O. N. Ilinskaya, V. V. Ulyanova, D. R. Yarullina, and I. G. Gataullin, Secretome of Intestinal Bacilli: A Natural Guard against Pathologies, Front Microbiol, vol.8, 2017.

K. Bene, Z. Varga, V. O. Petrov, N. Boyko, and E. Rajnavolgyi, Gut Microbiota Species Can Provoke both Inflammatory and Tolerogenic Immune Responses in Human Dendritic Cells Mediated by Retinoic Acid Receptor Alpha Ligation, Front Immunol, vol.8, 2017.

H. Zhang, W. Li, and D. Xu, Mucosa-reparing and microbiota-balancing therapeutic effect of Bacillus subtilis alleviates dextrate sulfate sodium-induced ulcerative colitis in mice, Exp Ther Med, vol.12, issue.4, pp.2554-2562, 2016.

S. Miquel, R. Martín, and O. Rossi, Faecalibacterium prausnitzii and human intestinal health, Current Opinion in Microbiology, vol.16, issue.3, pp.255-261, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00842645

, Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients | PNAS, 2019.

R. J. Newton, S. L. Mclellan, and D. K. Dila, Sewage Reflects the Microbiomes of Human Populations, mBio, vol.6, issue.2, 2015.

M. Demirci, H. B. Tokman, and H. K. Uysal, Reduced Akkermansia muciniphila and Faecalibacterium prausnitzii levels in the gut microbiota of children with allergic asthma, Allergol Immunopathol (Madr), vol.47, issue.4, pp.365-371, 2019.

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

R. E. Ley, Obesity and the human microbiome, Curr Opin Gastroenterol, vol.26, issue.1, pp.5-11, 2010.

. Microbiologie-orale, Wikipédia. ; 2019. Accessed, 2019.

R. Nagpal, H. Tsuji, and T. Takahashi, Ontogenesis of the Gut Microbiota Composition in Healthy, Full-Term, Vaginally Born and Breast-Fed Infants over the First 3 Years of Life: A Quantitative Bird's-Eye View, Front Microbiol, vol.8, 2017.

. Campeotto, Mise en place de la flore intestinale du nouveau-n.pdf. Accessed, 2007.

E. Westerbeek, A. Van-den-berg, H. N. Lafeber, J. Knol, W. Fetter et al., The intestinal bacterial colonisation in preterm infants: A review of the literature, Clinical Nutrition, vol.25, issue.3, pp.361-368, 2006.

, Tannock et Fuller -Plasmid Profiling of Members of the Family Enterob.pdf. Accessed, 2019.

M. M. Grönlund, O. P. Lehtonen, E. Eerola, and P. Kero, Fecal microflora in healthy infants born by different methods of delivery: permanent changes in intestinal flora after cesarean delivery, J Pediatr Gastroenterol Nutr, vol.28, issue.1, pp.19-25, 1999.

M. N. Guediche, . Colonisation, N. Du, and . Infection,

. Moreau-m-c, . Thomasson-m, . Ducluzeau-r, P. Raibaud, and . Muller-m-c, Cinétique d'établissement de la microflore digestive chez le nouveau-né humain en fonction de la nature du lait, Reproduction Nutrition Développement, vol.26, issue.2B, pp.745-753, 1986.

, Intestinal colonization with Enterobacteriaceae in Pakistani and Swedish hospitaldelivered infants, 2019.

, Favard -Impact du microbiote intestinal sur le stress et l.pdf

, The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, 2019.

D. Vadder, F. Kovatcheva-datchary, P. Goncalves, and D. , Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits, Cell, vol.156, issue.1-2, pp.84-96, 2014.

, Microbiota-activated PPAR-? signaling inhibits dysbiotic Enterobacteriaceae expansion | Science, 2019.

T. Jung, J. H. Park, W. Jeon, and K. Han, Butyrate modulates bacterial adherence on LS174T human colorectal cells by stimulating mucin secretion

K. Oliphant and E. Allen-vercoe, Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health, Microbiome, vol.7, issue.1, p.91, 2019.

S. A. Gibson, C. Mcfarlan, S. Hay, and G. T. Macfarlane, Significance of microflora in proteolysis in the colon, Appl Environ Microbiol, vol.55, issue.3, pp.679-683, 1989.

G. T. Macfarlane, J. H. Cummings, and C. Allison, Protein Degradation by Human Intestinal Bacteria, Microbiology, vol.132, issue.6, pp.1647-1656, 1986.

B. E. Gustafsson, F. S. Daft, E. G. Mcdaniel, J. C. Smith, and R. J. Fitzgerald, Effects of vitamin Kactive compounds and intestinal microorganisms in vitamin K-deficient germfree rats, J Nutr, vol.78, issue.4, pp.461-468, 1962.

I. Rowland, G. Gibson, and A. Heinken, Gut microbiota functions: metabolism of nutrients and other food components, Eur J Nutr, vol.57, issue.1, pp.1-24, 2018.

C. V. Srikanth and B. A. Mccormick, Interactions of the Intestinal Epithelium with the Pathogen and the Indigenous Microbiota: A Three-Way Crosstalk, Interdiscip Perspect Infect Dis, 2008.

S. M. Jandhyala, Role of the normal gut microbiota, World Journal of Gastroenterology, vol.21, issue.29, p.8787, 2015.

C. R. Interets, . Microbiote, and . Et-probiotiques, , p.79, 2014.

P. D. Cani and W. M. De-vos, Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila, Front Microbiol, vol.8, 2017.

T. Yamanaka, L. Helgeland, I. N. Farstad, H. Fukushima, T. Midtvedt et al., Microbial colonization drives lymphocyte accumulation and differentiation in the follicle-associated epithelium of Peyer's patches, J Immunol, vol.170, issue.2, pp.816-822, 2003.

L. Bry, P. G. Falk, T. Midtvedt, and J. I. Gordon, A model of host-microbial interactions in an open mammalian ecosystem, Science, vol.273, issue.5280, pp.1380-1383, 1996.

S. Vaishnava, C. L. Behrendt, A. S. Ismail, L. Eckmann, and L. V. Hooper, Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface, Proceedings of the National Academy of Sciences, vol.105, issue.52, pp.20858-20863, 2008.

M. Carabotti, A. Scirocco, M. A. Maselli, and C. Severi, The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems, Ann Gastroenterol, vol.28, issue.2, pp.203-209, 2015.

G. Clarke, S. Grenham, and P. Scully, 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.

R. Diaz-heijtz, S. Wang, and F. Anuar, Normal gut microbiota modulates brain development and behavior, Proc Natl Acad Sci, vol.108, issue.7, pp.3047-3052, 2011.

G. D. Abrams and J. E. Bishop, Effect of the normal microbial flora on gastrointestin

H. Iwai, Y. Ishihara, J. Yamanaka, and T. Ito, Effects of bacterial flora on cecal size and transit rate of intestinal contents in mice, Jpn J Exp Med, vol.43, issue.4, pp.297-305, 1973.

J. A. Hawrelak, The Causes of Intestinal Dysbiosis: A Review, vol.9, p.18, 2004.

C. D. Filippo, D. Cavalieri, and M. D. Paola, Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, PNAS, vol.107, issue.33, pp.14691-14696, 2010.

J. Zimmer, B. Lange, and J. Frick, A vegan or vegetarian diet substantially alters the human colonic faecal microbiota, Eur J Clin Nutr, vol.66, issue.1, pp.53-60, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00665871

D. Palma, G. Nadal, I. Collado, M. C. Sanz, and Y. , Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects, Br J Nutr, vol.102, issue.8, pp.1154-1160, 2009.

G. T. Macfarlane and S. Macfarlane, Bacteria, colonic fermentation, and gastrointestinal health, J AOAC Int, vol.95, issue.1, pp.50-60, 2012.

A. Kilkkinen, P. Pietinen, T. Klaukka, J. Virtamo, P. Korhonen et al., Use of oral antimicrobials decreases serum enterolactone concentration, Am J Epidemiol, vol.155, issue.5, pp.472-477, 2002.

, The Role of the Microbiome in Asthma: The Gut-Lung Axis, 2019.

G. Major and R. Spiller, Irritable bowel syndrome, inflammatory bowel disease and the microbiome, Curr Opin Endocrinol Diabetes Obes, vol.21, issue.1, pp.15-21, 2014.

I. A. Harsch and P. C. Konturek, The Role of Gut Microbiota in Obesity and Type 2 and Type 1 Diabetes Mellitus: New Insights into "Old, Diseases. Med Sci (Basel), vol.6, issue.2, 2018.

A. M. Caricilli and M. Saad, The Role of Gut Microbiota on Insulin Resistance, Nutrients, vol.5, issue.3, pp.829-851, 2013.

. Netgen, Microbiote intestinal, obésité et résistance à l'insuline. Revue Médicale Suisse, 2019.

M. Arrieta, L. T. Stiemsma, and P. A. Dimitriu, Early infancy microbial and metabolic alterations affect risk of childhood asthma, Sci Transl Med, vol.7, issue.307, pp.307-152, 2015.

, Flore intestinale et asthme -Rôle du microbiote intestinal dans le développement de l'asthme allergique. OPA Pratique, vol.11, 2018.

A. Pianta, S. Arvikar, and K. Strle, Evidence for Immune Relevance of Prevotella copri, a Gut Microbe, in Patients with Rheumatoid Arthritis, Arthritis Rheumatol, vol.69, issue.5, pp.964-975, 2017.

, Polyarthrite rhumatoïde : une auto-immunité liée à des bactéries intestinales. Le Quotidien du médecin, 2020.

Y. Maeda, T. Kurakawa, and E. Umemoto, Dysbiosis Contributes to Arthritis Development via Activation of Autoreactive T Cells in the Intestine, Arthritis & Rheumatology, vol.68, issue.11, pp.2646-2661, 2016.

G. Horta-baas, S. Romero-figueroa-m-del, A. J. Montiel-jarquín, M. L. Pizano-zárate, J. García-mena et al., Intestinal Dysbiosis and Rheumatoid Arthritis: A Link between Gut Microbiota and the Pathogenesis of Rheumatoid Arthritis, J Immunol Res, 2017.

, The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials, 2020.

D. Kim, Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metabolism and Disposition, vol.43, issue.10, pp.1581-1589, 2015.

K. Lavrijsen, D. Van-dyck, and J. Van-houdt, Reduction of the prodrug loperamide oxide to its active drug loperamide in the gut of rats, dogs, and humans, Drug Metab Dispos, vol.23, issue.3, pp.354-362, 1995.

, Reduction of Sulindac to its active metabolite, sulindac sulfide: assay and role of the methionine sulfoxide reductase system -ScienceDirect, 2020.

, The reduction of sulphinpyrazone and sulindac by intestinal bacteria, 2019.

M. A. Peppercorn and P. Goldman, The Role of Intestinal Bacteria in the Metabolism of Salicylazosulfapyridine, J Pharmacol Exp Ther, vol.181, issue.3, pp.555-562, 1972.

H. J. Lee, H. Zhang, D. A. Orlovich, and J. P. Fawcett, The influence of probiotic treatment on sulfasalazine metabolism in rat, Xenobiotica, vol.42, issue.8, pp.791-797, 2012.

A. Khan, A. K. Piris, J. Truelove, and S. C. , An experiment to determine the active therapeutic moiety of sulphasalazine, Lancet, vol.2, issue.8044, pp.892-895, 1977.

C. Deloménie, S. Fouix, and S. Longuemaux, Identification and Functional Characterization of Arylamine N-Acetyltransferases in Eubacteria: Evidence for Highly Selective Acetylation of 5-Aminosalicylic Acid, Journal of Bacteriology, vol.183, issue.11, pp.3417-3427, 2001.

B. J. Dull, K. Salata, and P. Goldman, Role of the intestinal flora in the acetylation of sulfasalazine metabolites, Biochemical Pharmacology, vol.36, issue.21, pp.3772-3774, 1987.

, Effects of orally administered antibiotics on the bioavailability of amlodipine: gut microbiota-mediated drug interaction, 2019.

S. Kitamura, K. Sugihara, M. Kuwasako, and K. Tatsumi, The Role of Mammalian Intestinal Bacteria in the Reductive Metabolism of Zonisamide, Journal of Pharmacy and Pharmacology, vol.49, issue.3, pp.253-256, 1997.

G. W. Elmer and R. P. Remmel, Role of the intestinal microflora in clonazepam metabolism in the rat, Xenobiotica, vol.14, issue.11, pp.829-840, 1984.

J. Fujii, N. Inotsume, and M. Nakano, Degradation of bromazepam by the intestinal microflora, Chem Pharm Bull, vol.35, issue.10, pp.4338-4341, 1987.

J. Fujii, N. Inotsume, and M. Nakano, Effect of food on the bioavailability of bromazepam following oral administration in healthy volunteers, J Pharmacobio-dyn, vol.13, issue.5, pp.269-271, 1990.

F. Rafii, J. B. Sutherland, E. B. Hansen, and C. E. Cerniglia, Reduction of nitrazepam by Clostridium leptum, a nitroreductase-producing bacterium isolated from the human intestinal tract, Clin Infect Dis, vol.25, issue.2, pp.121-122, 1997.

H. J. Haiser, K. L. Seim, E. P. Balskus, and P. J. Turnbaugh, Mechanistic insight into digoxin inactivation by Eggerthella lenta augments our understanding of its pharmacokinetics, Gut Microbes, vol.5, issue.2, pp.233-238, 2014.

J. R. Saha, V. P. Butler, H. C. Neu, and J. Lindenbaum, Digoxin-inactivating bacteria: identification in human gut flora, Science, vol.220, issue.4594, pp.325-327, 1983.

H. J. Haiser, D. B. Gootenberg, K. Chatman, G. Sirasani, E. P. Balskus et al., Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta, Science, vol.341, issue.6143, pp.295-298, 2013.

V. M. Rekdal, E. N. Bess, J. E. Bisanz, P. J. Turnbaugh, and E. P. Balskus, Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism, Science, vol.364, issue.6445, p.6323, 2019.

, The role of catecholamines, 5-hydroxytryptamine and m-tyramine in the behavioural effects of m-tyrosine in the rat. -PubMed -NCBI. Accessed October 9, 2019.

P. S. Mcquade and P. L. Wood, The effects of administration of meta-tyramine and paratyramine on dopamine and its metabolites in the rat striatum, Prog Neuropsychopharmacol Biol Psychiatry, vol.8, pp.705-709, 1984.

B. D. Wallace, H. Wang, and K. T. Lane, Alleviating Cancer Drug Toxicity by Inhibiting a Bacterial Enzyme, Science, vol.330, issue.6005, pp.831-835, 2010.

A. Loguidice, B. D. Wallace, L. Bendel, M. R. Redinbo, and U. A. Boelsterli, Pharmacologic Targeting of Bacterial ?-Glucuronidase Alleviates Nonsteroidal Anti-Inflammatory Drug-Induced Enteropathy in Mice, J Pharmacol Exp Ther, vol.341, issue.2, pp.447-454, 2012.

X. Liang, K. Bittinger, X. Li, D. R. Abernethy, F. D. Bushman et al., Bidirectional interactions between indomethacin and the murine intestinal microbiota, eLife, vol.4, p.8973, 2015.

K. S. Saitta, C. Zhang, K. K. Lee, K. Fujimoto, M. R. Redinbo et al., Bacterial ?glucuronidase inhibition protects mice against enteropathy induced by indomethacin, ketoprofen or diclofenac: mode of action and pharmacokinetics, Xenobiotica, vol.44, issue.1, pp.28-35, 2014.

C. Panebianco, A. Andriulli, and V. Pazienza, Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies, vol.6, pp.92-105, 2018.

F. Scaldaferri, V. Gerardi, and F. Mangiola, Role and mechanisms of action of Escherichia coli Nissle 1917 in the maintenance of remission in ulcerative colitis patients: An update, World J Gastroenterol, vol.22, issue.24, pp.5505-5511, 2016.

Z. Matuskova, E. Anzenbacherova, R. Vecera, H. Tlaskalova-hogenova, M. Kolar et al., Administration of a Probiotic Can Change Drug Pharmacokinetics: Effect of E. coli Nissle 1917 on Amidarone Absorption in Rats, PLoS One, vol.9, issue.2, 2014.

C. A. Jacobi and P. Malfertheiner, Escherichia coli Nissle 1917 (Mutaflor): new insights into an old probiotic bacterium, Dig Dis, vol.29, issue.6, pp.600-607, 2011.

L. Vee, M. Lecureur, V. Stieger, B. Fardel, and O. , Regulation of drug transporter expression in human hepatocytes exposed to the proinflammatory cytokines tumor necrosis factoralpha or interleukin-6, Drug Metab Dispos, vol.37, issue.3, pp.685-693, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00673249

A. Koenen, H. K. Kroemer, M. Grube, M. Zu-schwabedissen, and . He, Current understanding of hepatic and intestinal OATP-mediated drug-drug interactions, Expert Rev Clin Pharmacol, vol.4, issue.6, pp.729-742, 2011.

. Accessed, , 2019.

T. Selmer and P. I. Andrei, p-Hydroxyphenylacetate decarboxylase from Clostridium difficile. A novel glycyl radical enzyme catalysing the formation of p-cresol, Eur J Biochem, vol.268, issue.5, pp.1363-1372, 2001.

N. Gamage, A. Barnett, and N. Hempel, Human sulfotransferases and their role in chemical metabolism, Toxicol Sci, vol.90, issue.1, pp.5-22, 2006.

J. Kim, M. S. Choi, and J. Jeong, Effect of Probiotics on Pharmacokinetics of Orally Administered Acetaminophen in Mice, Drug Metab Dispos, vol.46, issue.2, pp.122-130, 2018.

H. Al-salami, G. Butt, J. P. Fawcett, I. G. Tucker, S. Golocorbin-kon et al., Probiotic treatment reduces blood glucose levels and increases systemic absorption of gliclazide in diabetic rats, Eur J Drug Metab Pharmacokinet, vol.33, issue.2, pp.101-106, 2008.

I. S. Kim, D. Yoo, and J. , Reduced metabolic activity of gut microbiota by antibiotics can potentiate the antithrombotic effect of aspirin, Biochem Pharmacol, vol.122, pp.72-79, 2016.

R. Kato, H. Yuasa, and K. Inoue, Effect of Lactobacillus casei on the Absorption of Nifedipine from Rat Small Intestine. Drug Metabolism and Pharmacokinetics, vol.22, pp.96-102, 2007.

U. M. Zanger and M. Schwab, Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation, Pharmacol Ther, vol.138, issue.1, pp.103-141, 2013.

B. Björkholm, C. M. Bok, A. Lundin, J. Rafter, M. L. Hibberd et al., Intestinal Microbiota Regulate Xenobiotic Metabolism in the Liver, PLoS One, vol.4, issue.9, 2009.

S. Viaud, F. Saccheri, and G. Mignot, The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide, Science, vol.342, issue.6161, pp.971-976, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01204279

B. Mao, D. Li, and J. Zhao, In vitro fermentation of lactulose by human gut bacteria, J Agric Food Chem, vol.62, issue.45, pp.10970-10977, 2014.

M. Rogers and D. M. Aronoff, The influence of non-steroidal anti-inflammatory drugs on the gut microbiome, Clinical Microbiology and Infection, vol.22, issue.2, pp.178-179, 2016.

, Resistance of germfree rats to indomethacin-induced intestinal lesions, 2020.

, Role of intestinal bacteria in ileal ulcer formation in rats treated with a nonsteroidal antiinflammatory drug, 2020.

T. Iwahi, H. Satoh, and M. Nakao, Lansoprazole, a novel benzimidazole proton pump inhibitor, and its related compounds have selective activity against Helicobacter pylori, Antimicrob Agents Chemother, vol.35, issue.3, pp.490-496, 1991.

A. F. Andersson, M. Lindberg, H. Jakobsson, F. Bäckhed, P. Nyrén et al., Comparative analysis of human gut microbiota by barcoded pyrosequencing, PLoS ONE, vol.3, issue.7, p.2836, 2008.

S. P. Pereira, N. Gainsborough, and R. H. Dowling, Drug-induced hypochlorhydria causes high duodenal bacterial counts in the elderly, Aliment Pharmacol Ther, vol.12, issue.1, pp.99-104, 1998.

J. Bures, J. Cyrany, and D. Kohoutova, Small intestinal bacterial overgrowth syndrome, World J Gastroenterol, vol.16, issue.24, pp.2978-2990, 2010.

C. Williams and K. Mccoll, Review article: proton pump inhibitors and bacterial overgrowth, Aliment Pharmacol Ther, vol.23, issue.1, pp.3-10, 2006.

K. R. Reveles, C. N. Ryan, L. Chan, R. A. Cosimi, and W. L. Haynes, Proton pump inhibitor use associated with changes in gut microbiota composition, Gut, vol.67, issue.7, pp.1369-1370, 2018.

, Metformine et microbiote intestinal : des rapports complexes. Accessed, 2019.

N. Larsen, F. K. Vogensen, and F. Van-den-berg, Gut Microbiota in Human Adults with Type 2 Diabetes Differs from Non-Diabetic Adults, PLoS One, vol.5, issue.2, 2010.

K. Forslund, F. Hildebrand, and T. Nielsen, Disentangling the effects of type 2 diabetes and metformin on the human gut microbiota, Nature, vol.528, issue.7581, pp.262-266, 2015.

A. Whang, R. Nagpal, and H. Yadav, Bi-directional drug-microbiome interactions of antidiabetics, EBioMedicine, vol.39, pp.591-602, 2019.

L. Bastard, Q. Al-ghalith, G. A. Grégoire, and M. , Systematic review: human gut dysbiosis induced by non-antibiotic prescription medications, Alimentary Pharmacology & Therapeutics, vol.47, issue.3, pp.332-345, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02127919

H. Wu, E. Esteve, and V. Tremaroli, Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug, Nat Med, vol.23, issue.7, pp.850-858, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01608413

N. Runkel, F. G. Moody, and G. S. Smith, Alterations in rat intestinal transit by morphine promote bacterial translocation, Digest Dis Sci, vol.38, issue.8, pp.1530-1536, 1993.

M. E. Hilburger, M. W. Adler, and A. L. Truant, Morphine induces sepsis in mice, J Infect Dis, vol.176, issue.1, pp.183-188, 1997.

T. J. Khan, Y. M. Ahmed, and M. A. Zamzami, Atorvastatin Treatment Modulates the Gut Microbiota of the Hypercholesterolemic Patients, OMICS, vol.22, issue.2, pp.154-163, 2018.

, Enteric Microbiome Metabolites Correlate with Response to Simvastatin Treatment, 2019.

M. Li, A. Al-sarraf, G. Sinclair, and J. Frohlich, Fish odour syndrome, CMAJ, vol.183, issue.8, pp.929-931, 2011.

Y. Liu, X. Song, and H. Zhou, Gut Microbiome Associates With Lipid-Lowering Effect of Rosuvastatin in Vivo, Front Microbiol, vol.9, 2018.

S. Cussotto, G. Clarke, T. G. Dinan, and J. F. Cryan, Psychotropics and the Microbiome: a Chamber of Secrets?, Psychopharmacology, 2019.

L. Amaral and V. Lorian, Effects of chlorpromazine on the cell envelope proteins of Escherichia coli, Antimicrob Agents Chemother, vol.35, issue.9, pp.1923-1924, 1991.

J. Molnár, J. Király, and Y. Mándi, The antibacterial action and R-factor-inhibiting activity by chlorpromazine, Experientia, vol.31, issue.4, pp.444-445, 1975.

L. Amaral, J. Kristiansen, and V. Lorian, Synergic effect of chiorpromazine on the activity of some antibiotics, J Antimicrob Chemother, vol.30, issue.4, pp.556-558, 1992.

S. G. Dastidar, A. Chaudhury, S. Annadurai, S. Roy, M. Mookerjee et al., In vitro and in vivo antimicrobial action of fluphenazine, J Chemother, vol.7, issue.3, pp.201-206, 1995.

S. A. Flowers, S. J. Evans, K. M. Ward, M. G. Mcinnis, and V. L. Ellingrod, Interaction Between Atypical Antipsychotics and the Gut Microbiome in a Bipolar Disease Cohort, Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy, vol.37, issue.3, pp.261-267, 2017.

K. Hiippala, V. Kainulainen, M. Kalliomäki, P. Arkkila, and R. Satokari, Mucosal Prevalence and Interactions with the Epithelium Indicate Commensalism of Sutterella spp, Frontiers in Microbiology, vol.7, 2016.

S. M. Bahr, B. J. Weidemann, and A. N. Castro, Risperidone-induced weight gain is mediated through shifts in the gut microbiome and suppression of energy expenditure, EBioMedicine, vol.2, issue.11, pp.1725-1734, 2015.

A. P. Morgan, J. J. Crowley, and R. J. Nonneman, The Antipsychotic Olanzapine Interacts with the Gut Microbiome to Cause Weight Gain in Mouse, PLoS One, vol.9, issue.12, 2014.

K. J. Davey, P. D. Cotter, and O. O'sullivan, Antipsychotics and the gut microbiome: olanzapine-induced metabolic dysfunction is attenuated by antibiotic administration in the rat, Transl Psychiatry, vol.3, p.309, 2013.

A. Kao, S. Spitzer, D. C. Anthony, B. Lennox, and P. Burnet, Prebiotic attenuation of olanzapine-induced weight gain in rats: analysis of central and peripheral biomarkers and gut microbiota, Transl Psychiatry, vol.8, 2018.

S. Cussotto, C. R. Strain, and F. Fouhy, Differential effects of psychotropic drugs on microbiome composition and gastrointestinal function, Psychopharmacology (Berl), vol.236, issue.5, pp.1671-1685, 2019.

A. Mandal, C. Sinha, A. K. Jena, S. Ghosh, and A. Samanta, An investigation on in vitro and in vivo antimicrobial properties of the antidepressant: amitriptyline hydrochloride, Brazilian Journal of Microbiology, vol.41, issue.3, pp.635-642, 2010.

M. Ayaz, F. Subhan, and J. Ahmed, Sertraline enhances the activity of antimicrobial agents against pathogens of clinical relevance, J Biol Res (Thessalon), vol.22, issue.1, p.4, 2015.

, Synthesis, antimicrobial activity of lamotrigine and its ammonium derivatives | SpringerLink, 2020.

Z. Begec, A. Yucel, and Y. Yakupogullari, The antimicrobial effects of ketamine combined with propofol: An in vitro study, Braz J Anesthesiol, vol.63, issue.6, pp.461-465, 2013.

S. Gocmen, U. Buyukkocak, and O. Caglayan, In vitro investigation of the antibacterial effect of ketamine, Ups J Med Sci, vol.113, issue.1, pp.39-46, 2008.