E. L. Abel, Behavioral effects of isatin on open field activity and immobility in the forced swim test in rats, Physiol. Behav, vol.57, issue.3, pp.611-613, 1995.

A. Armario, N. Daviu, C. Muñoz-abellán, C. Rabasa, S. Fuentes et al., What can we know from pituitary-adrenal hormones about the nature and consequences of exposure to emotional stressors?, Cell. Mol. Neurobiol, vol.32, pp.749-758, 2012.

T. Baba, T. Ara, M. Hasegawa, Y. Takai, Y. Okumura et al., Construction of Escherichia coli K-12 inframe, single-gene knockout mutants: the Keio collection, Mol. Syst. Biol, vol.2, p.8, 2006.

T. Bansal, R. C. Alaniz, T. K. Wood, and A. Jayaraman, The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation, Proc. Natl Acad. Sci. USA, vol.107, issue.1, pp.228-233, 2010.

S. K. Bhattacharya, S. K. Mitra, and S. B. Acharya, Anxiogenic activity of isatin, a putative biological factor, in rodents, J. Psychopharmacol, vol.5, issue.3, pp.202-206, 1991.

R. Carpenedo, G. Mannaioni, and F. Moroni, Oxindole, a sedative tryptophan metabolite, accumulates in blood and brain of rats with acute hepatic failure, J. Neurochem, vol.70, issue.5, pp.1998-2003, 1998.

C. Chimerel, E. Emery, D. K. Summers, U. Keyser, F. M. Gribble et al., Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells, Cell Rep, vol.9, issue.4, pp.1202-1208, 2014.
DOI : 10.1016/j.celrep.2014.10.032

URL : https://doi.org/10.1016/j.celrep.2014.10.032

G. Clarke, S. Grenham, P. Scully, P. Fitzgerald, R. D. Moloney et al., The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner, Mol. Psychiatry, vol.18, issue.6, pp.666-673, 2013.

M. Crumeyrolle-arias, M. Jaglin, A. Bruneau, S. Vancassel, A. Cardona et al., Absence of the gut microbiota enhances anxietylike behavior and neuroendocrine response to acute stress in rats, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204296

, Psychoneuroendocrinology, vol.42, pp.207-217

J. F. Cryan and T. G. Dinan, Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour, Nat. Rev. Neurosci, vol.13, pp.701-712, 2012.

R. M. Deacon, Assessing nest building in mice, Nat Protoc, vol.1, issue.3, pp.1117-1119, 2006.

R. M. Deacon, Assessing burrowing, nest construction, and hoarding in mice, J. Vis. Exp, vol.59, p.2607, 2012.

G. De-palma, M. D. Lynch, J. Lu, V. T. Dang, Y. Deng et al., Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice, Sci. Transl. Med, vol.9, issue.379, 2017.

A. Ennaceur, S. Michalikova, A. Bradford, and S. Ahmed, Detailed analysis of the behavior of Lister and Wistar rats in anxiety, object recognition and object location tasks, Behav. Brain Res, vol.159, pp.247-266, 2005.

V. Fournet, A. Schweitzer, C. Chevarin, J. C. Deloulme, M. Hamon et al., The deletion of STOP/MAP6 protein in mice triggers highly altered mood and impaired cognitive performances, J. Neurochem, vol.121, issue.1, pp.99-114, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00734692

J. P. Guilloux, M. Seney, N. Edgar, and E. Sibille, Integrated behavioral z-scoring increases the sensitivity and reliability of behavioral phenotyping in mice: relevance to emotionality and sex, J. Neurosci. Methods, vol.197, issue.1, pp.21-31, 2011.

Y. Ibarguen-vargas, A. Surget, C. Touma, R. Palme, and C. Belzung, Multifaceted strain-specific effects in a mouse model of depression and of antidepressant reversal, Psychoneuroendocrinology, vol.33, issue.10, pp.1357-68, 2008.

M. Jaglin, M. Rhimi, C. Philippe, N. Pons, A. Bruneau et al., Indole, a signaling molecule produced by the gut microbiota, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01837669

. Neurosci, , vol.12, p.216

H. Jiang, Z. Ling, Y. Zhang, H. Mao, Z. Ma et al., Altered fecal microbiota composition in patients 104 with major depressive disorder, Brain Behav. Immun, vol.48, pp.186-194, 2015.

B. J. Jones, T. Tan, and S. Bloom, Minireview: glucagon in stress and energy homeostasis, Endocrinology, vol.153, issue.3, pp.1049-1054, 2012.

M. E. Keck, T. Welt, M. B. Müller, M. Uhr, F. Ohl et al., Reduction of hypothalamic vasopressinergic hyperdrive contributes to clinically relevant behavioral and neuroendocrine effects of chronic paroxetine treatment in a psychopathological rat model, Neuropsychopharmacology, vol.28, pp.235-243, 2003.

J. R. Kelly, Y. Borre, C. O'-brien, E. Patterson, S. El-aidy et al., Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat, 2016.

, J. Psychiatr. Res, vol.82, pp.109-118

L. J. King, D. V. Parke, and R. T. Williams, The metabolism of [2-14C] indole in the rat, Biochem. J, vol.98, issue.1, pp.266-277, 1966.

G. Krause, L. Winkler, S. L. Mueller, R. F. Haseloff, J. Piontek et al., Structure and function of claudins, Biochim. Biophys. Acta, vol.1778, issue.3, pp.631-645, 2008.

B. Lamas, M. L. Richard, V. Leducq, H. P. Pham, M. L. Michel et al., CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands, Nature Medicine, vol.22, issue.6, pp.598-605, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01314089

,

J. H. Lee, T. K. Wood, and J. Lee, Roles of indole as an interspecies and interkingdom signaling molecule, Trends Microbiol, vol.23, issue.11, pp.707-718, 2015.

J. Li, H. Jia, X. Cai, H. Zhong, Q. Feng et al., , p.105

M. Consortium, P. Bork, and J. Wang, An integrated catalog of reference genes in the human gut microbiome, 2014.

, Biotechnol, vol.32, issue.8, pp.834-841

J. Liu, M. W. Marino, G. Wong, D. Grail, A. Dunn et al., TNF is a potent anti-inflammatory cytokine in autoimmune-mediated demyelination, Nat. Med, vol.4, issue.1, pp.78-83, 1998.

A. Medvedev, N. Igosheva, M. Crumeyrolle-arias, and V. Glover, Isatin: role in stress and anxiety, Stress, vol.8, pp.175-183, 2005.

A. Naseribafrouei, K. Hestad, E. Avershina, M. Sekelja, A. Linløkken et al., Correlation between the human fecal microbiota and depression, 2014.

, Neurogastroenterol. Motil, vol.26, issue.8

J. M. Natividad, A. Agus, J. Planchais, B. Lamas, A. C. Jarry et al.,

B. Hansel, J. M. Launay, R. J. Xavier, H. Duboc, and H. Sokol, Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome, Cell Metab, vol.28, issue.5, pp.737-749, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01895943

N. Ohkura, K. Oishi, Y. Sekine, G. Atsumi, N. Ishida et al., Comparative study of circadian variation in numbers of peripheral blood cells among mouse strains: unique feature of C3H/HeN mice, Biol. Pharm. Bull, vol.30, issue.6, 2007.

C. M. Pariante and S. L. Lightman, The HPA axis in major depression: classical theories and new developments, Trends Neurosci, vol.31, issue.9, pp.464-468, 2008.

M. I. Pinto-sanchez, G. B. Hall, K. Ghajar, A. Nardelli, C. Bolino et al., Probiotic Bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: a pilot study in patients with irritable bowel syndrome, Gastroenterology, vol.153, pp.448-459, 2017.

E. M. Richard, J. C. Helbling, C. Tridon, A. Desmedt, A. M. Minni et al., Plasma transcortin influences endocrine and behavioral stress responses in mice, Endocrinology, vol.151, pp.649-659, 2010.

R. Rosenthal, D. Günzel, D. Theune, C. Czichos, J. D. Schulzke et al., Water channels and barriers formed by claudins, Annals of the New York Academy of Sciences, vol.1397, issue.1, pp.100-109, 2017.

A. Sarrieau, F. Chaouloff, V. Lemaire, and P. Mormède, Comparison of the neuroendocrine responses to stress in outbred, inbred and F1 hybrid rats, Life Sci, vol.63, pp.87-96, 1998.

Y. Shimada, M. Kinoshita, K. Harada, M. Mizutani, K. Masahata et al., Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon, PLoS ONE, vol.8, issue.11, pp.1-10, 2013.

H. A. Slotten, M. Kalinichev, J. J. Hagan, C. A. Marsden, and K. C. Fone, Long-lasting changes in behavioural and neuroendocrine indices in the rat following neonatal maternal separation: gender-dependent effects, Brain Res, vol.1097, pp.123-132, 2006.

R. Sonowal, A. Swimm, A. Sahoo, L. Luo, Y. Matsunaga et al., Indoles from commensal bacteria extend healthspan, Proc. Natl. Acad Sci. USA, vol.114, issue.36, 2017.
DOI : 10.1073/pnas.1706464114

URL : https://www.pnas.org/content/pnas/114/36/E7506.full.pdf

L. Steru, R. Chermat, B. Thierry, and P. Simon, The tail suspension test: a new method for screening antidepressants in mice, Psychopharmacology (Berl), vol.85, issue.3, pp.367-370, 1985.

N. Sudo, Y. Chida, Y. Aiba, J. Sonoda, N. Oyama et al., Postnatal microbial colonization programs the hypothalamic-pituitaryadrenal system for stress response in mice, J. Physiol, vol.558, issue.1, pp.263-275, 2004.

S. Vancassel, S. Leman, L. Hanonick, S. Denis, J. Roger et al., N-3 polyunsaturated fatty acid supplementation reverses stress-induced modifications on brain monoamine levels in mice, J. Lipid Res, vol.49, issue.2, pp.340-348, 2008.

K. Aagaard, J. Ma, M. Kathleen, A. Radhika, G. Petrosino et al., The Placenta Harbors a Unique Microbiome, Science Translational Medicine, vol.6, issue.237, pp.1-22, 2014.

E. L. Abel, Behavioral effects of isatin on open field activity and immobility in the forced swim test in rats, Physiology & Behavior, vol.57, issue.3, pp.611-613, 1995.

S. Adesso, T. Magnus, S. Cuzzocrea, M. Campolo, B. Rissiek et al., Indoxyl sulfate affects glial function increasing oxidative stress and neuroinflammation in chronic kidney disease: Interaction between astrocytes and microglia, Frontiers in Pharmacology, vol.8, pp.1-13, 2017.

E. Akimova, R. Lanzenberger, and S. Kasper, The Serotonin-1A Receptor in Anxiety Disorders, Biological Psychiatry, vol.66, issue.7, pp.627-635, 2009.

A. Anyanful, J. M. Dolan-livengood, T. Lewis, S. Sheth, M. N. Dezalia et al., Paralysis and killing of Caenorhabditis elegans by enteropathogenic Escherichia coli requires the bacterial tryptophanase gene, Molecular Microbiology, vol.57, issue.4, pp.988-1007, 2005.

A. Anyanful, K. A. Easley, G. M. Benian, and D. Kalman, Conditioning Protects C. elegans from Lethal Effects of Enteropathogenic E. coli by Activating Genes that Regulate Lifespan and Innate Immunity, Cell Host and Microbe, vol.5, issue.5, pp.450-462, 2009.

T. Arentsen, Y. Qian, S. Gkotzis, T. Femenia, T. Wang et al., The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior, Molecular Psychiatry, vol.22, issue.2, pp.257-266, 2017.

M. Arumugam, J. Raes, E. Pelletier, D. Paslier, . Le et al., Enterotypes of the human gut microbiome, Nature, vol.473, issue.7346, pp.174-180, 2013.
URL : https://hal.archives-ouvertes.fr/cea-00903625

A. Aziz and G. H. Anderson, Exendin-4, a GLP-1 receptor agonist, interacts with proteins and their products of digestion to suppress food intake in rats, The Journal of Nutrition, vol.133, pp.2326-2330, 2003.

T. L. Bale, A. Contarino, G. W. Smith, R. Chan, L. H. Gold et al.,

F. , Mice deficient for corticotropin-releasing hormone receptor-2 display anxiety-like behaviour and are hypersensitive to stress, Nature Genetics, vol.24, issue.4, pp.410-414, 2000.

E. Banoglu, G. G. Jha, and R. S. King, Hepatic microsomal metabolism of indole to indoxyl, a precursor of indoxyl sulfate, European Journal of Drug Metabolism and Pharmacokinetics, vol.26, issue.4, pp.235-240, 2001.

E. Banoglu and S. R. King, Sulfation of indoxyl by human and rat aryl (phenol) sulfotranferases to form indoxyl sulfate, European Journal of Drug Metabolism and Pharmacokinetics, vol.27, issue.2, pp.135-140, 2002.

T. Bansal, R. C. Alaniz, T. K. Wood, and A. Jayaraman, The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation, Proceedings of the National Academy of Sciences, vol.107, issue.1, pp.228-233, 2010.

R. C. Bean, W. C. Shepherd, and H. Chan, Permeability of Lipid Bilayer Membranes to Organic Solutes, The Journal of General Physiology, vol.52, pp.495-508, 1968.

P. Bercik, A. J. Park, D. Sinclair, A. Khoshdel, J. Lu et al., The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut -brain communication, Neurogastroenterology & Motility, vol.23, issue.12, pp.1132-1139, 2011.

A. Bharwani, M. F. Mian, M. G. Surette, J. Bienenstock, and P. Forsythe, Oral treatment with Lactobacillus rhamnosus attenuates behavioural deficits and immune changes in chronic social stress, BMC Medicine, vol.15, issue.7, pp.1-14, 2017.

S. K. Bhattacharya, S. K. Mitra, and S. B. Acharya, Anxiogenic activity of isatin, a putative biological factor in rodents, Journal of Psychopharmacology, vol.5, issue.3, pp.202-206, 1991.

S. K. Bhattacharya, A. Clow, A. Pzyborowska, J. Halket, V. Glover et al., Effect of aromatic amino acids, pentylenetetrazole and yohimbine on isatin and tribulin activity in rat brain, Neuroscience Letters, vol.132, issue.1, pp.44-46, 1991.

S. K. Bhattacharya and S. B. Acharya, Further investigations on the anxiogenic action of isatin, Biogenic Amines, vol.9, pp.453-463, 1993.

S. K. Bhattacharya, A. Chakrabarti, M. Sandler, and V. Glover, Anxiolytic activity of intraventricularly administered ANP in the rat, Neuropsychopharmacology, vol.15, pp.199-206, 1996.

B. Bommarius, A. Anyanful, Y. Izrayelit, S. Bhatt, E. Cartwright et al., A Family of Indoles Regulate Virulence and Shiga Toxin Production in Pathogenic E. coli, PLoS ONE, vol.8, issue.1, pp.1-16, 2013.

C. Booth, G. Tudor, J. Tudor, B. P. Katz, and T. Macvittie, The Acute Gastrointestinal Syndrome in High-Dose Irradiated Mice, Health Physics, vol.103, issue.4, pp.383-399, 2012.

J. A. Bravo, P. Forsythe, M. V. Chew, E. Escaravage, H. M. Savignac et al.,

J. F. Cryan, Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve, Proceedings of the National Academy of Sciences of the United States of America, vol.108, pp.16050-16055, 2011.

J. Bundzikova-osacka, S. Ghosal, B. A. Packard, Y. M. Ulrich-lai, and J. P. Herman, Role of nucleus of the solitary tract noradrenergic neurons in post-stress cardiovascular and hormonal control in male rats, Stress, vol.18, issue.2, pp.221-232, 2015.

A. Burokas, S. Arboleya, R. D. Moloney, V. L. Peterson, K. Murphy et al.,

J. F. Cryan, Targeting the Microbiota-Gut-Brain Axis : Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice, Biological Psychiatry, vol.82, issue.7, pp.472-487, 2017.

R. Carpenedo, G. Mannaioni, and F. Moroni, Oxindole, a sedative tryptophan metabolite, accumulates in blood and brain of rats with acute hepatic failure, Journal of Neurochemistry, vol.70, issue.5, 1998.

B. Chassaing, J. D. Aitken, M. Malleshappa, and M. Vijay-kumar, Dextran Sulfate Sodium (DSS)-Induced Colitis in Mice, Current Protocols in Immunology, vol.104, issue.1, 2014.

J. Chen, C. Zhou, P. Zheng, K. Cheng, H. Wang et al., Differential urinary metabolites related with the severity of major depressive disorder, Behavioural Brain Research, vol.332, pp.280-287, 2017.

C. Chimerel, C. M. Field, S. Piñero-fernandez, U. F. Keyser, and D. K. Summers, , 2012.

, Indole prevents Escherichia coli cell division by modulating membrane potential, Biochimica et Biophysica Acta -Biomembranes, vol.1818, issue.7, pp.1590-1594

C. Chimerel, E. Emery, D. K. Summers, U. Keyser, F. M. Gribble et al., , 2014.

, Bacterial Metabolite Indole Modulates Incretin Secretion from Intestinal Enteroendocrine L Cells. Cell Reports, vol.9, issue.4, pp.1202-1208

Z. V. Chirkova, M. V. Kabanova, S. I. Filimonov, I. G. Abramov, A. Petzer et al.,

K. Y. Suponitsky, Inhibition of monoamine oxidase by indole-5,6-dicarbonitrile derivatives, Bioorganic and Medicinal Chemistry Letters, vol.25, issue.6, pp.1206-1211, 2015.

Z. V. Chirkova, M. V. Kabanova, S. I. Filimonov, I. G. Abramov, A. Petzer et al.,

K. Yu, An evaluation of synthetic indole derivatives as inhibitors of monoamine oxidase, Bioorganic & Medicinal Chemistry Letters, vol.26, issue.9, pp.2214-2219, 2016.

G. Clarke, S. Grenham, P. Scully, P. Fitzgerald, R. D. Moloney et al., The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner, Molecular Psychiatry, vol.18, issue.6, pp.666-673, 2013.

Y. Clément, A. M. Le-guisquet, P. Venault, G. Chapouthier, and C. Belzung, , 2009.

, Pharmacological alterations of anxious behaviour in mice depending on both strain and the behavioural situation, PLoS ONE, vol.4, issue.11, pp.7745-7745

B. A. Cowell, D. J. Evans, and S. M. Fleiszig, Actin cytoskeleton disruption by ExoY and its effects on Pseudomonas aeruginosa invasion, FEMS Microbiology Letters, vol.250, issue.1, pp.71-76, 2005.

J. F. Cryan and T. G. Dinan, Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour, Nature Reviews Neuroscience, vol.13, issue.10, pp.701-712, 2012.

M. Crumeyrolle-arias, A. Medvedev, A. Cardona, D. Barritault, and V. Glover, In situ imaging of specific binding of [3 H] isatin in rat brain, Journal of Neurochemistry, vol.84, issue.3, pp.618-620, 2003.

M. Crumeyrolle-arias, M. C. Tournaire, A. Cane, J. M. Launay, and D. Barritault,

A. Medvedev, Inhibition of brain mitochondrial monoamine oxidases by the endogenous compound 5-hydroxyoxindole, Biochemical Pharmacology, vol.67, issue.5, pp.977-979, 2004.

M. Crumeyrolle-arias, O. Buneeva, V. Zgoda, A. Kopylov, A. Cardona et al.,

U. P. Vi, Isatin Binding Proteins in Rat Brain : In Situ Imaging , Quantitative Characterization of Specific [3 H] Isatin Binding, and Proteomic Profiling, Journal of Neuroscience Research, vol.87, issue.12, pp.2763-2772, 2009.

M. Crumeyrolle-arias, M. Jaglin, A. Bruneau, S. Vancassel, A. Cardona et al.,

S. Rabot, Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats, Psychoneuroendocrinology, vol.42, pp.207-217, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204296

D. L. Daubert, M. Mccowan, B. Erdos, and D. A. Scheuer, Nucleus of the solitary tract catecholaminergic neurons modulate the cardiovascular response to psychological stress in rats, The Journal of Physiology, vol.590, issue.19, pp.4881-4895, 2012.

H. E. Day, E. M. Kryskow, T. J. Nyhuis, L. Herlihy, and S. Campeau, , 2008.

, Conditioned fear inhibits c-fos mRNA expression in the central extended amygdala, Brain Research, vol.1229, issue.1, pp.137-146

M. De-angelis, M. Piccolo, L. Vannini, S. Siragusa, A. De-giacomo et al., Fecal Microbiota and Metabolome of Children with Autism and Pervasive Developmental Disorder Not Otherwise Specified, PLoS ONE, vol.8, issue.10, pp.76993-76993, 2013.

M. Deeley and C. Yanofsky, Nucleotide sequence of the structural gene for Tryptophanase of Escherichia coli K-12, Journal of Bacteriology, vol.147, issue.3, pp.787-796, 1981.

T. Deguchi, M. Nakamura, Y. Tsutsumi, A. Suenaga, and M. Otagiri, , 2003.

, Pharmacokinetics and Tissue Distribution of Uraemic Indoxyl Sulphate in Rats

, Biopharmaceutics & Drug Disposition, vol.24, pp.345-355

L. Di, The role of drug metabolizing enzymes in clearance, Expert Opinion on Drug Metabolism & Toxicology, vol.10, issue.3, pp.379-393, 2014.

R. Diaz, S. Wang, F. Anuar, Y. Qian, B. Björkholm et al., Normal gut microbiota modulates brain development and behavior, Proceedings of the National Academy of Sciences, vol.108, issue.7, pp.3047-3052, 2011.

J. Domka, J. Lee, and T. K. Wood, YliH (BssR) and YceP (BssS) Regulate Escherichia coli K-12 Biofilm Formation by Influencing Cell Signaling, Applied and Environmental Microbiology, vol.72, issue.4, pp.2449-2459, 2006.

M. Dragunow and R. Faull, The use of c-fos as a metabolic marker in neuronal pathway tracing, Journal of Neuroscience Methods, vol.29, issue.3, pp.261-265, 1989.

F. Escudié, L. Auer, M. Bernard, M. Mariadassou, L. Cauquil et al., FROGS: Find, Rapidly, OTUs with Galaxy Solution, Bioinformatics, vol.34, issue.8, pp.1287-1294, 2018.

P. Evenepoel, D. Claus, B. Geypens, M. Hiele, K. Geboes et al., Amount and fate of egg protein escaping assimilation in the small intestine of humans, The American Journal of Physiology, vol.277, issue.5, pp.935-943, 1999.

K. Franklin and G. Paxinos, The Mouse Brain in Stereotaxic Coordinates, 2008.

E. Edition, ;. T. Amsterdam, K. Shinohara, Y. Kishimoto, and A. Terada, Effect of miso soup containing Natto on the composition and metabolic activity of the human faecal flora, 2006.

, Microbial Ecology in Health and Disease, vol.18, issue.2, pp.79-84

G. Gareau, E. Wine, D. M. Rodrigues, J. H. Cho, M. T. Whary et al.,

P. M. Sherman, Bacterial infection causes stress-induced memory dysfunction in mice, Gut, vol.60, issue.3, pp.307-317, 2010.

E. M. Gillam, L. M. Notley, H. Cai, J. J. De-voss, and F. P. Guengerich, , 2000.

, Oxidation of indole by cytochrome P450 enzymes, Biochemistry, vol.39, issue.45, pp.13817-13824

V. Glover, J. M. Halket, P. J. Watkins, A. Clow, B. L. Goodwin et al., , 1988.

, Isatin: Identity with the Purified Endogenous Monoamine Oxidase Inhibitor Tribulin, Journal of Neurochemistry, vol.51, issue.2, pp.656-659

J. J. Godon, E. Zumstein, P. Dabert, F. Habouzit, and R. Moletta, Molecular microbial diversity of an anaerobic digestor as determined by small-subunit rDNA sequence analysis, Applied and Environmental Microbiology, vol.63, issue.7, pp.2802-2813, 1997.

G. Griebel, C. Belzung, G. Perrault, and D. J. Sanger, Differences in anxiety related behaviors and in sensitivity to diazepam in inbred and outbred strains of mice, 2000.

, Psychopharmacology, vol.148, issue.2, pp.164-170

E. Guyot, A. Chevallier, R. Barouki, and X. Coumoul, The AhR twist: Liganddependent AhR signaling and pharmaco-toxicological implications, Drug Discovery Today, vol.18, issue.9, pp.479-486, 2013.

N. Hamaue, N. Yamazaki, M. Terado, M. Minami, K. Ohno et al., Urinary isatin concentrations in patients with Parkinson's disease determined by a newly developed HPLC-UV method, Research communications in molecular pathology and pharmacology, vol.108, issue.1-2, p.11758976, 2000.

F. C. Happold and L. Hoyle, Tryptophane ans some indole derivatives, Biochemical Journal, vol.29, issue.8, p.16745862, 1935.

M. N. Hill, K. G. Hellemans, P. Verma, B. B. Gorzalka, and P. Sciences, , 2012.

, Neurobiology of chronic mild stress: Parallels to major depression, Neuroscience and Biobehavioral Reviews, vol.36, issue.9, pp.2085-2117

A. Högberg-raibaud, O. Raibaud, and M. E. Goldberg, Kinetic and equilibrium studies on the activation of Escherichia coli K12 tryptophanase by pyridoxal 5'-phosphate and monovalent cations, Journal of Biological Chemistry, vol.250, issue.9, p.1091651, 1975.

E. Y. Hsiao, S. W. Mcbride, S. Hsien, G. Sharon, E. R. Hyde et al., Microbiota Modulate Behavioral and Physiological Abnormalities Associated with Neurodevelopmental Disorders, Cell, vol.155, issue.7, pp.1451-1463, 2013.

T. D. Hubbard, I. A. Murray, W. H. Bisson, T. S. Lahoti, K. Gowda et al.,

G. H. Perdew, Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles, /srep12689 Human Metabolome Database : HMDB. (n.d.). Retrieved from, vol.5, pp.1-13, 2015.

A. Ishikado, T. Sato, and T. Mitsuoka, Suppressive effects of lactulose and magnesium oxide supplementation on fecal putrefactive metabolites with shortening gastrointestinal transit time, Microbial Ecology in Health and Disease, vol.19, issue.3, pp.184-190, 2007.

J. A. Izquierdo and A. O. Stoppani, Inhibition of smooth muscle contractility by indole and some indole compounds, British Journal of Pharmacology, vol.8, issue.4, pp.389-394, 1953.

M. Jaglin, M. Rhimi, C. Philippe, N. Pons, A. Bruneau et al., , 2018.

, Indole, a signaling molecule produced by the gut microbiota, negatively impacts emotional behaviors in rats, Frontiers in Neuroscience, vol.12, pp.1-19

H. Jiang, Z. Ling, Y. Zhang, H. Mao, Z. Ma et al., Altered fecal microbiota composition in patients with major depressive disorder, Brain, Behavior and Immunity, vol.48, pp.186-194, 2015.

D. A. Karlin, A. J. Mastromarino, R. D. Jones, J. R. Stroehlein, and O. Lorentz, , 1985.

, Fecal skatole and indole and breath methane and hydrogen in patients with large bowel polyps or cancer, Journal of Cancer Research and Clinical Oncology, vol.109, issue.2, pp.135-141

K. Kawamura-sato, K. Shibayama, T. Horii, Y. Iimuma, Y. Arakawa et al., , 1999.

, Role of multiple efflux pumps in Escherichia coli in indole expulsion, FEMS Microbiology Letters, vol.179, issue.2, pp.345-352

J. R. Kelly, Y. Borre, C. O. Brien, E. Patterson, S. El et al., , 2016.

, Transferring the blues : Depression-associated gut microbiota induces neurobehavioural changes in the rat, Journal of Psychiatric Research, vol.82, pp.109-118

L. J. King, D. V. Parke, and R. T. Williams, The metabolism of [2-14C] indole in the rat, The Biochemical Journal, vol.98, issue.1, p.5328168, 1966.

A. M. Krstulovic and C. Matzura, Rapid analysis of tryptophan metabolites using reversedphase high-performance liquid chromatography with fluorometric detection, Journal of Chromatography, vol.163, issue.1, p.528629, 1979.

S. Y. Ku, P. Yip, and P. L. Howell, Structure of Escherichia coli tryptophanase, Acta Crystallographica, vol.62, issue.7, pp.814-823, 2006.

B. Lamas, J. M. Natividad, and H. Sokol, Aryl hydrocarbon receptor and intestinal immunity, Mucosal Immunology, vol.11, issue.4, pp.1024-1038, 2018.

J. H. Lee and J. Lee, Indole as an intercellular signal in microbial communities, 2010.

, FEMS Microbiology Reviews, vol.34, issue.4, pp.426-444

J. H. Lee, H. S. Cho, Y. Kim, J. A. Kim, S. Banskota et al., , 2013.

, Indole and 7-benzyloxyindole attenuate the virulence of Staphylococcus aureus, Applied Microbiology and Biotechnology, vol.97, issue.10, pp.4543-4552

J. Lee, T. K. Wood, and J. Lee, Roles of Indole as an Interspecies and Interkingdom Signaling Molecule, Trends in Microbiology, vol.23, issue.11, pp.707-718, 2015.

P. Lepage, M. C. Leclerc, M. Joossens, S. Mondot, H. M. Blottière et al., A metagenomic insight into our gut's microbiome, Gut, vol.62, issue.1, pp.146-158, 2012.

W. Liu, H. Chuang, Y. Huang, C. Wu, G. Chou et al., Alteration of behavior and monoamine levels attributable to Lactobacillus plantarum PS128 in germfree mice, Behavioural Brain Research, vol.298, pp.202-209, 2016.

A. Macierzanka, F. Böttger, L. Lansonneur, R. Groizard, A. S. Jean et al.,

A. R. Mackie, The effect of gel structure on the kinetics of simulated gastrointestinal digestion of bovine ?-lactoglobulin, Food Chemistry, vol.134, issue.4, pp.2156-2163, 2012.

G. Mannaioni, R. Carpenedo, A. M. Pugliese, and R. Corradetti, Electrophysiological studies on oxindole, a neurodepressant tryptophan metabolite, British Journal of Pharmacology, vol.125, issue.8, pp.1751-1760, 1998.

G. Mannaioni, R. Carpenedo, R. Corradetti, V. Carla, I. Venturini et al.,

M. L. Zeneroli, Tryptophan metabolism and hepatic encephalopathy : Studies on the Sedative Properties of Oxindole, Tryptophan, Serotonin, and Melatonin -Advances in Experimental Medicine and Biology, vol.467, pp.155-167, 1999.

P. Marteau and J. Doré, Le microbiote intestinal, un organe à part entière, John Libbey Eurotext, 2017.

M. Matsumoto, R. Kibe, T. Ooga, Y. Aiba, S. Kurihara et al., Impact of Intestinal Microbiota on Intestinal Luminal Metabolome, Scientific Reports, vol.2, pp.1-10, 2011.

R. Mayerhofer, E. E. Fröhlich, F. Reichmann, A. Farzi, N. Kogelnik et al.,

P. Holzer, Diverse action of lipoteichoic acid and lipopolysaccharide on neuroinflammation, blood-brain barrier disruption, and anxiety in mice, Brain Behavior and Immunity, vol.60, pp.174-187, 2017.

J. G. Mccall, R. Al-hasani, E. R. Siuda, D. Y. Hong, A. J. Norris et al.,

M. R. , CRH engagement of the locus coeruleus noradrenergic system mediates stressinduced anxiety, Neuron, vol.87, issue.3, pp.605-620, 2015.

I. M. Mcintyre and T. R. Norman, Serotonergic effects of isatin: an endogenous MAO inhibitor related to tribulin, Journal of Neural Transmission/ General Section JNT, vol.79, issue.1-2, pp.35-40, 1990.

A. E. Medvedev and V. Glover, Tribulin and Endogenous MAO-Inhibitory Regulation In Vivo, NeuroToxicology, vol.25, issue.1-2, pp.185-192, 2004.

A. Medvedev, N. Igosheva, M. Crumeyrolle-arias, and V. Glover, Isatin: Role in stress and anxiety, Stress, vol.8, issue.3, pp.175-183, 2005.

M. Messaoudi, R. Lalonde, N. Violle, H. Javelot, D. Desor et al., Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects, British Journal of Nutrition, vol.105, issue.5, pp.755-764, 2011.

A. A. Mika, M. Gaffney, R. Roller, A. Hills, C. A. Bouchet et al.,

M. Fleshner, Neuroscience Letters, vol.677, pp.103-109, 2018.

M. Minami, N. Hamaue, M. Hirafuji, H. Saito, T. Hiroshige et al.,

. Isatin and . Inhibitor, and a rat model of Parkinson's disease induced by the Japanese encephalitis virus, Oxidative Stress and Neuroprotection -Journal of Neural Transmission, vol.71, pp.87-95

M. Minekus, M. Alminger, P. Alvito, S. Ballance, T. Bohn et al., A standardised static in vitro digestion method suitable for food-an international consensus, Food and Function, vol.5, issue.6, pp.1113-1124, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01195466

E. Mishima, S. Fukuda, C. Mukawa, A. Yuri, Y. Kanemitsu et al., Evaluation of the impact of gut microbiota on uremic solute accumulation by a CE-TOFMS-based metabolomics approach, Kidney International, vol.92, issue.3, pp.634-645, 2017.

J. E. Morley, S. A. Farr, and J. F. Flood, Isatin inhibits food intake in mice, European Journal of Pharmacology, vol.305, issue.1-3, pp.23-29, 1996.

A. Moya-pérez, A. Perez-villalba, A. Benítez-páez, I. Campillo, and Y. Sanz, , 2017.

, Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice, Brain, Behavior, and Immunity, vol.65, pp.43-56

B. Myers and B. Greenwood-van-meerveld, Role of anxiety in the pathophysiology of irritable bowel syndrome: Importance of the amygdala, Frontiers in Neuroscience, vol.3, pp.1-10, 2009.

A. Naseribafrouei, K. Hestad, E. Avershina, M. Sekelja, A. Linløkken et al., Correlation between the human fecal microbiota and depression, 2014.

, Neurogastroenterology & Motility, vol.26, issue.8, pp.1155-1162

J. M. Natividad, A. Agus, J. Planchais, B. Lamas, A. C. Jarry et al.,

, Impaired Aryl Hydrocarbon Receptor Ligand Production by the Gut Microbiota Is a Key Factor in Metabolic Syndrome, Cell Metabolism, vol.28, issue.5, pp.737-749

K. M. Neufeld, S. M. Mahony, A. E. Hoban, V. Waworuntu, B. M. Berg et al., Neurobehavioural effects of Lactobacillus rhamnosus GG alone and in combination with prebiotics polydextrose and galactooligosaccharide in male rats exposed to early-life stress, Nutritional Neuroscience, 2017.

M. Neunlist, L. Van-landeghem, M. M. Mahé, P. Derkinderen, and S. B. Des-varannes,

M. Rolli-derkinderen, The digestive neuronal-glial-epithelial unit: A new actor in gut health and disease, Nature Reviews Gastroenterology and Hepatology, vol.10, issue.2, pp.90-100, 2012.

J. K. Nicholson, E. Holmes, and I. D. Wilson, Gut microorganisms, mammalian metabolism and personalized health care, Nature Reviews Microbiology, vol.3, issue.5, pp.431-438, 2005.
DOI : 10.1038/nrmicro1152

B. Nielsen, N. Jérôme, A. Saint-albin, F. Joly, S. Rabot et al., Sexual responses of male rats to odours from female rats in oestrus are not affected by female germfree status, Behavioural Brain Research, vol.359, pp.686-693, 2018.

R. Nishino, K. Mikami, H. Takahashi, S. Tomonaga, M. Furuse et al., Commensal microbiota modulate murine behaviors in a strictly contamination-free environment confirmed by culture-based methods, Neurogastroenterology & Motility, vol.25, issue.6, pp.521-371, 2013.
DOI : 10.1111/nmo.12110

J. E. Norton, G. A. Wallis, F. Spyropoulos, P. J. Lillford, and I. T. Norton, , 2014.

, Designing Food Structures for Nutrition and Health Benefits, Annual Review of Food Science and Technology, vol.5, issue.1, pp.177-195

S. Oh, G. W. Go, E. Mylonakis, and Y. Kim, The bacterial signalling molecule indole attenuates the virulence of the fungal pathogen Candida albicans, Journal of Applied Microbiology, vol.113, issue.3, pp.622-628, 2012.

A. Ørgaard and J. J. Holst, The role of somatostatin in GLP-1-induced inhibition of glucagon secretion in mice, Diabetologia, vol.60, issue.9, pp.1731-1739, 2017.

G. Palma, . De, M. D. Lynch, J. Lu, V. T. Dang et al., , 2017.

, Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice, Science Translational Medicine, vol.9, issue.379, pp.1-14

S. Parois, O. Zemb, and A. Prunier, Influence des conditions de logement sur la production et le stockage du scatol et de l'indole chez le porc mâle entier, vol.49, pp.163-168, 2017.

S. Piñero-fernandez, C. Chimerel, U. F. Keyser, and D. K. Summers, Indole transport across Escherichia coli membranes, Journal of Bacteriology, vol.193, issue.8, pp.1793-1798, 2011.

M. I. Pinto-sanchez, G. B. Hall, K. Ghajar, A. Nardelli, C. Bolino et al., Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome, 2017.

, Gastroenterology, vol.153, issue.2, pp.448-459

K. E. Price, Aminoglycoside research 1975-1985: Prospects for development of improved agents, Antimicrobial Agents and Chemotherapy, vol.29, issue.4, pp.543-548, 1986.

O. Riggio, G. Mannaioni, L. Ridola, S. Angeloni, M. Merli et al., Peripheral and splanchnic indole and oxindole levels in cirrhotic patients: A study on the pathophysiology of hepatic encephalopathy, American Journal of Gastroenterology, vol.105, issue.6, pp.1374-1381, 2010.

V. Rothhammer, I. D. Mascanfroni, L. Bunse, M. C. Takenaka, E. Jessica et al.,

F. J. Quintana, Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and CNS inflammation via the aryl hydrocarbon receptor, Nature Medicine, vol.22, issue.6, pp.586-597, 2016.

M. Sakai, K. Tohyama, and M. Mutai, Effect of indole on adenylate energy charge and mitochondrial phosphorylative activity of rat liver, The International Journal of Biochemistry, vol.14, issue.7, pp.569-572, 1982.

M. Sandler, A. Przyborowska, J. Halket, P. Watkins, V. Glover et al., , 1991.

, Urinary but Not Brain Isatin Levels Are Reduced in Germ-Free Rats, Journal of Neurochemistry, vol.57, issue.3, pp.1074-1075

A. Sarrieau, F. Chaouloff, V. Lemaire, and P. Morme`de, Comparison of the neuroendocrine responses to stress in outbred inbred and F1 hybrid rats, Life Sciences, vol.63, issue.2, pp.87-96, 1998.

H. M. Savignac, Y. Couch, M. Stratford, D. M. Bannerman, G. Tzortzis et al., Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-? levels in male mice, Brain, Behavior and Immunity, vol.52, pp.120-131, 2016.

J. R. Schank, L. C. Liles, and D. Weinshenker, Norepinephrine Signaling Through ?-Adrenergic Receptors is Critical for Expression of Cocaine-Induced Anxiety, Biological Psychiatry, vol.63, issue.11, pp.1007-1012, 2008.

R. Sender, S. Fuchs, and R. Milo, Revised Estimates for the Number of Human and Bacteria Cells in the Body, PLoS Biology, vol.14, issue.8, pp.1-14, 2016.

M. Senzacqua, I. Severi, J. Perugini, S. Acciarini, S. Cinti et al., Action of administered ciliary neurotrophic factor on the mouse dorsal vagal complex, Frontiers in Neuroscience, vol.10, pp.1-17, 2016.

Y. Shimada, M. Kinoshita, K. Harada, M. Mizutani, K. Masahata et al., Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon, PLoS ONE, vol.8, issue.11, pp.1-10, 2013.

R. Sonowal, A. Swimm, A. Sahoo, L. Luo, Y. Matsunaga et al., , 2017.

, Proceedings of the National Academy of Sciences, vol.114, pp.7506-7515

N. St?nciuc, I. Van-plancken, . Der, G. Rotaru, and M. Hendrickx, Denaturation impact in susceptibility of beta-lactoglobulin to enzymatic hydrolysis: A kinetic study, Revue Roumaine de Chimie, vol.53, issue.10, pp.921-929, 2008.

N. Sudo, Y. Chida, Y. Aiba, J. Sonoda, N. Oyama et al., , 2004.

, Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice, The Journal of Physiology, vol.558, issue.1, pp.263-275

Y. B. Tewari and R. N. Goldberg, An equilibrium and calorimetric investigation of the hydrolysis of L-tryptophan to (indole+pyruvate+ammonia), Journal of Solution Chemistry, vol.23, issue.2, pp.167-184, 1994.

F. Tronche, C. Kellendonk, O. Kretz, P. Gass, K. Anlag et al., Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety, Nature Genetics, vol.23, issue.1, pp.99-103, 1999.

Q. Wang, G. M. Garrity, J. M. Tiedje, and J. R. Cole, Naïve Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy, Applied and Environmental Microbiology, vol.73, issue.16, pp.5261-5267, 2007.

P. Watkins, A. Clow, V. Glover, J. Halket, A. Przyborowska et al., , 1990.

. Isatin, regional distribution in rat brain and tissues, Neurochemistry International, vol.17, issue.2, pp.321-323

D. D. Whitt and R. D. Demoss, Effect of microflora on the free amino acid distribution in various regions of the mouse gastrointestinal tract, Applied and Environmental Microbiology, vol.30, issue.4, pp.609-615, 1975.

W. R. Wikoff, A. T. Anfora, J. Liu, P. G. Schultz, S. Lesley et al., Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites, Proceedings of the National Academy of Sciences, vol.106, issue.10, pp.3698-3703, 2009.

K. Windey, V. De-preter, T. Louat, F. Schuit, J. Herman et al., Modulation of Protein Fermentation Does Not Affect Fecal Water Toxicity: A Randomized Cross-Over Study in Healthy Subjects, PLoS ONE, issue.12, p.52387, 2012.

C. Yanofsky, V. Horn, and P. Gollnick, Physiological studies of tryptophan transport and tryptophanase operon induction in Escherichia coli, Journal of Bacteriology, vol.173, issue.19, pp.6009-6017, 1991.

C. Yanofsky, RNA-based regulation of genes of tryptophan synthesis and degradation, in bacteria, RNA, vol.13, issue.8, pp.1141-1154, 2007.

H. Yang, L. Wang, S. V. Wu, J. Tay, M. Goulet et al., Peripheral secretin-induced Fos expression in the rat brain is largely vagal dependent, Neuroscience, vol.128, issue.1, pp.131-141, 2004.

T. Yatsunenko, F. E. Rey, M. J. Manary, I. Trehan, M. G. Dominguez-bello et al., Human gut microbiome viewed across age and geography, Nature, vol.486, issue.7402, pp.222-227, 2012.
DOI : 10.1038/nature11053

URL : http://europepmc.org/articles/pmc3376388?pdf=render

A. Yuwiler, The effect of isatin (tribulin) on metabolism of indoles in the rat brain and pineal: In vitro and in vivo studies, Neurochemical Research, vol.15, issue.1, pp.95-100, 1990.

T. Zelante, R. G. Iannitti, C. Cunha, A. Deluca, G. Giovannini et al.,

L. Romani, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, vol.39, issue.2, pp.372-385, 2013.

L. S. Zhang and S. S. Davies, Microbial metabolism of dietary components to bioactive metabolites: Opportunities for new therapeutic interventions, Genome Medicine, vol.8, issue.1, pp.1-18, 2016.

P. Zheng, B. Zeng, C. Zhou, M. Liu, Z. Fang et al., Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host ' s metabolism, pp.1-11, 2016.

Y. Zhou, Z. Zhao, and J. Xie, Effects of isatin on rotational behavior and DA levels in caudate putamen in Parkinsonian rats, Brain Research, vol.917, issue.1, pp.127-132, 2001.

S. Zhou, Y. Lu, H. Yao, and C. Owyang, Spatial organization of neurons in the dorsal motor nucleus of the vagus synapsing with intragastric cholinergic and nitric oxide/VIP neurons in the rat, American Journal of Physiology -Gastrointestinal and Liver Physiology, vol.294, issue.5, pp.1201-1209, 2008.

E. Zuccato, M. Venturi, G. Di-leo, L. Colombo, C. Bertolo et al., Role of bile acids and metabolic activity of colonic bacteria in increased risk of colon cancer after cholecystectomy, Digestive Diseases and Sciences, vol.38, issue.3, pp.514-519, 1993.