moreno-de-leblanc, S. Del-carmen, M. Zurita-turk, S. Rocha, and C. , Importance of IL-10 Modulation by Probiotic Microorganisms in Gastrointestinal Inflammatory Diseases, ISRN Gastroenterol, p.892971, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01204334

E. Denou, R. D. Pridmore, B. Berger, and J. M. Panoff, Identification of genes associated with the long-gutpersistence phenotype of the probiotic Lactobacillus johnsonii strain NCC533 using a combination of genomics and transcriptome analysis, J. Bacteriol, vol.190, pp.3161-3168, 2008.

S. Deutsch, M. Mariadassou, P. Nicolas, and S. Parayre, Identification of proteins involved in the antiinflammatory properties of Propionibacterium freudenreichii by means of a multi-strain study, Sci. Rep, vol.7, p.46409, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01510019

S. Deutsch, M. Mariadassou, I. Díaz-muñiz, D. S. Banavara, M. F. Budinich et al., Lactobacillus casei metabolic potential to utilize citrate as an energy source in ripening cheese: A bioinformatics approach, J. Appl. Microbiol, vol.101, pp.872-882, 2006.

R. P. Dickson, J. R. Erb-downward, and G. B. Huffnagle, The role of the bacterial microbiome in lung disease, Expert Rev. Respir. Med, vol.7, pp.245-257, 2013.

X. Didelot, R. Bowden, D. J. Wilson, and T. Peto, Transforming clinical microbiology with bacterial genome sequencing, Nat. Rev. Genet, vol.13, pp.601-612, 2012.

T. G. Dinan and J. F. Cryan, Microbes, Immunity and Behavior: Psychoneuroimmunology Meets the Microbiome, vol.42, pp.178-192, 2017.

F. Do-carmo, H. Rabah, S. Huang, and F. Gaucher, Mutation of the Surface Layer Protein SlpB Has Pleiotropic Effects in the Probiotic Propionibacterium freudenreichii CIRM-BIA 129, Front. Microbiol, vol.8, p.1807, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01857987

P. Douillard, A. Ribbera, H. M. Järvinen, and R. Kant, Comparative Genomic and Functional Analysis of Lactobacillus casei and Lactobacillus rhamnosus Strains Marketed as Probiotics, Appl. Environ. Microbiol, vol.79, pp.1923-1933, 2013.

J. Duan, H. Chung, T. E. Kasper, and D. L. , Microbial Colonization Drives Expansion of IL-1 Receptor 1-Expressing and IL-17-Producing ?/? T Cells, Cell Host Microbe, vol.7, pp.140-150, 2010.

G. Dubourg, J. C. Lagier, C. Robert, and F. Armougom, Culturomics and pyrosequencing evidence of the reduction in gut microbiota diversity in patients with broad-spectrum antibiotics, Int. J. Antimicrob. Agents, vol.44, pp.117-124, 2014.

W. B. Dunn and D. I. Ellis, Metabolomics: Current Analytical Platforms and Methodologies, Trends Anal Chem, vol.24, pp.285-294, 2005.

B. Foligné, D. C. Pot, and B. , Probiotics from research to market: the possibilities, risks and challenges, Curr. Opin. Microbiol, vol.16, pp.284-292, 2013.

B. Foligné, S. Deutsch, J. Breton, and F. J. Cousin, 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.

M. Francino, Early Development of the Gut Microbiota and Immune Health, Pathogens, vol.3, pp.769-790, 2014.

R. Frei, A. M. , O. , and L. , Prebiotics, probiotics, synbiotics, and the immune system, Curr. Opin. Gastroenterol, vol.31, pp.153-158, 2015.

V. Gagnaire, J. Jardin, H. Rabah, and V. Briard-bion, Emmental Cheese Environment Enhances Propionibacterium freudenreichii Stress Tolerance, PLoS One, vol.10, p.135780, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01372936

R. L. Gallo and L. V. Hooper, Epithelial antimicrobial defence of the skin and intestine, Nat. Rev. Immunol, vol.12, pp.503-516, 2012.

J. E. Garneau and S. Moineau, Bacteriophages of lactic acid bacteria and their impact on milk fermentations, Microb. Cell Fact, vol.10, p.20, 2011.

A. Gandhi and N. P. Shah, Integrating Omics to Unravel the Stress-Response Mechanisms in Probiotic Bacteria: Approaches, Challenges, and Prospects, Crit Rev Food Sci, vol.57, pp.3464-3471, 2017.

O. Gilad, B. Svensson, A. H. Viborg, and B. Stuer-lauridsen, The extracellular proteome of Bifidobacterium animalis subsp. lactis BB-12 reveals proteins with putative roles in probiotic effects, Proteomics, vol.11, pp.2503-2514, 2011.

Y. J. Goh and T. R. Klaenhammer, Functional roles of aggregation-promoting-like factor in stress tolerance and adherence of Lactobacillus acidophilus NCFM, Appl. Environ. Microbiol, vol.76, pp.5005-5012, 2010.

A. Gratia, Sur un remarquable exemple d'antagonisme entre deux souches de colibacille, Compt. Rend. Soc. Biol, vol.93, pp.1040-1042, 1925.

E. Hamon, P. Horvatovich, E. Izquierdo, and F. Bringel, Comparative proteomic analysis of Lactobacillus plantarum for the identification of key proteins in bile tolerance, BMC Microbiol, vol.11, p.63, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00644083

J. Handelsman, Metagenomics: Application of Genomics to Uncultured Microorganisms. Microbiol, Mol. Biol. Rev, vol.69, pp.195-195, 2005.

J. Handelsmanl, &. Rondon, . Mr, S. F. Brady, and J. Clardy, Molecular biological access to the chemistry of unknown 05SP 09 soil microbes: a new frontier for natural products E4 + qfs, Chem. Biol, vol.5, pp.245-249, 1998.

H. Harris, M. Bourin, C. Mj, and O. Pw, Phylogenomics and comparative genomics of Lactobacillus salivarius, a mammalian gut commensal, Microb. Genomics, vol.3, pp.1-16, 2017.

F. He, A. C. Ouwehand, H. Hashimoto, and E. Isolauri, Adhesion of Bifidobacterium spp. to human intestinal mucus, Microbiol. Immunol, vol.45, pp.259-262, 2001.

H. Da and D. Artis, Intestinal Bacteria and the Regulation of Immune Cell Homeostasis, Annu. Rev. Immunol, vol.28, pp.623-667, 2010.

A. Hirsch, E. Grinsted, C. Hr, and A. Mattick, A note on the inhibition of an anaerobic sporeformer in Swiss-type cheese by a nisin-producing streptococcus, J. Dairy Res, vol.18, pp.205-206, 1951.

Y. Hong, Y. Nishimura, D. Hishikawa, and H. Tsuzuki, Acetate and Propionate Short Chain Fatty Acids Stimulate Adipogenesis via GPCR43, Endocrinology, vol.146, pp.5092-5099, 2005.

A. Hosono, Functions of fermented milk. Challenges for the health sciences, pp.61-78, 1992.

T. H. Howell, Metchnikoff and prolongation of life, Age Ageing, vol.17, pp.420-421, 1988.

D. W. Huang, S. Bt, and R. A. Lempicki, Bioinformatics Enrichment Tools: Paths toward the Comprehensive Functional Analysis of Large Gene Lists, Nucleic Acids Res, vol.37, pp.1-13, 2009.

P. Hugon, J. C. Dufour, P. Colson, and P. E. Fournier, A comprehensive repertoire of prokaryotic species identified in human beings, Lancet Infect. Dis, vol.15, pp.1211-1219, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01643991

R. W. Jack, J. R. Tagg, and B. Ray, Bacteriocins of Gram-Positive Bacteria, Microbiol. Rev, vol.59, pp.171-200, 1995.

G. Jan, A. Belzacq, D. Haouzi, and A. Rouault, Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria, Cell Death Differ, vol.9, pp.179-188, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00088749

G. Jan, P. Leverrier, V. Pichereau, and P. Boyaval, Changes in Protein Synthesis and Morphology during Acid Adaptation of Propionibacterium freudenreichii, Appl. Environ. Microbiol, vol.67, pp.2029-2036, 2001.

R. Janik, L. Thomason, A. M. Stanisz, and P. Forsythe, Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, N-acetyl aspartate and glutamate, Neuroimage, vol.125, pp.988-995, 2016.

M. L. Jiménez-pranteda, A. Pérez-davó, M. Monteoliva-sánchez, and A. Ramos-cormenzana, Food Omics Validation: Towards Understanding Key Features for Gut Microbiota, Probiotics and Human Health, Food Analytical Methods, vol.8, issue.2, pp.272-289, 2015.

J. Br and T. R. Klaenhammer, Impact of genomics on the field of probiotic research: historical perspectives to modern paradigms, Anto Leeuw Int. J. G, vol.106, pp.141-156, 2014.

A. M. Kabat, N. Srinivasan, and K. J. Maloy, Modulation of immune development and function by intestinal microbiota, Trends Immunol, vol.35, pp.507-517, 2014.

H. Kang and S. Im, Probiotics as an Immune Modulator, S103-S105, vol.61, 2015.

M. Kankainen, L. Paulin, S. Tynkkynen, V. Ossowski, and I. , Comparative genomic analysis of Lactobacillus rhamnosus GG reveals pili containing a human-mucus binding protein, Proc. Natl. Acad. Sci, vol.106, pp.17193-17198, 2009.

N. G. Kapse, A. S. Engineer, V. Gowdaman, and S. Wagh, Functional annotation of the genome unravels probiotic potential of Bacillus coagulans HS243, Genomics, vol.111, pp.921-929, 2018.

T. M. Karpi?ski and A. K. Szkaradkiewicz, Characteristic of bacteriocines and their application, Polish J. Microbiol, vol.62, pp.223-235, 2013.

K. Khalid, An overview of lactic acid bacteria, Int. J. Biosci, vol.1, pp.1-13, 2011.

T. Klaenhammer, Genetics of bacteriocins produced by lactic acid bacteria, FEMS Microbiol. Rev, vol.12, issue.93, p.90057, 1993.

T. Klaenhammer, E. Altermann, F. Arigoni, and A. Bolotin, Discovering lactic acid bacteria by genomics, Anto Leeuw Int. J. G, vol.82, pp.29-58, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01190023

T. R. Klaenhammer, M. Kleerebezem, M. V. Kopp, and M. Rescigno, The impact of probiotics and prebiotics on the immune system, Nat. Rev. Immunol, vol.12, pp.728-734, 2012.

M. Kleerebezem, J. Boekhorst, R. Van-kranenburg, and D. Molenaar, Complete genome sequence of Lactobacillus plantarum WCFS1, Proc. Natl. Acad. Sci, vol.100, 1990.

S. R. Konstantinov, H. Smidt, W. M. De-vos, . Bruijn, and . Scm, S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions, Proc. Natl. Acad. Sci, vol.105, pp.19474-19479, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01454074

G. Konuray and Z. Erginkaya, Potential Use of Bacillus coagulans in the Food Industry, p.92, 2018.

K. Koskenniemi, K. Laakso, J. Koponen, and M. Kankainen, Proteomics and Transcriptomics Characterization of Bile Stress Response in Probiotic Lactobacillus rhamnosus GG, Mol. Cell. Proteomics, vol.10, 2011.

L. Fata, G. , W. P. Mohajeri, and M. H. , Probiotics and the Gut Immune System: Indirect Regulation, Probiotics Antimicrob. Proteins, vol.10, pp.11-21, 2018.

J. Lagier, G. Dubourg, M. Million, and F. Cadoret, Culturing the human microbiota and culturomics, Nat. Rev. Microbiol, vol.16, pp.540-550, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01910305

J. Lagier, S. Khelaifia, M. T. Alou, and S. Ndongo, Culture of previously uncultured members of the human gut microbiota by culturomics, Nat. Microbiol, vol.1, p.16203, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01453324

J. C. Lagier, F. Armougom, M. Million, and P. Hugon, Microbial culturomics: Paradigm shift in the human gut microbiome study, Clin. Microbiol. Infect, vol.18, pp.1185-1193, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01478655

A. Lan, A. Bruneau, M. Bensaada, and C. Philippe, Increased induction of apoptosis by Propionibacterium freudenreichii TL133 in colonic mucosal crypts of human microbiota-associated rats treated with 1,2-dimethylhydrazine, Br. J. Nutr, vol.100, pp.1251-1259, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01454096

S. Lavasani, B. Dzhambazov, M. Nouri, and F. Fåk, A Novel Probiotic Mixture Exerts a Therapeutic Effect on Experimental Autoimmune Encephalomyelitis Mediated by IL-10 Producing Regulatory T Cells, PLoS One, vol.5, p.9009, 2010.

L. Maréchal, C. Peton, V. Plé, C. Vroland, and C. , Surface proteins of Propionibacterium freudenreichii are involved in its anti-inflammatory properties, J. Proteomics, vol.113, pp.447-461, 2015.

S. Lebeer, I. Claes, T. Verhoeven, and C. Shen, Impact of luxS and Suppressor Mutations on the Gastrointestinal Transit of Lactobacillus rhamnosus GG, Appl. Environ. Microbiol, vol.74, pp.4711-4718, 2008.

S. Lebeer, J. Vanderleyden, D. Keersmaecker, and S. , Host interactions of probiotic bacterial surface molecules: Comparison with commensals and pathogens, Nat. Rev. Microbiol, vol.8, pp.171-184, 2010.

E. Lee, E. Song, Y. Nam, and L. , Probiotics in human health and disease: from nutribiotics to pharmabiotics, J. Microbiol, vol.56, pp.773-782, 2018.

P. Leverrier, D. Dimova, V. Pichereau, and Y. Auffray, Susceptibility and Adaptive Response to Bile Salts in Propionibacterium freudenreichii: Physiological and Proteomic Analysis, Appl. Environ. Microbiol, vol.69, pp.3809-3818, 2003.

P. Leverrier, J. Vissers, A. Rouault, and P. Boyaval, Mass spectrometry proteomic analysis of stress adaptation reveals both common and distinct response pathways in Propionibacterium freudenreichii, Arch. Microbiol, vol.181, pp.215-230, 2004.

P. Li, X. Li, Q. Gu, and X. Lou, Comparative genomic analysis of Lactobacillus plantarum ZJ316 reveals its genetic adaptation and potential probiotic profiles, J Zhejiang Univ-Sci B (Biomed Biotechnol), vol.17, pp.569-579, 2016.

A. Llewellyn and A. Foey, Probiotic Modulation of Innate Cell Pathogen Sensing and Signaling Events, Nutrients, vol.9, p.1156, 2017.

L. V. Mcfarland, Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review, BMJ Open, vol.4, pp.5047-005047, 2014.

V. Medina, B. Edmonds, G. P. Young, and R. James, Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin a (Inhibitors of histone deacetylase): Dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway, Cancer Res, vol.57, pp.3697-3707, 1997.

S. Moineau and C. Lévesque, Control of Bacteriophages in Industrial Fermentations, Bacteriophages Biology and Applications, pp.1-12, 2004.

F. Mozzi, M. E. Ortiz, J. Bleckwedel, D. Vuyst, and L. , Metabolomics as a tool for the comprehensive understanding of fermented and functional foods with lactic acid bacteria, Food Res. Int, vol.54, pp.1152-1161, 2013.

K. Nobutani, D. Sawada, S. Fujiwara, and Y. Kuwano, The effects of administration of the Lactobacillus gasseri strain CP2305 on quality of life, clinical symptoms and changes in gene expression in patients with irritable bowel syndrome, J. Appl. Microbiol, vol.122, pp.212-224, 2017.

S. O'flaherty and T. R. Klaenhammer, The role and potential of probiotic bacteria in the gut, and the communication between gut microflora and gut/host, Int. Dairy J, vol.20, pp.262-268, 2010.

L. C. Oliveira, T. Saraiva, W. M. Silva, and U. P. Pereira, Analyses of the probiotic property and stress resistancerelated genes of Lactococcus lactis subsp. lactis NCDO 2118 through comparative genomics and in vitro assays, PLoS One, vol.12, p.175116, 2017.

A. C. Ouwehand, S. Salminen, and E. Isolauri, Probiotics an overview of beneficial effects, Anto Leeuw Int. J. G, vol.82, pp.279-289, 2002.

K. Papadimitriou, G. Zoumpopoulou, B. Foligné, and V. Alexandraki, Discovering probiotic microorganisms: in vitro, in vivo, genetic and omics approaches, Front Microbiol, vol.6, p.58, 2015.

N. Perakakis, A. Yazdani, G. E. Karniadakis, and C. Mantzoros, Omics, Big Data and Machine Learning as Tools to Propel Understanding of Biological Mechanisms and to Discover Novel Diagnostics and Therapeutics, Metabolism, vol.87, 2018.

E. A. Pfeiler and T. R. Klaenhammer, The genomics of lactic acid bacteria, Trends Microbiol, vol.15, pp.546-553, 2007.

A. Pfleiderer, J. C. Lagier, F. Armougom, and C. Robert, Culturomics identified 11 new bacterial species from a single anorexia nervosa stool sample, Eur. J. Clin. Microbiol. Infect. Dis, vol.32, pp.1471-1481, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00964140

C. K. Porter, R. Gormley, D. R. Tribble, and B. D. Cash, The Incidence and Gastrointestinal Infectious Risk of Functional Gastrointestinal Disorders in a Healthy US Adult Population, Am. J. Gastroenterol, vol.106, pp.130-138, 2011.

J. L. Round and S. K. Mazmanian, The gut microbiota shapes intestinal immune responses during health and disease, Nat. Rev. Immunol, vol.9, pp.313-323, 2009.

P. Russo, M. Mohedano, L. De-la, V. Capozzi, and P. F. De-palencia, Comparative proteomic analysis of Lactobacillus plantarum WCFS1 and ?ctsR mutant strains under physiological and heat stress conditions, Int. J. Mol. Sci, vol.13, pp.10680-10696, 2012.

S. Salminen, A. Wright, L. Morelli, and P. Marteau, Demonstration of safety of probiotics -a review, Int. J. Food Microbiol, vol.44, pp.93-106, 1998.

M. Salque, P. I. Bogucki, J. Pyzel, and I. Sobkowiak-tabaka, Earliest evidence for cheese making in the sixth millennium bc in northern Europe, Nature, vol.493, pp.522-525, 2013.

E. Salvetti, H. Harris, G. E. Felis, and O. Pw, Comparative genomics reveals robust phylogroups in the genus Lactobacillus as the basis for reclassification, Appl. Environ. Microbiol, vol.84, pp.993-1011, 2018.

B. Sánchez, M. Champomier-vergè, P. Anglade, and F. Baraige, Proteomic Analyis of Global Changes in Protein Expression during Bile Salt Exposure of Bifidobacterium longum NCIMB 8809, J. Bacteriol, vol.187, pp.5799-5808, 2005.

. Sander, Use of probiotics for infection prevention in trauma patients, Nurs. Older People, vol.29, p.11, 2017.

T. Saraoui, S. Parayre, G. Guernec, and V. Loux, A unique in vivo experimental approach reveals metabolic adaptation of the probiotic Propionibacterium freudenreichii to the colon environment, BMC Genomics, vol.14, p.911, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01209705

D. M. Saulnier, F. Santos, S. Roos, and T. Mistretta, Exploring Metabolic Pathway Reconstruction and Genome-Wide Expression Profiling in Lactobacillus reuteri to Define Functional Probiotic Features, PLoS One, vol.6, p.18783, 2011.

H. Schmidt and M. Hensel, Pathogenicity islands in bacterial pathogenesis, Clin. Microbiol. Rev, vol.17, pp.14-56, 2004.

S. Senan, J. B. Prajapati, and C. G. Joshi, Comparative genome-scale analysis of niche-based stress-responsive genes in Lactobacillus helveticus strains, Genome, vol.57, pp.185-192, 2014.

S. Senan, J. B. Prajapati, and C. G. Joshi, Feasibility of Genome-Wide Screening for Biosafety Assessment of Probiotics: A Case Study of Lactobacillus helveticus MTCC 5463, Probiotics Antimicrob. Proteins, vol.7, pp.249-258, 2015.

M. Sharma and G. Shukla, Metabiotics: One Step ahead of Probiotics; an Insight into Mechanisms Involved in Anticancerous Effect in Colorectal Cancer, Front. Microbiol, vol.7, p.1940, 2016.

Y. Shen and R. Nahas, Complementary and alternative medicine for treatment of irritable bowel syndrome, Can. Fam. physician Me?decin Fam. Can, vol.55, pp.143-151, 2009.

B. A. Shenderov, Metabiotics: novel idea or natural development of probiotic conception, Microb. Ecol. Heal. Dis, vol.24, 2013.

C. Shortt, The probiotic century: Historical and current perspectives, Trends Food Sci. Technol, vol.10, pp.411-417, 1999.

V. V. Smeianov, P. Wechter, J. R. Broadbent, and J. E. Hughes, Comparative High-Density Microarray Analysis of Gene Expression during Growth of Lactobacillus helveticus in Milk versus Rich Culture Medium, Appl. Environ. Microbiol, vol.73, pp.2661-2672, 2007.

B. A. Smit, J. Van-hylckama-vlieg, W. Engels, and L. Meijer, Identification, Cloning, and Characterization of a Lactococcus lactis Branched-Chain -Keto Acid Decarboxylase Involved in Flavor Formation, Appl. Environ. Microbiol, vol.71, pp.303-311, 2005.

J. So, H. Kwon, C. Lee, and H. Yi, Lactobacillus casei suppresses experimental arthritis by downregulating T helper 1 effector functions, Mol. Immunol, vol.45, pp.2690-2699, 2008.

S. Soares, A. Luiz, R. Thiago, and J. Ramos, Plasticidade Genômica e Evolução Bacteriana. Microbiol. Foco, vol.4, pp.31-38, 2011.

A. Sood, V. Midha, G. K. Makharia, and V. Ahuja, The Probiotic Preparation, VSL#3 Induces Remission in Patients With Mild-to-Moderately Active Ulcerative Colitis, Clin. Gastroenterol. Hepatol, vol.7, pp.1202-1209, 2009.

G. E. Soto and S. J. Hultgren, Bacterial Adhesins : Common Themes and Variations in Architecture and Assembly MINIREVIEW Bacterial Adhesins : Common Themes and Variations in Architecture and Assembly, vol.181, pp.1059-1071, 1999.

H. Sugahara, T. Odamaki, S. Fukuda, and T. Kato, Probiotic Bifidobacterium longum alters gut luminal metabolism through modification of the gut microbial community, Sci. Rep, vol.5, p.13548, 2015.

W. C. Summers, Bacteriophage Research: Early History, pp.1-485, 2005.

W. Sybesma, D. Molenaar, W. Van-ijcken, and K. Venema, Genome Instability in Lactobacillus rhamnosus GG, Appl. Environ. Microbiol, vol.79, pp.2233-2239, 2013.

M. Tidjani-alou, M. Million, S. I. Traore, and D. Mouelhi, Gut Bacteria Missing in Severe Acute Malnutrition, Can We Identify Potential Probiotics by, Culturomics? Front. Microbiol, vol.8, p.899, 2017.

E. M. Tuomola and S. J. Salminen, Adhesion of some probiotic and dairy Lactobacillus strains to Caco-2 cell cultures, Int. J. Food Microbiol, vol.41, pp.33-39, 1998.

P. J. Turnbaugh, R. E. Ley, M. A. Mahowald, and V. Magrini, An obesity-associated gut microbiome with increased capacity for energy harvest, Nature, vol.444, pp.1027-1031, 2006.

A. Tursi, G. Brandimarte, A. Papa, and A. Giglio, Treatment of Relapsing Mild-to-Moderate Ulcerative Colitis With the Probiotic VSL#3 as Adjunctive to a Standard Pharmaceutical Treatment: A Double-Blind, Randomized, Placebo-Controlled Study, Am. J. Gastroenterol, vol.105, pp.2218-2227, 2010.

Ö. Umu, C. Bäuerl, M. Oostindjer, and P. B. Pope, The Potential of Class II Bacteriocins to Modify Gut Microbiota to Improve Host Health, PLoS One, vol.11, p.164036, 2016.

P. Baarlen, F. Troost, C. Van-der-meer, and G. Hooiveld, Human mucosal in vivo transcriptome responses to three lactobacilli indicate how probiotics may modulate human cellular pathways, Proc. Natl. Acad. Sci, vol.108, pp.4562-4569, 2011.

M. J. Van-belkum and M. E. Stiles, Nonlantibiotic antibacterial peptides from lactic acid bacteria, Nat. Prod. Rep, vol.17, pp.323-335, 2000.

M. Van-de-guchte, E. Sd, E. Maguin, J. A. Kok, and J. , Transcriptome landscape of Lactococcus lactis reveals many novel RNAs including a small regulatory RNA involved in carbon uptake and metabolism, J. Appl. Microbiol, vol.91, pp.353-366, 2001.

M. P. Vélez, D. Keersmaecker, S. Vanderleyden, and J. , Adherence factors of Lactobacillus in the human gastrointestinal tract, FEMS Microbiol. Lett, vol.276, pp.140-148, 2007.

J. C. Venter, Environmental Genome Shotgun Sequencing of the Sargasso Sea, Science, vol.304, pp.66-74, 2004.

A. Verma and G. Shukla, Administration of prebiotic inulin suppresses 1,2 dimethylhydrazine dihydrochloride induced procarcinogenic biomarkers fecal enzymes and preneoplastic lesions in early colon carcinogenesis in Sprague Dawley rats, J. Funct. Foods, vol.5, pp.991-996, 2013.

M. Viljanen, E. Pohjavuori, T. Haahtela, and R. Korpela, Induction of inflammation as a possible mechanism of probiotic effect in atopic eczema-dermatitis syndrome, J. Allergy Clin. Immunol, vol.115, pp.1254-1259, 2005.

G. Weiss and L. Jespersen, Transcriptional Analysis of Genes Associated with Stress and Adhesion in Lactobacillus acidophilus NCFM during the Passage through an in vitro Gastrointestinal Tract Model, J. Mol. Microbiol. Biotechnol, vol.18, pp.206-214, 2010.

S. Westfall, N. Lomis, I. Kahouli, and S. Y. Dia, Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis, Cell. Mol. Life Sci, vol.74, pp.3769-3787, 2017.

K. E. Wommack and R. R. Colwell, Virioplankton: viruses in aquatic ecosystems. Microbiol, Mol. Biol. Rev, vol.64, pp.69-114, 2000.

J. Yang, Y. Ji, H. Park, and J. Lee, Selection of functional lactic acid bacteria as starter cultures for the fermentation of Korean leek (Allium tuberosum Rottler ex Sprengel.), Int. J. Food Microbiol, vol.191, pp.164-171, 2014.

J. Yuan, B. Wang, Z. Sun, and X. Bo, Analysis of Host-Inducing Proteome Changes in Bifidobacterium longum NCC2705 Grown in Vivo, J. Proteome Res, vol.7, pp.375-385, 2008.

J. Zheng, S. Wittouck, E. Salvetti, and C. Franz, A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae, Int. J. Syst. Evol. Microbiol, vol.70, 2020.

L. Zhong, X. Zhang, and M. Covasa, Emerging roles of lactic acid bacteria in protection against colorectal cancer, World J. Gastroenterol, vol.20, p.7878, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204393