G. R. Gibson and M. B. Roberfroid, Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics, J Nutr, vol.125, pp.1401-1413, 1995.

A. Gruss, E. Borezee-durant, and D. Lechardeur, Environmental heme utilization by heme-auxotrophic bacteria, Adv Microb Physiol, vol.61, pp.69-124, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01053114

E. P. Rocha and C. J. Smith, Heme and Iron Metabolism in Bacteroides, Iron uptake and homeostasis in microorganisms, pp.155-65, 2010.

J. De-vogel, W. B. Van-eck, A. L. Sesink, D. S. Jonker-termont, J. Kleibeuker et al., Dietary heme injures surface epithelium resulting in hyperproliferation, inhibition of apoptosis and crypt hyperplasia in rat colon, Carcinogenesis, vol.29, pp.398-403, 2008.

N. Ijssennagger, A. Rijnierse, N. J. De-wit, M. V. Boekschoten, J. Dekker et al., Dietary heme induces acute oxidative stress, but delayed cytotoxicity and compensatory hyperproliferation in mouse colon, Carcinogenesis, vol.34, pp.1628-1663, 2013.

F. Pierre, S. Tache, C. R. Petit, R. Van-der-meer, and D. E. Corpet, Meat and cancer: haemoglobin and haemin in a low-calcium diet promote colorectal carcinogenesis at the aberrant crypt stage in rats, Carcinogenesis, vol.24, pp.1683-90, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00411766

G. R. Zuckerman, C. Prakash, M. P. Askin, and B. S. Lewis, AGA technical review on the evaluation and management of occult and obscure gastrointestinal bleeding, Gastroenterology, vol.118, pp.201-222, 2000.

N. Ijssennagger, M. Derrien, G. M. Van-doorn, A. Rijnierse, B. Van-den-bogert et al., Dietary heme alters microbiota and mucosa of mouse colon without functional changes in host-microbe cross-talk, PLoS One, vol.7, issue.12, p.49868, 2012.

J. F. Sperry, M. D. Appleman, and T. D. Wilkins, Requirement of heme for growth of Bacteroides fragilis, Appl Environ Microbiol, vol.34, pp.386-90, 1977.

F. R. Silva, C. T. Nabeshima, M. H. Bellini, N. Schor, N. D. Vieira et al., Study of protoporphyrin IX elimination by body excreta: a new noninvasive cancer diagnostic method?, J. Fluorescence, vol.23, pp.131-136, 2013.

J. W. Van-den-berg, R. Koole-lesuis, A. Edixhoven-bosdijk, and N. Brouwers, Automating the quantification of heme in feces, Clin Chem, vol.34, pp.2125-2131, 1988.

L. G. Van-rossum, A. F. Van-rijn, R. J. Laheij, M. G. Van-oijen, P. Fockens et al., Random comparison of guaiac and immunochemical fecal occult blood tests for colorectal cancer in a screening population, Gastroenterol, vol.135, pp.82-90, 2008.

D. R. Caldwell, D. C. White, M. P. Bryant, and R. N. Doetsch, Specificity of the heme requirement for growth of Bacteroides ruminicola, J Bacteriol, vol.90, pp.1645-54, 1965.

P. Duwat, S. Sourice, B. Cesselin, G. Lamberet, K. Vido et al., Respiration capacity of the fermenting bacterium Lactococcus lactis and its positive effects on growth and survival, J Bacteriol, vol.183, pp.4509-4525, 2001.

D. A. Ahlquist, D. B. Mcgill, S. Schwartz, W. F. Taylor, M. Ellefson et al., HemoQuant, a new quantitative assay for fecal hemoglobin. Comparison with Hemoccult, Annals Int Med, vol.101, pp.297-302, 1984.

P. Vaananen and R. Tenhunen, Rapid immunochemical detection of fecal occult blood by use of a latex-agglutination test, Clin Chem, vol.34, pp.1763-1769, 1988.

S. L. Saraf, X. Zhang, T. Kanias, J. P. Lash, R. E. Molokie et al., Haemoglobinuria is associated with chronic kidney disease and its progression in patients with sickle cell anaemia, Brit J Haematol, vol.164, pp.729-768, 2014.

J. P. Wang, L. Qi, B. Zheng, F. Liu, M. R. Moore et al., Porphyrins as early biomarkers for arsenic exposure in animals and humans, Cell Mol Biol, vol.48, pp.835-878, 2002.

S. Fanaro, R. Chierici, P. Guerrini, and V. Vigi, Intestinal microflora in early infancy: composition and development, Acta Paediatrica, vol.41, pp.48-55, 2003.

K. Orrhage and C. E. Nord, Factors controlling the bacterial colonization of the intestine in breastfed infants, Acta Paediatr Suppl, vol.88, issue.430, pp.47-57, 1999.

C. Palmer, E. M. Bik, D. B. Digiulio, D. A. Relman, and P. O. Brown, Development of the human infant intestinal microbiota, PLoS Biol, vol.5, issue.7, p.177, 2007.

K. Aagaard, J. Ma, K. M. Antony, R. Ganu, J. Petrosino et al., The placenta harbors a unique microbiome, Sci Transl Med, vol.6, pp.237-265, 2014.

M. C. Abt, L. C. Osborne, L. A. Monticelli, T. A. Doering, T. Alenghat et al., Commensal bacteria calibrate the activation threshold of innate antiviral immunity, Immunity, vol.37, pp.158-170, 2012.

I. Adlerberth, Factors influencing the establishment of the intestinal microbiota in infancy, Nestle Nutr Workshop Ser Pediatr Program, vol.62, pp.29-33, 2008.

I. Adlerberth and A. E. Wold, Establishment of the gut microbiota in Western infants, Acta Paediatr, vol.98, pp.229-238, 2009.

D. A. Ahlquist, D. B. Mcgill, S. Schwartz, W. F. Taylor, M. Ellefson et al., , 1984.

. Hemoquant, Comparison with Hemoccult. Ann Intern Med, vol.101, pp.297-302

T. A. Ajslev, C. S. Andersen, M. Gamborg, T. I. Sorensen, J. et al., Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics, Int J Obes (Lond), vol.35, pp.522-529, 2011.

A. Albrecht, G. Caliskan, M. S. Oitzl, U. Heinemann, and O. Stork, Long-lasting increase of corticosterone after fear memory reactivation: anxiolytic effects and network activity modulation in the ventral hippocampus, Neuropsychopharmacology, vol.38, pp.386-394, 2013.

D. An, C. Na, J. Bielawski, Y. A. Hannun, and D. L. Kasper, Membrane sphingolipids as essential molecular signals for Bacteroides survival in the intestine, Proc Natl Acad Sci U S A, vol.108, pp.4666-4671, 2011.

S. C. Andrews, The Ferritin-like superfamily: Evolution of the biological iron storeman from a rubrerythrin-like ancestor, Biochim Biophys Acta, vol.1800, pp.691-705, 2010.

S. C. Andrews, A. K. Robinson, and F. Rodriguez-quinones, Bacterial iron homeostasis, FEMS Microbiol Rev, vol.27, pp.215-237, 2003.

T. Arora and F. Backhed, The gut microbiota and metabolic disease: current understanding and future perspectives, J Intern Med, 2016.

M. Arumugam, J. Raes, E. Pelletier, D. Le-paslier, T. Yamada et al., Enterotypes of the human gut microbiome, Nature, vol.473, pp.174-180, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00903625

P. Ascenzi, A. Di-masi, L. Leboffe, E. Frangipani, M. Nardini et al., Structural Biology of Bacterial Haemophores, Adv Microb Physiol, vol.67, pp.127-176, 2015.

H. Atamna and W. H. Frey, A role for heme in Alzheimer's disease: heme binds amyloid beta and has altered metabolism, Proc Natl Acad Sci U S A, vol.101, pp.11153-11158, 2004.

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

F. Backhed, H. Ding, T. Wang, L. V. Hooper, G. Y. Koh et al., The gut microbiota as an environmental factor that regulates fat storage, Proc Natl Acad Sci U S A, vol.101, pp.15718-15723, 2004.

F. Backhed, R. E. Ley, J. L. Sonnenburg, D. A. Peterson, G. et al., Host-bacterial mutualism in the human intestine, Science, vol.307, pp.1915-1920, 2005.

F. Backhed, J. Roswall, Y. Peng, Q. Feng, H. Jia et al., Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life, Cell Host Microbe, vol.17, p.852, 2015.

N. M. Bastide, F. Chenni, M. Audebert, R. L. Santarelli, S. Tache et al., A central role for heme iron in colon carcinogenesis associated with red meat intake, Cancer Res, vol.75, pp.870-879, 2015.

A. D. Baughn and M. H. Malamy, The essential role of fumarate reductase in haemdependent growth stimulation of Bacteroides fragilis, Microbiology, vol.149, pp.1551-1558, 2003.

A. D. Baughn and M. H. Malamy, The strict anaerobe Bacteroides fragilis grows in and benefits from nanomolar concentrations of oxygen, Nature, vol.427, pp.441-444, 2004.

C. Belzer, G. K. Gerber, G. Roeselers, M. Delaney, A. Dubois et al., Dynamics of the microbiota in response to host infection, PLoS One, vol.9, p.95534, 2014.

N. Benevides-matos, C. Wandersman, and F. Biville, HasB, the Serratia marcescens TonB paralog, is specific to HasR, J Bacteriol, vol.190, pp.21-27, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00319813

Y. Benno, K. Sawada, and T. Mitsuoka, The intestinal microflora of infants: composition of fecal flora in breast-fed and bottle-fed infants, Microbiol Immunol, vol.28, pp.975-986, 1984.

R. D. Berg, The indigenous gastrointestinal microflora, Trends Microbiol, vol.4, pp.430-435, 1996.

L. G. Bermudez-humaran, N. G. Cortes-perez, S. Ah-leung, F. Lefevre, G. Yang et al., Current prophylactic and therapeutic uses of a recombinant Lactococcus lactis strain secreting biologically active interleukin-12, J Mol Microbiol Biotechnol, vol.14, pp.80-89, 2008.

E. Biagi, S. Quercia, A. Aceti, I. Beghetti, S. Rampelli et al., The Bacterial Ecosystem of Mother's Milk and Infant's Mouth and Gut, 2017.

F. Biville, H. Cwerman, S. Letoffe, M. S. Rossi, V. Drouet et al.,

, Haemophore-mediated signalling in Serratia marcescens: a new mode of regulation for an extra cytoplasmic function (ECF) sigma factor involved in haem acquisition, Mol Microbiol, vol.53, pp.1267-1277

F. R. Blattner, G. Plunkett, C. A. Bloch, N. T. Perna, V. Burland et al., The complete genome sequence of Escherichia coli K-12, Science, vol.277, pp.1453-1462, 1997.

A. Boleij, E. M. Hechenbleikner, A. C. Goodwin, R. Badani, E. M. Stein et al., The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients, Clin Infect Dis, vol.60, pp.208-215, 2015.

G. H. Bornside, W. E. Donovan, and M. B. Myers, Intracolonic tensions of oxygen and carbon dioxide in germfree, conventional, and gnotobiotic rats, Proc Soc Exp Biol Med, vol.151, pp.437-441, 1976.

K. E. Bouter, D. H. Van-raalte, A. K. Groen, and M. Nieuwdorp, Role of the Gut Microbiome in the Pathogenesis of Obesity and Obesity-Related Metabolic Dysfunction, Gastroenterology, 2017.

V. Bouvard, D. Loomis, K. Z. Guyton, Y. Grosse, F. E. Ghissassi et al., and International Agency for Research on Cancer Monograph Working, Lancet Oncol, vol.16, pp.1599-1600, 2015.

J. Breton, C. Daniel, J. Dewulf, S. Pothion, N. Froux et al., Gut microbiota limits heavy metals burden caused by chronic oral exposure, Toxicol Lett, vol.222, pp.132-138, 2013.

I. Brook, Enhancement of growth of aerobic and facultative bacteria in mixed infections with Bacteroides species, Infect Immun, vol.50, pp.929-931, 1985.

I. Brook, Pathogenicity of the Bacteroides fragilis group, Ann Clin Lab Sci, vol.19, pp.360-376, 1989.

C. G. Buffie and E. G. Pamer, Microbiota-mediated colonization resistance against intestinal pathogens, Nat Rev Immunol, vol.13, pp.790-801, 2013.

D. P. Burkitt, A. R. Walker, and N. S. Painter, Dietary fiber and disease, JAMA, vol.229, pp.1068-1074, 1974.

D. R. Caldwell, D. C. White, M. P. Bryant, and R. N. Doetsch, Specificity of the heme requirement for growth of Bacteroides ruminicola, J Bacteriol, vol.90, pp.1645-1654, 1965.

M. L. Cartron, S. Maddocks, P. Gillingham, C. J. Craven, and S. C. Andrews, Feo--transport of ferrous iron into bacteria, Biometals, vol.19, pp.143-157, 2006.

A. Castellani, C. , and A. J. , Manual of tropical medecine, pp.959-960, 1919.

E. P. Cato, J. , and J. L. , Reinstatement of Species Rank for Bacteroides fragilis, B. ovatus, B. distasonis, B. thetaiotaomicron, and B. vulgatus: Designation of Neotype Strains for Bacteroides fragilis (Veillon and Zuber) Castellani and Chalmers and Bacteroides thetaiotaomicron (Distaso) Castellani and Chalmers, Int J Syst Bacteriol, vol.26, pp.230-237, 1976.

N. I. Chalmers, R. J. Palmer, . Jr, J. O. Cisar, and P. E. Kolenbrander, Characterization of a Streptococcus sp.-Veillonella sp. community micromanipulated from dental plaque, J Bacteriol, vol.190, pp.8145-8154, 2008.

D. Chen, J. D. Brown, Y. Kawasaki, J. Bommer, and J. Y. Takemoto, Scalable production of biliverdin IXalpha by Escherichia coli, BMC Biotechnol, vol.12, p.89, 2012.

C. Cherbuy, E. Honvo-houeto, A. Bruneau, C. Bridonneau, C. Mayeur et al., Microbiota matures colonic epithelium through a coordinated induction of cell cycle-related proteins in gnotobiotic rat, Am J Physiol Gastrointest Liver Physiol, vol.299, pp.348-357, 2010.

C. A. Cherrington, M. Hinton, G. R. Pearson, and I. Chopra, Short-chain organic acids at ph 5.0 kill Escherichia coli and Salmonella spp. without causing membrane perturbation, J Appl Bacteriol, vol.70, pp.161-165, 1991.

J. E. Choby and E. P. Skaar, Heme Synthesis and Acquisition in Bacterial Pathogens, J Mol Biol, vol.428, pp.3408-3428, 2016.

D. M. Chu, J. Ma, A. L. Prince, K. M. Antony, M. D. Seferovic et al., , 2017.

, Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery, Nat Med, vol.23, pp.314-326

S. M. Collins, M. Surette, and P. Bercik, The interplay between the intestinal microbiota and the brain, Nat Rev Microbiol, vol.10, pp.735-742, 2012.

L. E. Comstock and M. J. Coyne, Bacteroides thetaiotaomicron: a dynamic, nicheadapted human symbiont, Bioessays, vol.25, pp.926-929, 2003.

P. L. Conway, S. L. Gorbach, and B. R. Goldin, Survival of lactic acid bacteria in the human stomach and adhesion to intestinal cells, J Dairy Sci, vol.70, pp.1-12, 1987.

D. E. Corpet, Red meat and colon cancer: should we become vegetarians, or can we make meat safer?, Meat Sci, vol.89, pp.310-316, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00592966

M. J. Coyne, W. Kalka-moll, A. O. Tzianabos, D. L. Kasper, and L. E. Comstock, , 2000.

, Bacteroides fragilis NCTC9343 produces at least three distinct capsular polysaccharides: cloning, characterization, and reassignment of polysaccharide B and C biosynthesis loci, Infect Immun, vol.68, pp.6176-6181

A. J. Cross, L. M. Ferrucci, A. Risch, B. I. Graubard, M. H. Ward et al., A large prospective study of meat consumption and colorectal cancer risk: an investigation of potential mechanisms underlying this association, Cancer Res, vol.70, pp.2406-2414, 2010.

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

J. H. Cummings and G. T. Macfarlane, The control and consequences of bacterial fermentation in the human colon, J Appl Bacteriol, vol.70, pp.443-459, 1991.

M. De-angelis, R. Francavilla, M. Piccolo, A. De-giacomo, and M. Gobbetti, Autism spectrum disorders and intestinal microbiota, Gut Microbes, vol.6, pp.207-213, 2015.

M. De-paepe, M. Leclerc, C. R. Tinsley, and M. A. Petit, Bacteriophages: an underestimated role in human and animal health? Front Cell Infect Microbiol 4, 39, Curr Opin Microbiol, vol.2, pp.289-295, 1999.

A. Distaso, Contribution à l'étude sur l'intoxication intestinale, Zbl Bakt, I Abt Orig, vol.62, pp.433-468, 1912.

A. Distaso, Flora of wounds and flora of putrefaction. The Lancet, pp.74-75, 1916.

C. Dobell, Antony van Leeuwenhoek and his" Little Animals, 1932.

J. L. Domingo and M. Nadal, Carcinogenicity of consumption of red meat and processed meat: A review of scientific news since the IARC decision, Food Chem Toxicol, vol.105, pp.256-261, 2017.

R. M. Donaldson and . Jr, Normal Bacterial Populations of the Intestine and Their Relation to Intestinal Function, N Engl J Med, vol.270, pp.1050-1056, 1964.

R. M. Donaldson and . Jr, Normal Bacterial Populations of the Intestine and Their Relation to Intestinal Function, N Engl J Med, vol.270, pp.994-1001, 1964.

R. M. Donaldson and . Jr, Normal Bacterial Populations of the Intestine and Their Relation to Intestinal Function, N Engl J Med, vol.270, pp.938-945, 1964.

B. S. Drasar, Cultivation of anaerobic intestinal bacteria, J Pathol Bacteriol, vol.94, pp.417-427, 1967.

B. S. Drasar and M. J. Hill, Human Intestinal Flora, 1974.

B. S. Drasar, M. Shiner, and G. M. Mcleod, Studies on the intestinal flora. I. The bacterial flora of the gastrointestinal tract in healthy and achlorhydric persons, Gastroenterology, vol.56, pp.71-79, 1969.

D. L. Dunn and R. L. Simmons, The role of anaerobic bacteria in intraabdominal infections, Rev Infect Dis, vol.6, issue.1, pp.139-146, 1984.

F. F. Dutra and M. T. Bozza, Heme on innate immunity and inflammation, Front Pharmacol, vol.5, p.115, 2014.

P. Duwat, S. Sourice, B. Cesselin, G. Lamberet, K. Vido et al., Respiration capacity of the fermenting bacterium Lactococcus lactis and its positive effects on growth and survival, J Bacteriol, vol.183, pp.4509-4516, 2001.

A. H. Eggerth and B. H. Gagnon, The Bacteroides of Human Feces, J Bacteriol, vol.25, pp.389-413, 1933.

T. Escherich, Die Darmbakterien des Neugeborenen und Säuglings (Les bactéries intestinale du nouveau-né et de l'enfant), Fortschritte der Medicin, vol.3, pp.515-557, 1885.

A. Everard, C. Belzer, L. Geurts, J. P. Ouwerkerk, C. Druart et al., Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity, Proc Natl Acad Sci U S A, vol.110, pp.9066-9071, 2013.

W. Fang, M. Shi, L. Huang, J. Chen, W. et al., Antagonism of lactic acid bacteria towards Staphylococcus aureus and Escherichia coli on agar plates and in milk, Vet Res, vol.27, pp.3-12, 1996.
URL : https://hal.archives-ouvertes.fr/hal-00902393

A. Fernandez, D. Lechardeur, A. Derre-bobillot, E. Couve, P. Gaudu et al., Two coregulated efflux transporters modulate intracellular heme and protoporphyrin IX availability in Streptococcus agalactiae, PLoS Pathog, vol.6, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01606046

S. M. Finegold, Therapy and epidemiology of autism--clostridial spores as key elements, Med Hypotheses, vol.70, pp.508-511, 2008.

A. Fleming, On the bacteriology of septic wounds. The Lancet, pp.638-643, 1915.

L. Flohe, S. Toppo, G. Cozza, and F. Ursini, A comparison of thiol peroxidase mechanisms, Antioxid Redox Signal, vol.15, pp.763-780, 2011.

M. H. Foley, D. W. Cockburn, and N. M. Koropatkin, The Sus operon: a model system for starch uptake by the human gut Bacteroidetes, Cell Mol Life Sci, vol.73, pp.2603-2617, 2016.

N. Frankenberg, J. Moser, J. , and D. , Bacterial heme biosynthesis and its biotechnological application, Appl Microbiol Biotechnol, vol.63, pp.115-127, 2003.

R. Freter and G. D. Abrams, Function of various intestinal bacteria in converting germfree mice to the normal state, Infect Immun, vol.6, pp.119-126, 1972.

R. Freter, H. Brickner, J. Fekete, M. M. Vickerman, and K. E. Carey, Survival and implantation of Escherichia coli in the intestinal tract, Infect Immun, vol.39, pp.686-703, 1983.

S. Fukuda, H. Toh, K. Hase, K. Oshima, Y. Nakanishi et al., Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature, vol.469, pp.543-547, 2011.

M. J. Gasson, Plasmid complements of Streptococcus lactis NCDO 712 and other lactic streptococci after protoplast-induced curing, J Bacteriol, vol.154, pp.1-9, 1983.

G. H. Gauss, P. Benas, B. Wiedenheft, M. Young, T. Douglas et al., Structure of the DPS-like protein from Sulfolobus solfataricus reveals a bacterioferritin-like dimetal binding site within a DPS-like dodecameric assembly, Biochemistry, vol.45, pp.10815-10827, 2006.

L. Geurts, A. M. Neyrinck, N. M. Delzenne, C. Knauf, and P. D. Cani, Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics, Benef Microbes, vol.5, pp.3-17, 2014.

J. M. Ghigo, S. Letoffe, and C. Wandersman, A new type of hemophore-dependent heme acquisition system of Serratia marcescens reconstituted in Escherichia coli, J Bacteriol, vol.179, pp.3572-3579, 1997.

S. Ghosh, C. Dai, K. Brown, E. Rajendiran, S. Makarenko et al., Colonic microbiota alters host susceptibility to infectious colitis by modulating inflammation, redox status, and ion transporter gene expression, Am J Physiol Gastrointest Liver Physiol, vol.301, pp.39-49, 2011.

R. J. Gibbons and J. B. Macdonald, Hemin and vitamin K compounds as required factors for the cultivation of certain strains of Bacteroides melaninogenicus, J Bacteriol, vol.80, pp.164-170, 1960.

L. Gibold, E. Garenaux, G. Dalmasso, C. Gallucci, D. Cia et al., The Vat-AIEC protease promotes crossing of the intestinal mucus layer by Crohn's disease-associated Escherichia coli, Cell Microbiol, vol.18, pp.617-631, 2016.

V. A. Gilbertsen, R. Mchugh, L. Schuman, and S. E. Williams, The earlier detection of colorectal cancers: a preliminary report of the results of the Occult Blood Study, Cancer, vol.45, pp.2899-2901, 1980.

S. R. Gill, M. Pop, R. T. Deboy, P. B. Eckburg, P. J. Turnbaugh et al., Metagenomic analysis of the human distal gut microbiome, Science, vol.312, pp.1355-1359, 2006.

M. Glei, S. Klenow, J. Sauer, U. Wegewitz, K. Richter et al., Hemoglobin and hemin induce DNA damage in human colon tumor cells HT29 clone 19A and in primary human colonocytes, Mutat Res, vol.594, pp.162-171, 2006.

E. J. Goldstein, Anaerobic bacteremia, Clin Infect Dis, vol.23, issue.1, pp.97-101, 1996.

S. Gomez-de-la-torre-canny and J. F. Rawls, Baby, It's Cold Outside: Host-Microbiota Relationships Drive Temperature Adaptations, Cell Host Microbe, vol.18, pp.635-636, 2015.

P. Gorwood, C. Blanchet-collet, N. Chartrel, J. Duclos, P. Dechelotte et al., New Insights in Anorexia Nervosa, vol.10, p.256, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02330466

C. Gower-rousseau, Epidémiologie des Maladies Inflammatoires Chroniques de l'Intestin en France : apportdu Registre EPIMAD, 2012.

A. Granger, M. M. Richer, and S. Roustel, Microbiochimie et alimentation, 2007.

A. Gruss, E. Borezee-durant, and D. Lechardeur, Environmental heme utilization by heme-auxotrophic bacteria, Adv Microb Physiol, vol.61, pp.69-124, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01053114

T. Haahtela, S. Holgate, R. Pawankar, C. A. Akdis, S. Benjaponpitak et al., The biodiversity hypothesis and allergic disease: world allergy organization position statement, World Allergy Organ J, vol.6, p.3, 2013.

K. E. Hagman, W. Pan, B. G. Spratt, J. T. Balthazar, R. C. Judd et al., Resistance of Neisseria gonorrhoeae to antimicrobial hydrophobic agents is modulated by the mtrRCDE efflux system, Microbiology, vol.141, pp.611-622, 1995.

R. L. Hahnke, J. P. Meier-kolthoff, M. Garcia-lopez, S. Mukherjee, M. Huntemann et al., Genome-Based Taxonomic Classification of Bacteroidetes, Front Microbiol, vol.7, 2003.

B. Halliwell and J. M. Gutteridge, Oxygen toxicity, oxygen radicals, transition metals and disease, Biochem J, vol.219, pp.1-14, 1984.

D. Halpern and A. Gruss, A sensitive bacterial-growth-based test reveals how intestinal Bacteroides meet their porphyrin requirement, BMC Microbiol, vol.15, p.282, 2015.

N. D. Hammer and E. P. Skaar, Molecular mechanisms of Staphylococcus aureus iron acquisition, Annu Rev Microbiol, vol.65, pp.129-147, 2011.

K. Hantke, Is the bacterial ferrous iron transporter FeoB a living fossil, Trends Microbiol, vol.11, pp.192-195, 2003.

I. U. Heinemann, M. Jahn, J. , and D. , The biochemistry of heme biosynthesis, Arch Biochem Biophys, vol.474, pp.238-251, 2008.

J. D. Helmann, The extracytoplasmic function (ECF) sigma factors, Adv Microb Physiol, vol.46, pp.47-110, 2002.

C. D. Herren, E. R. Rocha, and C. J. Smith, Genetic analysis of an important oxidative stress locus in the anaerobe Bacteroides fragilis, Gene, vol.316, pp.167-175, 2003.

L. V. Holdeman, I. J. Good, and W. E. Moore, Human fecal flora: variation in bacterial composition within individuals and a possible effect of emotional stress, Appl Environ Microbiol, vol.31, pp.359-375, 1976.

E. Holmes, J. V. Li, J. R. Marchesi, and J. K. Nicholson, Gut microbiota composition and activity in relation to host metabolic phenotype and disease risk, Cell Metab, vol.16, pp.559-564, 2012.

K. Honda and D. R. Littman, The microbiome in infectious disease and inflammation, Annu Rev Immunol, vol.30, pp.759-795, 2012.

L. C. Hoskins, M. Agustines, W. B. Mckee, E. T. Boulding, M. Kriaris et al., Mucin degradation in human colon ecosystems. Isolation and properties of fecal strains that degrade ABH blood group antigens and oligosaccharides from mucin glycoproteins, J Clin Invest, vol.75, pp.944-953, 1985.

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, pp.1451-1463, 2013.

N. Ijssennagger, C. Belzer, G. J. Hooiveld, J. Dekker, S. W. Van-mil et al., Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon, Proc Natl Acad Sci U S A, vol.112, pp.10038-10043, 2015.

N. Ijssennagger, M. Derrien, G. M. Van-doorn, A. Rijnierse, . Van-den et al., Dietary heme alters microbiota and mucosa of mouse colon without functional changes in host-microbe cross-talk, PLoS One, vol.7, p.49868, 2012.

J. A. Imlay, Cellular defenses against superoxide and hydrogen peroxide, Annu Rev Biochem, vol.77, pp.755-776, 2008.

J. A. Imlay, How obligatory is anaerobiosis ?, Mol Microbiol, vol.68, pp.801-804, 2008.

T. F. Imperiale, D. F. Ransohoff, S. H. Itzkowitz, T. R. Levin, P. Lavin et al., Multitarget stool DNA testing for colorectal-cancer screening, N Engl J Med, vol.370, pp.1287-1297, 2014.

S. A. Jones, T. Gibson, R. C. Maltby, F. Z. Chowdhury, V. Stewart et al.,

, Anaerobic respiration of Escherichia coli in the mouse intestine, Infect Immun, vol.79, pp.4218-4226

T. Jost, C. Lacroix, C. P. Braegger, F. Rochat, and C. Chassard, Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding, Environ Microbiol, vol.16, pp.2891-2904, 2014.

L. Joubert, J. B. Dagieu, A. Fernandez, A. Derre-bobillot, E. Borezee-durant et al., Visualization of the role of host heme on the virulence of the heme auxotroph Streptococcus agalactiae. Sci Rep, vol.7, p.40435, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01594946

L. Joubert, A. Derre-bobillot, P. Gaudu, A. Gruss, and D. Lechardeur, HrtBA and menaquinones control haem homeostasis in Lactococcus lactis, Mol Microbiol, vol.93, pp.823-833, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01204405

M. Jungano and A. Distaso, , 1910.

N. Kamada, G. Y. Chen, N. Inohara, and G. Nunez, Control of pathogens and pathobionts by the gut microbiota, Nat Immunol, vol.14, pp.685-690, 2013.

M. Kanehisa, A database for post-genome analysis, Trends Genet, vol.13, pp.375-376, 1997.

F. H. Karlsson, F. Fak, I. Nookaew, V. Tremaroli, B. Fagerberg et al., Symptomatic atherosclerosis is associated with an altered gut metagenome, Nat Commun, vol.3, p.1245, 2012.

K. Kato, R. Tanaka, S. Sano, A. Tanaka, and H. Hosaka, Identification of a gene essential for protoporphyrinogen IX oxidase activity in the cyanobacterium Synechocystis sp. PCC6803, Proc Natl Acad Sci U S A, vol.107, pp.16649-16654, 2010.

I. Kirschner-zilber, E. Rabizadeh, and N. Shaklai, The interaction of hemin and bilirubin with the human red cell membrane, Biochim Biophys Acta, vol.690, pp.20-30, 1982.

N. Klijn, A. H. Weerkamp, and W. M. De-vos, Genetic marking of Lactococcus lactis shows its survival in the human gastrointestinal tract, Appl Environ Microbiol, vol.61, pp.2771-2774, 1995.

D. Knights, T. L. Ward, C. E. Mckinlay, H. Miller, A. Gonzalez et al., Rethinking "enterotypes, Cell Host Microbe, vol.16, pp.433-437, 2014.

J. E. Koenig, A. Spor, N. Scalfone, A. D. Fricker, J. Stombaugh et al., Succession of microbial consortia in the developing infant gut microbiome, Proc Natl Acad Sci U S A, vol.108, pp.4578-4585, 2011.

O. Koren, A. Spor, J. Felin, F. Fak, J. Stombaugh et al., Human oral, gut, and plaque microbiota in patients with atherosclerosis, Proc Natl Acad Sci U S A, vol.108, pp.4592-4598, 2011.

N. R. Krieg, J. T. Staley, D. R. Brown, B. P. Hedlund, B. J. Paster et al., Bergey's Manual of Systematic Bacteriology, 2010.

S. Kumar and U. Bandyopadhyay, Free heme toxicity and its detoxification systems in human, Toxicol Lett, vol.157, pp.175-188, 2005.

T. Kuwahara, A. Yamashita, H. Hirakawa, H. Nakayama, H. Toh et al., Genomic analysis of Bacteroides fragilis reveals extensive DNA inversions regulating cell surface adaptation, Proc Natl Acad Sci U S A, vol.101, pp.14919-14924, 2004.

S. J. Kwon, A. L. De-boer, R. Petri, and C. Schmidt-dannert, High-level production of porphyrins in metabolically engineered Escherichia coli: systematic extension of a pathway assembled from overexpressed genes involved in heme biosynthesis, Appl Environ Microbiol, vol.69, pp.4875-4883, 2003.

D. J. Lane, D. H. Bae, A. M. Merlot, S. Sahni, and D. R. Richardson, Duodenal cytochrome b (DCYTB) in iron metabolism: an update on function and regulation, Nutrients, vol.7, pp.2274-2296, 2015.

J. P. Laplace, Le transit digestif chez les monogastriques, Ann Zootech, vol.24, pp.489-552, 1975.
URL : https://hal.archives-ouvertes.fr/hal-00887804

K. Latalova, M. Hajda, and J. Prasko, Can gut microbes play a role in mental disorders and their treatment?, Psychiatr Danub, vol.29, pp.28-30, 2017.

C. K. Lau, K. D. Krewulak, and H. J. Vogel, Bacterial ferrous iron transport: the Feo system, FEMS Microbiol Rev, vol.40, pp.273-298, 2016.

G. D. Launoy, H. J. Bertrand, C. Berchi, V. Y. Talbourdet, A. V. Guizard et al., Evaluation of an immunochemical fecal occult blood test with automated reading in screening for colorectal cancer in a general average-risk population, Int J Cancer, vol.115, pp.493-496, 2005.

D. Lechardeur, B. Cesselin, U. Liebl, M. H. Vos, A. Fernandez et al., Discovery of intracellular heme-binding protein HrtR, which controls heme efflux by the conserved HrtB-HrtA transporter in Lactococcus lactis, J Biol Chem, vol.287, pp.4752-4758, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00324331

S. Letoffe, L. Debarbieux, N. Izadi, P. Delepelaire, and C. Wandersman, Ligand delivery by haem carrier proteins: the binding of Serratia marcescens haemophore to its outer membrane receptor is mediated by two distinct peptide regions, Mol Microbiol, vol.50, pp.77-88, 2003.
URL : https://hal.archives-ouvertes.fr/pasteur-00366306

S. Letoffe, P. Delepelaire, and C. Wandersman, The housekeeping dipeptide permease is the Escherichia coli heme transporter and functions with two optional peptide binding proteins, Proc Natl Acad Sci U S A, vol.103, pp.12891-12896, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00189637

S. Letoffe, J. M. Ghigo, and C. Wandersman, Iron acquisition from heme and hemoglobin by a Serratia marcescens extracellular protein, Proc Natl Acad Sci U S A, vol.91, pp.9876-9880, 1994.

S. Letoffe, K. Wecker, M. Delepierre, P. Delepelaire, and C. Wandersman, Activities of the Serratia marcescens heme receptor HasR and isolated plug and beta-barrel domains: the beta-barrel forms a heme-specific channel, J Bacteriol, vol.187, pp.4637-4645, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00020364

M. D. Levitt, Oxygen tension in the gut, N Engl J Med, vol.282, pp.1039-1040, 1970.

J. P. Lewis, K. Plata, F. Yu, A. Rosato, and C. Anaya, Transcriptional organization, regulation and role of the Porphyromonas gingivalis W83 hmu haemin-uptake locus, Microbiology, vol.152, pp.3367-3382, 2006.

R. E. Ley, F. Backhed, P. Turnbaugh, C. A. Lozupone, R. D. Knight et al., Obesity alters gut microbial ecology, Proc Natl Acad Sci U S A, vol.102, pp.11070-11075, 2005.

R. E. Ley, P. J. Turnbaugh, S. Klein, G. , and J. I. , Microbial ecology: human gut microbes associated with obesity, Nature, vol.444, pp.1022-1023, 2006.

S. Lindquist and O. Hernell, Lipid digestion and absorption in early life: an update, Curr Opin Clin Nutr Metab Care, vol.13, pp.314-320, 2010.

V. Lombard, H. Golaconda-ramulu, E. Drula, P. M. Coutinho, and B. Henrissat, The carbohydrate-active enzymes database (CAZy) in 2013, Nucleic Acids Res, vol.42, pp.490-495, 2014.

B. Lorber and R. M. Swenson, The bacteriology of intra-abdominal infections, Surg Clin North Am, vol.55, pp.1349-1354, 1975.

P. Louis, Does the human gut microbiota contribute to the etiology of autism spectrum disorders?, Dig Dis Sci, vol.57, 1987.

P. Louis, G. L. Hold, and H. J. Flint, The gut microbiota, bacterial metabolites and colorectal cancer, Nat Rev Microbiol, vol.12, pp.661-672, 2014.

Z. Lu and J. A. Imlay, The Fumarate Reductase of Bacteroides thetaiotaomicron, unlike That of Escherichia coli, Is Configured so that It Does Not Generate Reactive Oxygen Species, MBio, vol.8, 2017.

T. D. Luckey, Introduction to intestinal microecology, Am J Clin Nutr, vol.25, pp.1292-1294, 1972.

R. I. Mackie, A. Sghir, and H. R. Gaskins, Developmental microbial ecology of the neonatal gastrointestinal tract, Am J Clin Nutr, vol.69, pp.1035-1045, 1999.

A. J. Macpherson, L. Hunziker, K. Mccoy, and A. Lamarre, IgA responses in the intestinal mucosa against pathogenic and non-pathogenic microorganisms, Microbes Infect, vol.3, pp.1021-1035, 2001.

A. J. Macpherson and T. Uhr, Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria, Science, vol.303, pp.1662-1665, 2004.

F. A. Macrae, D. J. St-john, P. Caligiore, L. S. Taylor, and J. W. Legge, Optimal dietary conditions for hemoccult testing, Gastroenterology, vol.82, pp.899-903, 1982.

J. Macy, I. Probst, and G. Gottschalk, Evidence for cytochrome involvement in fumarate reduction and adenosine 5'-triphosphate synthesis by Bacteroides fragilis grown in the presence of hemin, J Bacteriol, vol.123, pp.436-442, 1975.

J. C. Madan, R. C. Salari, D. Saxena, L. Davidson, G. A. O'toole et al., Gut microbial colonisation in premature neonates predicts neonatal sepsis, Arch Dis Child Fetal Neonatal Ed, vol.97, pp.456-462, 2012.

P. Manfredi, F. Lauber, F. Renzi, K. Hack, E. Hess et al., New iron acquisition system in Bacteroidetes, Infect Immun, vol.83, pp.300-310, 2015.

I. Marmol, C. Sanchez-de-diego, A. Pradilla-dieste, E. Cerrada, R. Yoldi et al., Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer, Int J Mol Sci, vol.18, 2017.

E. C. Martens, H. C. Chiang, G. , and J. I. , Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont, Cell Host Microbe, vol.4, pp.447-457, 2008.

R. Martin, F. Chain, S. Miquel, J. Lu, J. J. Gratadoux et al., The commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-induced chronic moderate and severe colitis models, Inflamm Bowel Dis, vol.20, pp.417-430, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00931932

M. Matsumoto, R. Kibe, T. Ooga, Y. Aiba, S. Kurihara et al., Impact of intestinal microbiota on intestinal luminal metabolome, Sci Rep, vol.2, p.233, 2012.

D. Maurzerall, Normal Porphyrin Metabolism, J Pediatr, vol.64, pp.5-16, 1964.

S. K. Mazmanian, J. L. Round, and D. L. Kasper, A microbial symbiosis factor prevents intestinal inflammatory disease, Nature, vol.453, pp.620-625, 2008.

J. M. Mccord, B. B. Keele, . Jr, and I. Fridovich, An enzyme-based theory of obligate anaerobiosis: the physiological function of superoxide dismutase, Proc Natl Acad Sci U S A, vol.68, pp.1024-1027, 1971.

A. T. Mckie, P. Marciani, A. Rolfs, K. Brennan, K. Wehr et al., A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation, Mol Cell, vol.5, pp.299-309, 2000.

B. M. Meehan, A. D. Baughn, R. Gallegos, and M. H. Malamy, Inactivation of a single gene enables microaerobic growth of the obligate anaerobe Bacteroides fragilis, Proc Natl Acad Sci U S A, vol.109, pp.12153-12158, 2012.

B. M. Meehan and M. H. Malamy, Fumarate reductase is a major contributor to the generation of reactive oxygen species in the anaerobe Bacteroides fragilis, Microbiology, vol.158, pp.539-546, 2012.

M. Mehta, D. S. Goldfarb, and L. Nazzal, The role of the microbiome in kidney stone formation, Int J Surg, vol.36, pp.607-612, 2016.

S. M. Mense and L. Zhang, Heme: a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases, Cell Res, vol.16, pp.681-692, 2006.

J. C. Meslin, E. Sacquet, and J. L. Guenet, Action de la flore bactérienne sur la morphologie et la surface de la muqueuse de l'intestin grêle du rat, Ann Biol anim Bioch Biophys, vol.13, pp.203-214, 1973.

J. C. Meslin, E. Sacquet, and P. Raibaud, Action d'une flore microbienne qui ne déconjugue pas les sels biliaires sur la morphologie et le renouvellement cellulaire de la muqueuse de l'intestin grêle du rat, Ann Biol anim Bioch Biophys, vol.14, pp.709-720, 1974.

E. Metchnikoff, Les microbes intestinaux, Bulletin de l'Institut Pasteur I, 1903.
URL : https://hal.archives-ouvertes.fr/pasteur-00724013

E. Metchnikoff, The nature of man: studies in optimistic philosophy: Heinemann), 1903.

M. Miethke, Molecular strategies of microbial iron assimilation: from high-affinity complexes to cofactor assembly systems, Metallomics, vol.5, pp.15-28, 2013.

M. K. Mirzaei, M. , and C. F. , Menage a trois in the human gut: interactions between host, bacteria and phages, Nat Rev Microbiol, 2017.

S. Mishra and J. A. Imlay, An anaerobic bacterium, Bacteroides thetaiotaomicron, uses a consortium of enzymes to scavenge hydrogen peroxide, Mol Microbiol, vol.90, pp.1356-1371, 2013.

T. Mitsuoka, Intestinal flora and human health, Asia Pac J Clin Nutr, vol.5, pp.2-9, 1996.

T. Mitsuoka, Establishment of intestinal bacteriology, Biosci Microbiota Food Health, vol.33, pp.99-116, 2014.

J. L. Mokili, F. Rohwer, and B. E. Dutilh, Metagenomics and future perspectives in virus discovery, Curr Opin Virol, vol.2, pp.63-77, 2012.

W. E. Moore and L. V. Holdeman, Human fecal flora: the normal flora of 20 Japanese-Hawaiians, Appl Microbiol, vol.27, pp.961-979, 1974.

M. U. Muckenthaler, S. Rivella, M. W. Hentze, and B. Galy, A Red Carpet for Iron Metabolism, Cell, vol.168, pp.344-361, 2017.

J. B. Neilands, Microbial iron compounds, Annu Rev Biochem, vol.50, pp.715-731, 1981.

N. Noinaj, M. Guillier, T. J. Barnard, and S. K. Buchanan, TonB-dependent transporters: regulation, structure, and function, Annu Rev Microbiol, vol.64, pp.43-60, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00555668

G. Nuttall and H. Thierfelder, Thierisches Leben ohne Bakterien im Verdauungskanal (Vie animale sans bactérie dans le tractus digestif). Ztschr f physiol Chem 21, p.22, 1895.

M. R. O'brian and L. Thony-meyer, Biochemistry, regulation and genomics of haem biosynthesis in prokaryotes, Adv Microb Physiol, vol.46, pp.257-318, 2002.

P. S. Oates and A. R. West, Heme in intestinal epithelial cell turnover, differentiation, detoxification, inflammation, carcinogenesis, absorption and motility, World J Gastroenterol, vol.12, pp.4281-4295, 2006.

T. Olczak, A. Sroka, J. Potempa, and M. Olczak, Porphyromonas gingivalis HmuY and HmuR: further characterization of a novel mechanism of heme utilization, Arch Microbiol, vol.189, pp.197-210, 2008.

W. W. Oliver and W. B. Wherry, Notes on some bacterial parasites of the human mucous membranes, J Infectious Deseases, vol.28, pp.2341-2344, 1921.

B. R. Otto, J. G. Kusters, J. Luirink, F. K. De-graaf, and B. Oudega, Molecular characterization of a heme-binding protein of Bacteroides fragilis BE1, Infect Immun, vol.64, pp.4345-4350, 1996.

B. R. Otto, M. Sparrius, D. J. Wors, F. K. De-graaf, and D. M. Maclaren, Utilization of haem from the haptoglobin-haemoglobin complex by Bacteroides fragilis, Microb Pathog, vol.17, pp.137-147, 1994.

B. R. Otto, S. J. Van-dooren, C. M. Dozois, J. Luirink, and B. Oudega, Escherichia coli hemoglobin protease autotransporter contributes to synergistic abscess formation and hemedependent growth of Bacteroides fragilis, Infect Immun, vol.70, pp.5-10, 2002.

C. Palmer, E. M. Bik, D. B. Digiulio, D. A. Relman, and P. O. Brown, Development of the human infant intestinal microbiota, PLoS Biol, vol.5, p.177, 2007.

N. Pan and J. A. Imlay, How does oxygen inhibit central metabolism in the obligate anaerobe Bacteroides thetaiotaomicron, Mol Microbiol, vol.39, pp.1562-1571, 2001.

D. S. Parker, The measurement of production rates of volatile fatty acids in the caecum of the conscious rabbit, Br J Nutr, vol.36, pp.61-70, 1976.

D. Perkins-balding, M. Ratliff-griffin, and I. Stojiljkovic, Iron transport systems in Neisseria meningitidis, Microbiol Mol Biol Rev, vol.68, pp.154-171, 2004.

F. Pierre, S. Tache, C. R. Petit, R. Van-der-meer, and D. E. Corpet, Meat and cancer: haemoglobin and haemin in a low-calcium diet promote colorectal carcinogenesis at the aberrant crypt stage in rats, Carcinogenesis, vol.24, pp.1683-1690, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00411766

K. A. Pittman and M. P. Bryant, Peptides and Other Nitrogen Sources for Growth of Bacteroides Ruminicola, J Bacteriol, vol.88, pp.401-410, 1964.

C. T. Privalle and E. M. Gregory, Superoxide dismutase and O2 lethality in Bacteroides fragilis, J Bacteriol, vol.138, pp.139-145, 1979.

L. Pumbwe, C. A. Skilbeck, V. Nakano, M. J. Avila-campos, R. M. Piazza et al., Bile salts enhance bacterial co-aggregation, bacterial-intestinal epithelial cell adhesion, biofilm formation and antimicrobial resistance of Bacteroides fragilis, Microb Pathog, vol.43, pp.78-87, 2007.

J. Qin, R. Li, J. Raes, M. Arumugam, K. S. Burgdorf et al., A human gut microbial gene catalogue established by metagenomic sequencing, Nature, vol.464, pp.59-65, 2010.
URL : https://hal.archives-ouvertes.fr/cea-00908974

J. Qin, Y. Li, Z. Cai, S. Li, J. Zhu et al., , 2012.

, A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature, vol.490, pp.55-60

G. Quinto, Nutrition of five Bacteroides strains, J Bacteriol, vol.84, pp.559-562, 1962.

R. Rai, V. A. Saraswat, and R. K. Dhiman, Gut microbiota: its role in hepatic encephalopathy, J Clin Exp Hepatol, vol.5, pp.29-36, 2015.

A. R. Reeves, J. N. Elia, J. Frias, and A. A. Salyers, A Bacteroides thetaiotaomicron outer membrane protein that is essential for utilization of maltooligosaccharides and starch, J Bacteriol, vol.178, pp.823-830, 1996.

A. R. Reeves, G. R. Wang, and A. A. Salyers, Characterization of four outer membrane proteins that play a role in utilization of starch by Bacteroides thetaiotaomicron, J Bacteriol, vol.179, pp.643-649, 1997.

S. P. Riepe, J. Goldstein, A. , and D. H. , Effect of secreted Bacteroides proteases on human intestinal brush border hydrolases, J Clin Invest, vol.66, pp.314-322, 1980.

A. Riiser, The human microbiome, asthma, and allergy, Allergy Asthma Clin Immunol, vol.11, p.35, 2015.

K. P. Robertson, C. J. Smith, A. M. Gough, and E. R. Rocha, Characterization of Bacteroides fragilis hemolysins and regulation and synergistic interactions of HlyA and HlyB, Infect Immun, vol.74, pp.2304-2316, 2006.

E. R. Rocha, C. D. Herren, D. J. Smalley, and C. J. Smith, The complex oxidative stress response of Bacteroides fragilis: the role of OxyR in control of gene expression, Anaerobe, vol.9, pp.165-173, 2003.

E. R. Rocha and A. S. Krykunivsky, Anaerobic utilization of Fe(III)-xenosiderophores among Bacteroides species and the distinct assimilation of Fe(III)-ferrichrome by Bacteroides fragilis within the genus, 2017.

E. R. Rocha and C. J. Smith, Role of the alkyl hydroperoxide reductase (ahpCF) gene in oxidative stress defense of the obligate Anaerobe bacteroides fragilis, J Bacteriol, vol.181, pp.5701-5710, 1999.

E. R. Rocha and C. J. Smith, Haem and Iron Metabolism in Bacteroides. Iron uptake and homeostasis in microorganisms, p.153, 2010.

E. R. Rocha and C. J. Smith, Ferritin-like family proteins in the anaerobe Bacteroides fragilis: when an oxygen storm is coming, take your iron to the shelter, Biometals, vol.26, pp.577-591, 2013.

T. Rochat, L. Bermudez-humaran, J. J. Gratadoux, C. Fourage, C. Hoebler et al., Anti-inflammatory effects of Lactobacillus casei BL23 producing or not a manganese-dependant catalase on DSS-induced colitis in mice, Microb Cell Fact, vol.6, p.22, 2007.

D. C. Rockey, Occult gastrointestinal bleeding, N Engl J Med, vol.341, pp.38-46, 1999.

J. M. Rodriguez, The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation?, Adv Nutr, vol.5, pp.779-784, 2014.

M. S. Rossi, A. Paquelin, J. M. Ghigo, and C. Wandersman, Haemophore-mediated signal transduction across the bacterial cell envelope in Serratia marcescens: the inducer and the transported substrate are different molecules, Mol Microbiol, vol.48, pp.1467-1480, 2003.

O. D. Rotstein, T. L. Pruett, C. L. Wells, and R. L. Simmons, The role of Bacteroides encapsulation in the lethal synergy between Escherichia coli and Bacteroides species studied in a rat fibrin clot peritonitis model, J Infect, vol.15, pp.135-146, 1987.

O. D. Rotstein, C. L. Wells, T. L. Pruett, J. J. Sorenson, and R. L. Simmons, Succinic acid production by Bacteroides fragilis. A potential bacterial virulence factor, Arch Surg, vol.122, pp.93-98, 1987.

A. A. Salyers, J. R. Vercellotti, S. E. West, and T. D. Wilkins, Fermentation of mucin and plant polysaccharides by strains of Bacteroides from the human colon, Appl Environ Microbiol, vol.33, pp.319-322, 1977.

T. Sawa, T. Akaike, K. Kida, Y. Fukushima, K. Takagi et al., Lipid peroxyl radicals from oxidized oils and heme-iron: implication of a high-fat diet in colon carcinogenesis, Cancer Epidemiol Biomarkers Prev, vol.7, pp.1007-1012, 1998.

R. W. Schaedler, R. Dubos, and R. Costello, Association of Germfree Mice with Bacteria Isolated from Normal Mice, J Exp Med, vol.122, pp.77-82, 1965.

T. D. Schiano, Treatment options for hepatic encephalopathy, Pharmacotherapy, vol.30, pp.16-21, 2010.

M. Schottelius, Die Bedeutung der Darmbakterien für die Ernährung (L'importance des bactéries intestinale dans la nutrition), Archiv für Hygiene, vol.34, p.210, 1899.

I. Schroder, E. Johnson, and S. De-vries, Microbial ferric iron reductases, FEMS Microbiol Rev, vol.27, pp.427-447, 2003.

H. Schwabacher, D. R. Lucas, R. , and C. , Bacterium melaninogenicum; a misnomer, J Gen Microbiol, vol.1, pp.109-120, 1947.

S. Schwartz, J. Dahl, M. Ellefson, A. , and D. , The "HemoQuant" test: a specific and quantitative determination of heme (hemoglobin) in feces and other materials, Clin Chem, vol.29, pp.2061-2067, 1983.

C. L. Sears, The toxins of Bacteroides fragilis, Toxicon, vol.39, pp.1737-1746, 2001.

I. Sekirov, S. L. Russell, L. C. Antunes, and B. B. Finlay, Gut microbiota in health and disease, Physiol Rev, vol.90, pp.859-904, 2010.

P. Seksik, H. Sokol, P. Lepage, N. Vasquez, C. Manichanh et al., Review article: the role of bacteria in onset and perpetuation of inflammatory bowel disease, Aliment Pharmacol Ther, vol.24, pp.11-18, 2006.

R. Sender, S. Fuchs, M. , and R. , Revised Estimates for the Number of Human and Bacteria Cells in the Body, PLoS Biol, vol.14, p.1002533, 2016.

A. L. Sesink, D. S. Termont, J. H. Kleibeuker, and R. Van-der-meer, Red meat and colon cancer: the cytotoxic and hyperproliferative effects of dietary heme, Cancer Res, vol.59, pp.5704-5709, 1999.

A. Sharff, C. Fanutti, J. Shi, C. Calladine, and B. Luisi, The role of the TolC family in protein transport and multidrug efflux. From stereochemical certainty to mechanistic hypothesis, Eur J Biochem, vol.268, pp.5011-5026, 2001.

G. Sharon, T. R. Sampson, D. H. Geschwind, and S. K. Mazmanian, The Central Nervous System and the Gut Microbiome, Cell, vol.167, pp.915-932, 2016.

J. R. Sheldon and D. E. Heinrichs, Recent developments in understanding the iron acquisition strategies of gram positive pathogens, FEMS Microbiol Rev, vol.39, pp.592-630, 2015.

J. W. Smalley and T. Olczak, Heme acquisition mechanisms of Porphyromonas gingivalis -strategies used in a polymicrobial community in a heme-limited host environment, Mol Oral Microbiol, vol.32, pp.1-23, 2017.

M. L. Snyder, The bacterial flora of meconium specimens collected from sixty-four infants within four hours after delivery, J Pediatrics, vol.9, pp.624-632, 1936.

H. Sokol, B. Pigneur, L. Watterlot, O. Lakhdari, L. G. Bermudez-humaran et al., Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Proc Natl Acad Sci U S A, vol.105, pp.16731-16736, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00652961

J. L. Sonnenburg, J. Xu, D. D. Leip, C. H. Chen, B. P. Westover et al., Glycan foraging in vivo by an intestine-adapted bacterial symbiont, Science, vol.307, pp.1955-1959, 2005.

J. F. Sperry, M. D. Appleman, and T. D. Wilkins, Requirement of heme for growth of Bacteroides fragilis, Appl Environ Microbiol, vol.34, pp.386-390, 1977.

A. Spor, O. Koren, and R. Ley, Unravelling the effects of the environment and host genotype on the gut microbiome, Nat Rev Microbiol, vol.9, pp.279-290, 2011.

D. L. Stauff, V. J. Torres, and E. P. Skaar, Signaling and DNA-binding activities of the Staphylococcus aureus HssR-HssS two-component system required for heme sensing, J Biol Chem, vol.282, pp.26111-26121, 2007.

B. Stecher and W. D. Hardt, The role of microbiota in infectious disease, Trends Microbiol, vol.16, pp.107-114, 2008.

L. F. Stinson, M. S. Payne, and J. A. Keelan, Planting the seed: Origins, composition, and postnatal health significance of the fetal gastrointestinal microbiota, Critical Reviews in Microbiology, pp.1-18, 2016.

I. Stojiljkovic, B. D. Evavold, and V. Kumar, Antimicrobial properties of porphyrins, Expert Opin Investig Drugs, vol.10, pp.309-320, 2001.

A. Suau, R. Bonnet, M. Sutren, J. J. Godon, G. R. Gibson et al., Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut, Appl Environ Microbiol, vol.65, pp.4799-4807, 1999.

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, J Physiol, vol.558, pp.263-275

C. J. Sund, E. R. Rocha, A. O. Tzianabos, W. G. Wells, J. M. Gee et al., The Bacteroides fragilis transcriptome response to oxygen and H2O2: the role of OxyR and its effect on survival and virulence, Mol Microbiol, vol.67, pp.129-142, 2008.

A. Swidsinski, J. Weber, V. Loening-baucke, L. P. Hale, and H. Lochs, Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease, J Clin Microbiol, vol.43, pp.3380-3389, 2005.

F. P. Tally, P. R. Stewart, V. L. Sutter, and J. E. Rosenblatt, Oxygen tolerance of fresh clinical anaerobic bacteria, J Clin Microbiol, vol.1, pp.161-164, 1975.

Y. P. Tang, M. M. Dallas, and M. H. Malamy, Characterization of the Batl (Bacteroides aerotolerance) operon in Bacteroides fragilis: isolation of a B. fragilis mutant with reduced aerotolerance and impaired growth in in vivo model systems, Mol Microbiol, vol.32, pp.139-149, 1999.

H. Tissier, Recherches sur la flore intestinale des nourissons : état normal et pathologique: G. Carré et C, 1900.

J. Tomas, J. Reygner, C. Mayeur, R. Ducroc, S. Bouet et al., Early colonizing Escherichia coli elicits remodeling of rat colonic epithelium shifting toward a new homeostatic state, ISME J, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01194140

J. Tomas, L. Wrzosek, N. Bouznad, S. Bouet, C. Mayeur et al., Primocolonization is associated with colonic epithelial maturation during conventionalization, FASEB J, vol.27, pp.645-655, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01003327

N. U. Toprak, A. Yagci, B. M. Gulluoglu, M. L. Akin, P. Demirkalem et al., A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer, Clin Microbiol Infect, vol.12, pp.782-786, 2006.

E. Turlin, G. Heuck, M. I. Simoes-brandao, N. Szili, J. R. Mellin et al., Protoporphyrin (PPIX) efflux by the MacAB-TolC pump in Escherichia coli, vol.3, pp.849-859, 2014.

A. O. Tzianabos, A. B. Onderdonk, B. Rosner, R. L. Cisneros, and D. L. Kasper, Structural features of polysaccharides that induce intra-abdominal abscesses, Science, vol.262, pp.416-419, 1993.

P. Väänänen and R. Tenhunen, Rapid immunochemical detection of fecal occult blood by use of a latex-agglutination test, Clin Chem, vol.34, pp.1763-1766, 1988.

J. M. Van-camp, V. Tomaselli, and A. G. Coran, Bacterial translocation in the neonate, Curr Opin Pediatr, vol.6, pp.327-333, 1994.

J. W. Van-den-berg, R. Koole-lesuis, A. Edixhoven-bosdijk, and N. Brouwers, Automating the quantification of heme in feces, Clin Chem, vol.34, pp.2125-2126, 1988.

L. W. Van-den-elsen, H. C. Poyntz, L. S. Weyrich, W. Young, and E. E. Forbes-blom, Embracing the gut microbiota: the new frontier for inflammatory and infectious diseases, Clin Transl Immunology, vol.6, p.125, 2017.

M. C. Van-lanschot, B. Carvalho, V. M. Coupe, M. Van-engeland, E. Dekker et al., Molecular stool testing as an alternative for surveillance colonoscopy: a cross-sectional cohort study, BMC Cancer, vol.17, p.116, 2017.

V. H. Varel and M. P. Bryant, Nutritional features of Bacteroides fragilis subsp. fragilis, Appl Microbiol, vol.28, pp.251-257, 1974.

N. Vasquez, I. Mangin, P. Lepage, P. Seksik, J. P. Duong et al., Patchy distribution of mucosal lesions in ileal Crohn's disease is not linked to differences in the dominant mucosa-associated bacteria: a study using fluorescence in situ hybridization and temporal temperature gradient gel electrophoresis, Inflamm Bowel Dis, vol.13, pp.684-692, 2007.

A. Veillon and A. Zuber, Recherches sur quelques microbes strictements anaérobies et leur rôle en pathologie, Arch Med Exp Anat Pathol Serie, vol.1, p.517, 1898.

J. M. Vieira, S. H. Seabra, D. C. Vallim, M. A. Americo, S. E. Fracallanza et al., Bacteroides fragilis induce necrosis on mice peritoneal macrophages: In vitro and in vivo assays, Biochem Biophys Res Commun, vol.387, pp.627-632, 2009.

J. M. Vieira, D. C. Vallim, E. O. Ferreira, S. H. Seabra, R. C. Vommaro et al., Bacteroides fragilis interferes with iNOS activity and leads to pore formation in macrophage surface, Biochem Biophys Res Commun, vol.326, pp.607-613, 2005.

N. F. Villarino, G. R. Lecleir, J. E. Denny, S. P. Dearth, C. L. Harding et al., Composition of the gut microbiota modulates the severity of malaria, Proc Natl Acad Sci U S A, vol.113, pp.2235-2240, 2016.

S. L. Vogt, J. Pena-diaz, and B. B. Finlay, Chemical communication in the gut: Effects of microbiota-generated metabolites on gastrointestinal bacterial pathogens, Anaerobe, vol.34, pp.106-115, 2015.

A. Vrieze, E. Van-nood, F. Holleman, J. Salojarvi, R. S. Kootte et al., Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome, Gastroenterology, vol.143, pp.913-916, 2012.

H. E. Vuong and E. Y. Hsiao, Emerging Roles for the Gut Microbiome in Autism Spectrum Disorder, Biol Psychiatry, vol.81, pp.411-423, 2017.

C. Wandersman and P. Delepelaire, Bacterial iron sources: from siderophores to hemophores, Annu Rev Microbiol, vol.58, pp.611-647, 2004.

Z. Wang, E. Klipfell, B. J. Bennett, R. Koeth, B. S. Levison et al., Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature, vol.472, pp.57-63, 2011.

H. M. Wexler, Bacteroides: the good, the bad, and the nitty-gritty, Clin Microbiol Rev, vol.20, pp.593-621, 2007.

K. H. Wilson and R. B. Blitchington, Human colonic biota studied by ribosomal DNA sequence analysis, Appl Environ Microbiol, vol.62, pp.2273-2278, 1996.

C. R. Woese and G. E. Fox, Phylogenetic structure of the prokaryotic domain: the primary kingdoms, Proc Natl Acad Sci U S A, vol.74, pp.5088-5090, 1977.

H. Wojtowicz, T. Guevara, C. Tallant, M. Olczak, A. Sroka et al., Unique structure and stability of HmuY, a novel heme-binding protein of Porphyromonas gingivalis, PLoS Pathog, vol.5, 2009.

S. Wollers, G. Layer, R. Garcia-serres, L. Signor, M. Clemancey et al., Iron-sulfur (Fe-S) cluster assembly: the SufBCD complex is a new type of Fe-S scaffold with a flavin redox cofactor, J Biol Chem, vol.285, pp.23331-23341, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01053728

A. Woting and M. Blaut, The Intestinal Microbiota in Metabolic Disease, Nutrients, vol.8, 2016.

C. Y. Wu, L. G. Bermudez-humaran, F. Yue, M. Li, and L. P. Zhang, Intranasal administration with recombinant Lactococcus lactis expressing heme oxygenase-1 reduces hyperoxia-induced lung inflammation in rat pups, Biotechnol Lett, vol.37, pp.1203-1211, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01535267

G. D. Wu, J. Chen, C. Hoffmann, K. Bittinger, Y. Y. Chen et al., Linking long-term dietary patterns with gut microbial enterotypes, Science, vol.334, pp.105-108, 2011.

S. Wu, K. C. Lim, J. Huang, R. F. Saidi, and C. L. Sears, Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin, Proc Natl Acad Sci U S A, vol.95, pp.14979-14984, 1998.

J. Xu, M. K. Bjursell, J. Himrod, S. Deng, L. K. Carmichael et al., A genomic view of the human-Bacteroides thetaiotaomicron symbiosis, Science, vol.299, pp.2074-2076, 2003.

J. Xu, G. , and J. I. , Honor thy symbionts, Proc Natl Acad Sci U S A, vol.100, pp.10452-10459, 2003.

Y. Yamamoto, C. Poyart, P. Trieu-cuot, G. Lamberet, A. Gruss et al., Respiration metabolism of Group B Streptococcus is activated by environmental haem and quinone and contributes to virulence, Mol Microbiol, vol.56, pp.525-534, 2005.

S. Zambolin, B. Clantin, M. Chami, S. Hoos, A. Haouz et al., Structural basis for haem piracy from host haemopexin by Haemophilus influenzae, Nat Commun, vol.7, p.11590, 2016.

H. I. Zgurskaya, G. Krishnamoorthy, A. Ntreh, and S. Lu, Mechanism and Function of the Outer Membrane Channel TolC in Multidrug Resistance and Physiology of Enterobacteria, Front Microbiol, vol.2, p.189, 2011.