A. Ait-belgnaoui, H. Durand, C. Cartier, G. Chaumaz, H. Eutamene et al., Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats, Psychoneuroendocrinology, vol.37, pp.1885-1895, 2012.

A. Ait-belgnaoui, A. Colom, V. Braniste, L. Ramalho, A. Marrot et al., Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.510-520, 2014.

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, 2008.

F. Barreau, L. Ferrier, J. Fioramonti, and L. Bueno, New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models, Pediatr. Res, vol.62, pp.240-245, 2007.

P. Bercik, E. F. Verdu, J. A. Foster, J. Macri, M. Potter et al., Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice, Gastroenterology, vol.139, pp.2102-2112, 2010.

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, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.23, pp.1132-1139, 2011.

N. A. Bokulich, S. Subramanian, J. J. Faith, D. Gevers, J. I. Gordon et al., Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing, Nat. Methods, vol.10, pp.57-59, 2013.

K. Bradford, W. Shih, E. J. Videlock, A. P. Presson, B. D. Naliboff et al., Association Between Early Adverse Life Events and Irritable Bowel Syndrome, Clin. Gastroenterol. Hepatol, vol.10, pp.385-390, 2012.

I. Branchi, D. Santucci, and E. Alleva, Ultrasonic vocalisation emitted by infant rodents: a tool for assessment of neurobehavioural development, Behav. Brain Res, vol.125, pp.49-56, 2001.

A. Burokas, R. D. Moloney, T. G. Dinan, and J. F. Cryan, Microbiota regulation of the Mammalian gutbrain axis, Adv. Appl. Microbiol, vol.91, pp.1-62, 2015.

D. P. Chapman, C. L. Whitfield, V. J. Felitti, S. R. Dube, V. J. Edwards et al., Adverse childhood experiences and the risk of depressive disorders in adulthood, J. Affect. Disord, vol.82, pp.217-225, 2004.

D. K. Chitkara, M. A. Van-tilburg, N. Blois-martin, and W. E. Whitehead, Early life risk factors that contribute to irritable bowel syndrome in adults: a systematic review, Am. J. Gastroenterol, vol.103, pp.765-774, 2008.

L. Crouzet, E. Gaultier, C. Del'homme, C. Cartier, E. Delmas et al., The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.25, pp.272-282, 2013.

G. De-palma, P. Blennerhassett, J. Lu, Y. Deng, A. J. Park et al., Microbiota and host determinants of behavioural phenotype in maternally separated mice, Nat. Commun, vol.6, p.7735, 2015.

L. Desbonnet, L. Garrett, G. Clarke, B. Kiely, J. F. Cryan et al., Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression, Neuroscience, vol.170, pp.1179-1188, 2010.

T. Dinan and J. Cryan, Regulation of the stress response by the gut microbiota: implications for psychoneuroendocrinology, 2012.

D. Prado, C. H. Narahari, T. Holland, F. H. Lee, H. Murthy et al., Effects of early adolescent environmental enrichment on cognitive dysfunction, prefrontal cortex development, and inflammatory cytokines after early life stress, Dev. Psychobiol, 2015.

S. El-aidy, A. S. Ramsteijn, F. Dini-andreote, R. Van-eijk, D. J. Houwing et al., Serotonin Transporter Genotype Modulates the Gut Microbiota Composition in Young Rats, an Effect Augmented by Early Life Stress, Front. Cell. Neurosci, vol.11, p.222, 2017.

F. Escudie, L. Auer, M. Bernard, L. Cauquil, K. Vidal et al., FROGS: Find Rapidly OTU with Galaxy Solution, The Environmental Genomic Conference, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01886442

D. G. Folks, The interface of psychiatry and irritable bowel syndrome, Curr. Psychiatry Rep, vol.6, pp.210-215, 2004.

J. A. Foster, M. Neufeld, and K. , Gut-brain axis: how the microbiome influences anxiety and depression, Trends Neurosci, vol.36, pp.305-312, 2013.

P. J. Mcmurdie and S. Holmes, Phyloseq: An R Package for Reproducible Interactive Analysis and Graphics of Microbiome Census Data, PLoS ONE, vol.8, 2013.

P. J. Mcmurdie and S. Holmes, Waste Not, Want Not: Why Rarefying Microbiome Data Is Inadmissible, PLoS Comput. Biol, vol.10, 2014.

N. Moussaoui, V. Braniste, A. Ait-belgnaoui, M. Gabanou, S. Sekkal et al., Changes in intestinal glucocorticoid sensitivity in early life shape the risk of epithelial barrier defect in maternal-deprived rats, PloS One, vol.9, p.88382, 2014.

A. Moya-pérez, A. Perez-villalba, A. Benítez-páez, I. Campillo, and Y. Sanz, Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice, Brain. Behav. Immun, 2017.

T. Murakami, K. Kamada, K. Mizushima, Y. Higashimura, K. Katada et al., Changes in Intestinal Motility and Gut Microbiota Composition in a Rat Stress Model, Digestion, vol.95, pp.55-60, 2017.

S. M. O'mahony, N. P. Hyland, T. G. Dinan, and J. F. Cryan, Maternal separation as a model of brain-gut axis dysfunction, Psychopharmacology (Berl.), vol.214, pp.71-88, 2011.

G. Paxinos and C. Watson, The Rat Brain in Stereotaxic Coordinates, 2013.

M. Pigrau, B. K. Rodiño-janeiro, M. Casado-bedmar, B. Lobo, M. Vicario et al., The joint power of sex and stress to modulate brain-gut-microbiota axis and intestinal barrier homeostasis: implications for irritable bowel syndrome, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.28, pp.463-486, 2016.

R. M. Pinheiro, M. N. De-lima, B. C. Portal, S. B. Busato, L. Falavigna et al., Long-lasting recognition memory impairment and alterations in brain levels of cytokines and BDNF induced by maternal deprivation: effects of valproic acid and topiramate, J. Neural Transm, 2014.

M. M. Pusceddu, S. El-aidy, F. Crispie, O. O'sullivan, P. Cotter et al., N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota, PloS One, vol.10, 2015.

R. Development-core and R. Team, R: A Language and Environment for Statistical Computing. R Found, Stat. Comput, vol.1, p.409, 2011.

J. Reunanen, V. Kainulainen, L. Huuskonen, N. Ottman, C. Belzer et al., Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer, Appl. Environ. Microbiol, vol.81, pp.3655-3662, 2015.

G. Z. Réus, G. C. Fernandes, A. B. De-moura, R. H. Silva, A. C. Darabas et al., Early life experience contributes to the developmental programming of depressive-like behaviour, neuroinflammation and oxidative stress, J. Psychiatr. Res, vol.95, pp.196-207, 2017.

M. Rincel, A. L. Lépinay, P. Delage, J. Fioramonti, V. S. Théodorou et al., Maternal high-fat diet prevents developmental programming by early-life stress, Transl. Psychiatry, vol.6, p.966, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602432

A. C. Rios, P. K. Maurya, M. Pedrini, M. Zeni-graiff, E. Asevedo et al., Microbiota abnormalities and the therapeutic potential of probiotics in the treatment of mood disorders, Rev. Neurosci, 2017.

S. Roque, A. R. Mesquita, J. A. Palha, N. Sousa, and M. Correia-neves, The Behavioral and Immunological Impact of Maternal Separation: A Matter of Timing, Front. Behav. Neurosci, vol.8, 2014.

H. M. Savignac, B. Kiely, T. G. Dinan, and J. F. Cryan, Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.1615-1627, 2014.

J. Schindelin, I. Arganda-carreras, E. Frise, V. Kaynig, M. Longair et al., Fiji: an open-source platform for biological-image analysis, Nat. Methods, vol.9, pp.676-682, 2012.

N. Segata, J. Izard, L. Waldron, D. Gevers, L. Miropolsky et al., Metagenomic biomarker discovery and explanation, Genome Biol, vol.12, p.60, 2011.

A. Wieck, S. L. Andersen, H. C. Brenhouse, . Supplementary-references, N. A. Bokulich et al., Evidence for a neuroinflammatory mechanism in delayed effects of early life adversity in rats: Relationship to cortical NMDA receptor expression, Brain. Behav. Immun, vol.28, pp.57-59, 2013.

B. J. Callahan, K. Sankaran, J. A. Fukuyama, P. J. Mcmurdie, and S. P. Holmes, Bioconductor Workflow for Microbiome Data Analysis: from raw reads to community analyses, 1492.

C. Camacho, G. Coulouris, V. Avagyan, N. Ma, J. Papadopoulos et al., BLAST+: architecture and applications, BMC Bioinformatics, vol.10, p.421, 2009.

C. Deroulers, D. Ameisen, M. Badoual, C. Gerin, A. Granier et al., Analyzing huge pathology images with open source software, Diagn. Pathol, vol.8, p.92, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00842931

V. F. Labrousse, L. Costes, A. Aubert, M. Darnaudéry, G. Ferreira et al., Impaired interleukin-1beta and c-Fos expression in the hippocampus is associated with a spatial memory deficit in P2X(7) receptor-deficient mice, PloS One, vol.4, p.6006, 2009.

M. I. Love, W. Huber, A. , and S. , Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

T. Mago? and S. Salzberg, FLASH: fast length adjustment of short reads to improve genome assemblies, 2011.

F. Mahé, T. Rognes, C. Quince, C. De-vargas, and M. Dunthorn, Swarm: robust and fast clustering method for amplicon-based studies, PeerJ, vol.2, p.593, 2014.

P. J. Mcmurdie and S. Holmes, Waste Not, Want Not: Why Rarefying Microbiome Data Is Inadmissible, PLoS Comput. Biol, vol.10, 2014.

G. Paxinos, C. Watson, M. Pennisi, and A. Topple, Bregma, lambda and the interaural midpoint in stereotaxic surgery with rats of different sex, strain and weight, J. Neurosci. Methods, vol.13, pp.139-143, 1985.

M. Rincel, A. L. Lépinay, P. Delage, J. Fioramonti, V. S. Théodorou et al., Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats, R60. REFERENCES Ait-Belgnaoui, vol.6, pp.1885-1895, 2011.

C. Belzung and G. Griebel, Measuring normal and pathological anxiety-like behaviour in mice: a review, Behav. Brain Res, vol.125, pp.141-149, 2001.

S. C. Bischoff, G. Barbara, W. Buurman, T. Ockhuizen, J. Schulzke et al., Intestinal permeability--a new target for disease prevention and therapy, BMC Gastroenterol, vol.14, p.189, 2014.

S. A. Blair, S. V. Kane, D. R. Clayburgh, and J. R. Turner, Epithelial myosin light chain kinase expression and activity are upregulated in inflammatory bowel disease, Lab. Investig. J. Tech. Methods Pathol, vol.86, pp.191-201, 2006.

S. R. Bornstein, A. Schuppenies, M. L. Wong, and J. Licinio, Approaching the shared biology of obesity and depression: the stress axis as the locus of gene-environment interactions, Mol. Psychiatry, vol.11, pp.892-902, 2006.

K. Bradford, W. Shih, E. J. Videlock, A. P. Presson, B. D. Naliboff et al., Association Between Early Adverse Life Events and Irritable Bowel Syndrome, Clin. Gastroenterol. Hepatol, vol.10, pp.385-390, 2012.

V. Braniste, M. Al-asmakh, C. Kowal, F. Anuar, A. Abbaspour et al., The gut microbiota influences blood-brain barrier permeability in mice, Sci. Transl. Med, vol.6, pp.263-158, 2014.

T. Buie, D. B. Campbell, G. J. Fuchs, G. T. Furuta, J. Levy et al., Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report, Pediatrics, vol.125, issue.1, pp.1-18, 2010.

A. Burokas, R. D. Moloney, T. G. Dinan, and J. F. Cryan, Microbiota regulation of the Mammalian gut-brain axis, Adv. Appl. Microbiol, vol.91, pp.1-62, 2015.

A. Burokas, S. Arboleya, R. D. Moloney, V. L. Peterson, K. Murphy et al., Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice, Biol. Psychiatry, vol.82, pp.472-487, 2017.

N. G. Cascella, D. Kryszak, B. Bhatti, P. Gregory, D. L. Kelly et al., Prevalence of celiac disease and gluten sensitivity in the United States clinical antipsychotic trials of intervention effectiveness study population, Schizophr. Bull, vol.37, pp.94-100, 2011.

D. K. Chitkara, M. A. Van-tilburg, N. Blois-martin, and W. E. Whitehead, Early life risk factors that contribute to irritable bowel syndrome in adults: a systematic review, Am. J. Gastroenterol, vol.103, pp.765-774, 2008.

D. R. Clayburgh, T. A. Barrett, Y. Tang, J. B. Meddings, L. J. Van-eldik et al., Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo, J. Clin. Invest, vol.115, pp.2702-2715, 2005.

C. D. Conrad, L. A. Galea, Y. Kuroda, and B. S. Mcewen, Chronic stress impairs rat spatial memory on the Y maze, and this effect is blocked by tianeptine pretreatment, Behav. Neurosci, vol.110, pp.1321-1334, 1996.

L. F. De-cossío, C. Fourrier, J. Sauvant, A. Everard, L. Capuron et al., Impact of prebiotics on metabolic and behavioral alterations in a mouse model of metabolic syndrome, Brain. Behav. Immun, vol.64, pp.33-49, 2017.

N. A. Datson, M. C. Morsink, O. C. Meijer, and E. R. De-kloet, Central corticosteroid actions: Search for gene targets, Eur. J. Pharmacol, vol.583, pp.272-289, 2008.

G. De-palma, P. Blennerhassett, J. Lu, Y. Deng, A. J. Park et al., Microbiota and host determinants of behavioural phenotype in maternally separated mice, Nat. Commun, vol.6, p.7735, 2015.

C. Deroulers, D. Ameisen, M. Badoual, C. Gerin, A. Granier et al., Analyzing huge pathology images with open source software, Diagn. Pathol, vol.8, p.92, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00842931

P. D'eufemia, M. Celli, R. Finocchiaro, L. Pacifico, L. Viozzi et al., Abnormal intestinal permeability in children with autism, Acta Paediatr. Oslo Nor, vol.85, pp.1076-1079, 1992.

T. Dinan and J. Cryan, Regulation of the stress response by the gut microbiota: implications for psychoneuroendocrinology, 2012.

P. Luczynski, K. Mcvey-neufeld, C. S. Oriach, G. Clarke, T. G. Dinan et al., Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior, Int. J. Neuropsychopharmacol, vol.19, 2016.

F. S. Luppino, L. M. De-wit, P. F. Bouvy, T. Stijnen, P. Cuijpers et al., Overweight, obesity, and depression: a systematic review and meta-analysis of longitudinal studies, Arch. Gen. Psychiatry, vol.67, pp.220-229, 2010.

M. Maes, M. Kubera, and J. Leunis, The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression, Neuro Endocrinol. Lett, vol.29, pp.117-124, 2008.

L. De-magistris, V. Familiari, A. Pascotto, A. Sapone, A. Frolli et al., Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives, J. Pediatr. Gastroenterol. Nutr, vol.51, pp.418-424, 2010.

O. Malkesman, M. L. Scattoni, D. Paredes, T. Tragon, B. Pearson et al., The Female Urine Sniffing Test: A Novel Approach for Assessing Reward-Seeking Behavior in Rodents, Biol. Psychiatry, vol.67, pp.864-871, 2010.

J. G. Markle, D. N. Frank, S. Mortin-toth, C. E. Robertson, L. M. Feazel et al., Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity, Science, vol.339, pp.1084-1088, 2013.

M. Nébot-vivinus, C. Harkat, H. Bzioueche, C. Cartier, R. Plichon-dainese et al., Multispecies probiotic protects gut barrier function in experimental models, World J. Gastroenterol, vol.20, pp.6832-6843, 2014.

B. D. Oladeji and O. Gureje, The comorbidity between depression and diabetes, Curr. Psychiatry Rep, vol.15, p.390, 2013.

G. Paxinos, F. , and K. , Paxinos and Franklin's the Mouse Brain in Stereotaxic Coordinates, 2001.

J. Reunanen, V. Kainulainen, L. Huuskonen, N. Ottman, C. Belzer et al., Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer, Appl. Environ. Microbiol, vol.81, pp.3655-3662, 2015.

E. M. Richard, J. 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.

M. Rincel, A. L. Lépinay, P. Delage, J. Fioramonti, V. S. Théodorou et al., Maternal high-fat diet prevents developmental programming by early-life stress, Transl. Psychiatry, vol.6, p.966, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602432

A. C. Rios, P. K. Maurya, M. Pedrini, M. Zeni-graiff, E. Asevedo et al., Microbiota abnormalities and the therapeutic potential of probiotics in the treatment of mood disorders, Rev. Neurosci, 2017.

L. Shen, E. D. Black, E. D. Witkowski, W. I. Lencer, V. Guerriero et al., Myosin light chain phosphorylation regulates barrier function by remodeling tight junction structure, J. Cell Sci, vol.119, pp.2095-2106, 2006.

A. Slyepchenko, M. Maes, R. Machado-vieira, G. Anderson, M. Solmi et al., Intestinal Dysbiosis, Gut Hyperpermeability and Bacterial Translocation: Missing Links Between Depression, Obesity and Type 2 Diabetes, Curr. Pharm. Des, vol.22, pp.6087-6106, 2016.

B. R. Stevens, R. Goel, K. Seungbum, E. M. Richards, R. C. Holbert et al., Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression, 2017.

L. Su, L. Shen, D. R. Clayburgh, S. C. Nalle, E. A. Sullivan et al., Targeted Epithelial Tight Junction Dysfunction Causes Immune Activation and Contributes to Development of Experimental Colitis, Gastroenterology, vol.136, pp.551-563, 2009.

P. De-timary, S. Leclercq, P. Stärkel, and N. Delzenne, A dysbiotic subpopulation of alcoholdependent subjects, Gut Microbes, vol.6, pp.388-391, 2015.

L. Van-oudenhove, H. Törnblom, S. Störsrud, J. Tack, and M. Simrén, Depression and Somatization Are Associated With Increased Postprandial Symptoms in Patients With Irritable Bowel Syndrome, Gastroenterology, vol.150, pp.866-874, 2016.

K. Wilpart, H. Törnblom, J. Svedlund, J. F. Tack, M. Simrén et al., Coping Skills Are Associated With Gastrointestinal Symptom Severity and Somatization in Patients With Irritable Bowel Syndrome, Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc, vol.15, pp.1565-1571, 2017.

M. Altemus, Sex differences in depression and anxiety disorders: potential biological determinants, Horm. Behav, vol.50, pp.534-538, 2006.

Y. Aoyama, A. Mouri, K. Toriumi, T. Koseki, S. Narusawa et al., Clozapine ameliorates epigenetic and behavioral abnormalities induced by phencyclidine through activation of dopamine D1 receptor, Int. J. Neuropsychopharmacol, vol.17, pp.723-737, 2014.

K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara et al., Induction of colonic regulatory T cells by indigenous Clostridium species, Science, vol.331, pp.337-341, 2011.

F. Barreau, L. Ferrier, J. Fioramonti, and L. Bueno, New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models, Pediatr. Res, vol.62, pp.240-245, 2007.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, J. R. Stat. Soc. Ser. B, 1995.

P. Bercik, E. F. Verdu, J. A. Foster, J. Macri, M. Potter et al., Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice, Gastroenterology, vol.139, pp.2102-2112, 2010.

B. M. Bolstad, R. A. Irizarry, M. Astrand, and T. P. Speed, A comparison of normalization methods for high density oligonucleotide array data based on variance and bias, Bioinforma. Oxf. Engl, vol.19, pp.185-193, 2003.

Y. E. Borre, G. W. O'keeffe, G. Clarke, C. Stanton, T. G. Dinan et al., Microbiota and neurodevelopmental windows: implications for brain disorders, Trends Mol. Med, vol.20, pp.509-518, 2014.

J. Boué, L. Basso, N. Cenac, C. Blanpied, M. Rolli-derkinderen et al., Endogenous regulation of visceral pain via production of opioids by colitogenic CD4(+) T cells in mice, Gastroenterology, vol.146, pp.166-175, 2014.

J. A. Bravo, P. Forsythe, M. V. Chew, E. Escaravage, H. M. Savignac et al., Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve, Proc. Natl. Acad. Sci, vol.108, pp.16050-16055, 2011.

A. S. Brown, The environment and susceptibility to schizophrenia, Prog. Neurobiol, vol.93, pp.23-58, 2011.

A. S. Brown, A. Sourander, S. Hinkka-yli-salomäki, I. W. Mckeague, J. Sundvall et al., Elevated maternal C-reactive protein and autism in a national birth cohort, Mol. Psychiatry, vol.19, pp.259-264, 2014.

S. A. Buffington, G. V. Di-prisco, T. A. Auchtung, N. J. Ajami, J. F. Petrosino et al., Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring, Cell, vol.165, pp.1762-1775, 2016.

J. R. Cole, Q. Wang, J. A. Fish, B. Chai, D. M. Mcgarrell et al., Ribosomal Database Project: data and tools for high throughput rRNA analysis, Nucleic Acids Res, vol.42, pp.633-642, 2014.

E. M. Cotella, I. Mestres-lascano, L. Franchioni, G. M. Levin, and M. M. Suárez, Long-term effects of maternal separation on chronic stress response suppressed by amitriptyline treatment, Stress Amst. Neth, vol.16, pp.477-481, 2013.

L. Coutellier, S. Beraki, P. M. Ardestani, N. L. Saw, and M. Shamloo, Npas4: a neuronal transcription factor with a key role in social and cognitive functions relevant to developmental disorders, PloS One, vol.7, p.46604, 2012.

L. Coutellier, V. Gilbert, and R. Shepard, Npas4 deficiency increases vulnerability to juvenile stress in mice, Behav. Brain Res, vol.295, pp.17-25, 2015.

G. De-palma, P. Blennerhassett, J. Lu, Y. Deng, A. J. Park et al., Digging and marble burying in mice: simple methods for in vivo identification of biological impacts, Nat. Commun, vol.6, pp.122-124, 2006.

J. Deslauriers, A. Larouche, P. Sarret, and S. Grignon, Combination of prenatal immune challenge and restraint stress affects prepulse inhibition and dopaminergic/GABAergic markers, Prog. Neuropsychopharmacol. Biol. Psychiatry, vol.45, pp.156-164, 2013.

J. Deslauriers, W. Racine, P. Sarret, and S. Grignon, Preventive effect of ?-lipoic acid on prepulse inhibition deficits in a juvenile two-hit model of schizophrenia, Neuroscience, vol.272, pp.261-270, 2014.

A. Dinel, C. André, A. Aubert, G. Ferreira, S. Layé et al., Cognitive and emotional alterations are related to hippocampal inflammation in a mouse model of metabolic syndrome, PloS One, vol.6, 2011.

C. Dominianni, R. Sinha, J. J. Goedert, Z. Pei, L. Yang et al., Sex, body mass index, and dietary fiber intake influence the human gut microbiome, PloS One, vol.10, p.124599, 2015.

R. Edgar, M. Domrachev, and A. E. Lash, Gene Expression Omnibus: NCBI gene expression and hybridization array data repository, Nucleic Acids Res, vol.30, pp.207-210, 2002.

J. R. Emge, K. Huynh, E. N. Miller, M. Kaur, C. Reardon et al., Modulation of the microbiota-gut-brain axis by probiotics in a murine model of inflammatory bowel disease, Am. J. Physiol. Gastrointest. Liver Physiol, vol.310, pp.989-998, 2016.

X. Fang, X. Zhong, G. Yu, S. Shao, Y. et al., Vascular protective effects of KLF2 on A?induced toxicity: Implications for Alzheimer's disease, Brain Res, vol.1663, pp.174-183, 2017.

M. R. Farrell, F. H. Holland, R. M. Shansky, and H. C. Brenhouse, Sex-specific effects of early life stress on social interaction and prefrontal cortex dendritic morphology in young rats, Behav. Brain Res, vol.310, pp.119-125, 2016.

E. Flinkkilä, A. Keski-rahkonen, M. Marttunen, and A. Raevuori, Prenatal Inflammation, Infections and Mental Disorders, Psychopathology, vol.49, pp.317-333, 2016.

K. A. Foley, K. Ossenkopp, M. Kavaliers, and D. F. Macfabe, Pre-and neonatal exposure to lipopolysaccharide or the enteric metabolite, propionic acid, alters development and behavior in adolescent rats in a sexually dimorphic manner, PloS One, vol.9, p.87072, 2014.

K. A. Foley, D. F. Macfabe, A. Vaz, K. Ossenkopp, and M. Kavaliers, Sexually dimorphic effects of prenatal exposure to propionic acid and lipopolysaccharide on social behavior in neonatal, adolescent, and adult rats: implications for autism spectrum disorders, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.39, pp.68-78, 2014.

K. A. Foley, D. F. Macfabe, M. Kavaliers, and K. Ossenkopp, Sexually dimorphic effects of prenatal exposure to lipopolysaccharide, and prenatal and postnatal exposure to propionic acid, on acoustic startle response and prepulse inhibition in adolescent rats: relevance to autism spectrum disorders, Behav. Brain Res, vol.278, pp.244-256, 2015.

E. Fombonne, The prevalence of autism, JAMA, vol.289, pp.87-89, 2003.

F. Fransen, A. A. Van-beek, T. Borghuis, B. Meijer, F. Hugenholtz et al., The Impact of, Gut Microbiota on Gender-Specific Differences in Immunity. Front. Immunol, vol.8, p.754, 2017.

M. Gacias, S. Gaspari, P. G. Santos, S. Tamburini, M. Andrade et al., Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior, 2016.

C. L. García-ródenas, G. E. Bergonzelli, S. Nutten, A. Schumann, C. Cherbut et al., Nutritional approach to restore impaired intestinal 28 barrier function and growth after neonatal stress in rats, J. Pediatr. Gastroenterol. Nutr, vol.43, pp.16-24, 2006.

R. C. Gentleman, V. J. Carey, D. M. Bates, B. Bolstad, M. Dettling et al., Bioconductor: open software development for computational biology and bioinformatics, vol.5, p.80, 2004.

J. Ghia, P. Blennerhassett, and S. M. Collins, Impaired parasympathetic function increases susceptibility to inflammatory bowel disease in a mouse model of depression, J. Clin. Invest, vol.118, pp.2209-2218, 2008.

S. Giovanoli, H. Engler, A. Engler, J. Richetto, M. Voget et al., Stress in puberty unmasks latent neuropathological consequences of prenatal immune activation in mice, Science, vol.339, pp.1095-1099, 2013.

S. Giovanoli, H. Engler, A. Engler, J. Richetto, J. Feldon et al., Preventive effects of minocycline in a neurodevelopmental two-hit model with relevance to schizophrenia, Transl. Psychiatry, vol.6, pp.903-917, 2005.

F. Guida, F. Turco, M. Iannotta, D. De-gregorio, I. Palumbo et al., Antibiotic-induced microbiota perturbation causes gut endocannabinoidome changes, hippocampal neuroglial reorganization and depression in mice, Brain. Behav. Immun, 2017.

C. Hammen, R. Henry, and S. E. Daley, Depression and sensitization to stressors among young women as a function of childhood adversity, J. Consult. Clin. Psychol, vol.68, pp.782-787, 2000.

C. Helmeke, W. Ovtscharoff, G. Poeggel, and K. Braun, Imbalance of immunohistochemically characterized interneuron populations in the adolescent and adult rodent medial prefrontal cortex after repeated exposure to neonatal separation stress, Neuroscience, vol.152, pp.18-28, 2008.

M. R. Herbert, Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders, Curr. Opin. Neurol, vol.23, pp.103-110, 2010.

R. A. Hill, M. Klug, S. Kiss-von-soly, M. D. Binder, A. J. Hannan et al., Sexspecific disruptions in spatial memory and anhedonia in a "two hit" rat model correspond with alterations in hippocampal brain-derived neurotrophic factor expression and signaling: Sex-Specific Effects of Stress on BDNF, Cognition, and Anhedonia. Hippocampus, vol.24, pp.1197-1211, 2014.

E. B. Hollister, C. Gao, and J. Versalovic, Compositional and functional features of the gastrointestinal microbiome and their effects on human health, Gastroenterology, vol.146, pp.1449-1458, 2014.

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.

R. L. Huot, P. M. Plotsky, R. H. Lenox, and R. K. Mcnamara, Neonatal maternal separation reduces hippocampal mossy fiber density in adult Long Evans rats, Brain Res, vol.950, pp.52-63, 2002.

H. E. Jakobsson, A. M. Rodríguez-piñeiro, A. Schütte, A. Ermund, P. Boysen et al., The composition of the gut microbiota shapes the colon mucus barrier, EMBO Rep, vol.16, pp.164-177, 2015.

E. Ja?arevi?, K. E. Morrison, and T. L. Bale, Sex differences in the gut microbiome-brain axis across the lifespan, Philos. Trans. R. Soc. Lond. B. Biol. Sci, vol.371, 2016.

S. Kim, H. Kim, Y. S. Yim, S. Ha, K. Atarashi et al., Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring, Nature, vol.549, pp.528-532, 2017.

M. Klug and M. Van-den-buuse, Chronic cannabinoid treatment during young adulthood induces sex-specific behavioural deficits in maternally separated rats, Behav. Brain Res, vol.233, pp.305-313, 2012.

N. Kratsman, D. Getselter, E. , and E. , Sodium butyrate attenuates social behavior deficits and modifies the transcription of inhibitory/excitatory genes in the frontal cortex of an autism model, Neuropharmacology, vol.102, pp.136-145, 2016.

J. H. Krystal, A. Anticevic, G. J. Yang, G. Dragoi, N. R. Driesen et al., Impaired Tuning of Neural Ensembles and the Pathophysiology of Schizophrenia: A Translational and Computational Neuroscience Perspective, Biol. Psychiatry, vol.81, pp.874-885, 2017.

M. A. Labouesse, W. Langhans, and U. Meyer, Long-term pathological consequences of prenatal infection: beyond brain disorders, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.309, pp.1-12, 2015.

M. A. Labouesse, E. Dong, D. Grayson, A. Guidotti, and U. Meyer, Maternal immune activation induces GAD1 and GAD2, 2015.

V. F. Labrousse, A. Nadjar, C. Joffre, L. Costes, A. Aubert et al., Short-term long chain omega3 diet protects from neuroinflammatory processes and memory impairment in aged mice, PloS One, vol.7, p.36861, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01191175

S. Leclercq, F. M. Mian, A. M. Stanisz, L. B. Bindels, E. Cambier et al., Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior, Nat. Commun, vol.8, p.15062, 2017.

J. Lehmann, H. Russig, J. Feldon, P. , and C. R. , Effect of a single maternal separation at different pup ages on the corticosterone stress response in adult and aged rats, Pharmacol. Biochem. Behav, vol.73, pp.141-145, 2002.

S. Liang, T. Wang, X. Hu, J. Luo, W. Li et al., Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress, Neuroscience, vol.310, pp.561-577, 2015.

Y. Lin, B. L. Bloodgood, J. L. Hauser, A. D. Lapan, A. C. Koon et al., Activity-dependent regulation of inhibitory synapse development by Npas4, Nature, vol.455, pp.1198-1204, 2008.

J. Liu, J. Sun, F. Wang, X. Yu, Z. Ling et al., , 2015.

, Neuroprotective Effects of Clostridium butyricum against Vascular Dementia in Mice via Metabolic Butyrate, BioMed Res. Int, p.412946, 2015.

Y. Liu, W. Liu, C. Wu, Y. Juan, Y. Wu et al., Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naïve adult mice, Brain Res, vol.1631, pp.1-12, 2016.

S. Lundberg, M. Martinsson, I. Nylander, R. , and E. , Altered corticosterone levels and social play behavior after prolonged maternal separation in adolescent male but not female Wistar rats, Horm. Behav, vol.87, pp.137-144, 2017.

M. Ma?kowiak, P. Mordalska, and K. W?dzony, Neuroligins, synapse balance and neuropsychiatric disorders, Pharmacol. Rep. PR, vol.66, pp.830-835, 2014.

J. G. Markle, D. N. Frank, S. Mortin-toth, C. E. Robertson, L. M. Feazel et al., Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity, Science, vol.339, pp.1084-1088, 2013.

D. Mattei, A. Djodari-irani, R. Hadar, A. Pelz, L. F. De-cossío et al., Minocycline rescues decrease in neurogenesis, increase in microglia cytokines and deficits in sensorimotor gating in an animal model of schizophrenia, Brain. Behav. Immun, vol.38, pp.175-184, 2014.

E. A. Mayer, D. Padua, and K. Tillisch, Altered brain-gut axis in autism: comorbidity or causative mechanisms?, BioEssays News Rev. Mol. Cell. Dev. Biol, vol.36, pp.933-939, 2014.

T. M. Maynard, L. Sikich, J. A. Lieberman, and A. S. Lamantia, Neural development, cell-cell signaling, and the "two-hit" hypothesis of schizophrenia, Schizophr. Bull, vol.27, pp.457-476, 2001.

B. S. Mcewen, Stress, adaptation, and disease. Allostasis and allostatic load, Ann. N. Y. Acad. Sci, vol.840, pp.33-44, 1998.

K. A. Mclaughlin, L. D. Kubzansky, E. C. Dunn, R. Waldinger, G. Vaillant et al., Childhood social environment, emotional reactivity to stress, and mood and anxiety disorders across the life course, Depress. Anxiety, vol.27, pp.1087-1094, 2010.

A. S. Monte, B. S. Mello, V. C. Borella, T. Da-silva-araujo, F. E. Da-silva et al., Two-hit model of schizophrenia induced by neonatal immune activation and peripubertal stress in rats: Study of sex differences and brain oxidative alterations, Behav. Brain Res, vol.331, pp.30-37, 2017.

V. Mourlon, A. Baudin, O. Blanc, A. Lauber, B. Giros et al., Maternal deprivation induces depressive-like behaviours only in female rats, Behav. Brain Res, vol.213, pp.278-287, 2010.

A. Moya-pérez, A. Perez-villalba, A. Benítez-páez, I. Campillo, and Y. Sanz, Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice, Brain. Behav. Immun, 2017.

T. Murakami, K. Kamada, K. Mizushima, Y. Higashimura, K. Katada et al., Changes in Intestinal Motility and Gut Microbiota Composition in a Rat Stress Model, Digestion, vol.95, pp.55-60, 2017.

E. Nederhof and M. V. Schmidt, Mismatch or cumulative stress: Toward an integrated hypothesis of programming effects, Physiol. Behav, vol.106, pp.691-700, 2012.

S. B. Nelson and V. Valakh, Excitatory/Inhibitory Balance and Circuit Homeostasis in Autism Spectrum Disorders, Neuron, vol.87, pp.684-698, 2015.

S. M. O'mahony, N. P. Hyland, T. G. Dinan, and J. F. Cryan, Maternal separation as a model of brain-gut axis dysfunction, Psychopharmacology (Berl.), vol.214, pp.71-88, 2011.

A. Ornoy, L. Weinstein-fudim, and Z. Ergaz, Prenatal factors associated with autism spectrum disorder (ASD), Reprod. Toxicol. Elmsford N, vol.56, pp.155-169, 2015.

A. M. Patterson, I. E. Mulder, A. J. Travis, A. Lan, N. Cerf-bensussan et al., Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity, Front. Immunol, vol.8, p.1166, 2017.

N. K. Phillips, C. L. Hammen, P. A. Brennan, J. M. Najman, and W. Bor, Early Adversity and the Prospective Prediction of Depressive and Anxiety Disorders in Adolescents, J. Abnorm. Child Psychol, vol.33, pp.13-24, 2005.

, R: A language and environment for statistical computing. R Found, Stat. Comput. Vienna Austria ISBN, 2008.

A. V. Rao, A. C. Bested, T. M. Beaulne, M. A. Katzman, C. Iorio et al., A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome, Gut Pathog, vol.1, p.6, 2009.

J. Reunanen, V. Kainulainen, L. Huuskonen, N. Ottman, C. Belzer et al., Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer, Appl. Environ. Microbiol, vol.81, pp.3655-3662, 2015.

A. Riba, M. Olier, S. Lacroix-lamandé, C. Lencina, V. Bacquié et al., Paneth Cell Defects Induce Microbiota Dysbiosis In Mice And Promote Visceral Hypersensitivity. Gastroenterology, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01603303

E. M. Richard, J. 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.

M. Rincel, A. L. Lépinay, P. Delage, J. Fioramonti, V. S. Théodorou et al., Maternal high-fat diet prevents developmental programming by early-life stress, Transl. Psychiatry, vol.6, p.966, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602432

M. Salzberg, G. Kumar, L. Supit, N. C. Jones, M. J. Morris et al., Early postnatal stress confers enduring vulnerability to limbic epileptogenesis, Epilepsia, vol.48, pp.2079-2085, 2007.

A. Sarkar, P. Chachra, P. Kennedy, C. J. Pena, L. A. Desouza et al., , 2014.

, Hippocampal HDAC4 contributes to postnatal fluoxetine-evoked depression-like behavior, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.39, pp.2221-2232

R. Shepard, K. Heslin, and L. Coutellier, The transcription factor Npas4 contributes to adolescent development of prefrontal inhibitory circuits, and to cognitive and emotional functions: Implications for neuropsychiatric disorders, Neurobiol. Dis, vol.99, pp.36-46, 2017.

H. Shi, B. Sheng, F. Zhang, C. Wu, R. Zhang et al., Kruppel-like factor 2 protects against ischemic stroke by regulating endothelial blood brain barrier function, Am. J. Physiol. Heart Circ. Physiol, vol.304, pp.796-805, 2013.

S. Y. Shin, S. H. Han, R. Woo, S. H. Jang, M. et al., Adolescent mice show anxiety-and aggressive-like behavior and the reduction of long-term potentiation in mossy fiber-CA3 synapses after neonatal maternal separation, Neuroscience, vol.316, pp.221-231, 2016.

S. Yim, Y. Park, A. Berrios, J. Lafourcade, M. Pascual et al., Reversing behavioural abnormalities in mice exposed to maternal inflammation, Nature, vol.549, pp.482-487, 2017.

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. F. Slykerman, F. Hood, K. Wickens, J. M. Thompson, C. Barthow et al., Effect of Lactobacillus rhamnosus HN001 in Pregnancy on Postpartum Symptoms of Depression and Anxiety: A Randomised Double-blind, vol.24, pp.159-165, 2017.

G. K. Smyth, Linear Models and Empirical Bayes Methods for Assessing Differential Expression in Microarray Experiments, Stat Appl Genet Mol Biol, vol.3, 2004.

I. Spiegel, A. R. Mardinly, H. W. Gabel, J. E. Bazinet, C. H. Couch et al., Npas4 regulates excitatory-inhibitory balance within neural circuits through cell-type-specific gene programs, Cell, vol.157, pp.1216-1229, 2014.

Z. Steel, C. Marnane, C. Iranpour, T. Chey, J. W. Jackson et al., The global prevalence of common mental disorders: a systematic review and meta-analysis 1980-2013, Int. J. Epidemiol, vol.43, pp.476-493, 2014.

R. M. Stilling, M. Van-de-wouw, G. Clarke, C. Stanton, T. G. Dinan et al., The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis?, Neurochem. Int, vol.99, pp.110-132, 2016.

E. Suply, P. De-vries, R. Soret, F. Cossais, and M. Neunlist, Butyrate enemas enhance both cholinergic and nitrergic phenotype of myenteric neurons and neuromuscular transmission in newborn rat colon, Am. J. Physiol. Gastrointest. Liver Physiol, vol.302, pp.1373-1380, 2012.

A. T. Tang, J. P. Choi, J. J. Kotzin, Y. Yang, C. C. Hong et al., Endothelial TLR4 and the microbiome drive cerebral cavernous malformations, Nature, vol.545, pp.305-310, 2017.

M. Taniguchi, M. B. Carreira, Y. A. Cooper, A. Bobadilla, J. A. Heinsbroek et al., HDAC5 and Its Target Gene, Npas4, Function in the Nucleus Accumbens to Regulate Cocaine-Conditioned Behaviors, Neuron, vol.96, pp.130-144, 2017.

M. C. Tsuda, N. Yamaguchi, and S. Ogawa, Early life stress disrupts peripubertal development of aggression in male mice, Neuroreport, vol.22, pp.259-263, 2011.

P. Van-den-abbeele, C. Belzer, M. Goossens, M. Kleerebezem, W. M. De-vos et al., Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model, ISME J, vol.7, pp.949-961, 2013.

T. Vanhaecke, P. Aubert, P. Grohard, T. Durand, P. Hulin et al., L. fermentum CECT 5716 prevents stress-induced intestinal barrier dysfunction in newborn rats, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.29, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01712194

A. K. Varghese, E. F. Verdú, P. Bercik, W. I. Khan, P. A. Blennerhassett et al., Antidepressants attenuate increased susceptibility to colitis in a murine model of depression, Gastroenterology, vol.130, pp.1743-1753, 2006.

A. A. Walf and C. A. Frye, The use of the elevated plus maze as an assay of anxiety-related behavior in rodents, Nat. Protoc, vol.2, pp.322-328, 2007.

D. M. Werling and D. H. Geschwind, Sex differences in autism spectrum disorders, Curr. Opin. Neurol, vol.26, pp.146-153, 2013.

C. Wu, F. Li, G. Han, and Z. Liu, A?(1-42) disrupts the expression and function of KLF2 in Alzheimer's disease mediated by p53, Biochem. Biophys. Res. Commun, vol.431, pp.141-145, 2013.

O. Yizhar, L. E. Fenno, M. Prigge, F. Schneider, T. J. Davidson et al., Neocortical excitation/inhibition balance in information processing and social dysfunction, Nature, vol.477, pp.171-178, 2011.

A. Zager, M. L. Andersen, S. Tufik, and J. Palermo-neto, Maternal immune activation increases the corticosterone response to acute stress without affecting the hypothalamic monoamine content and sleep patterns in male mice offspring, Neuroimmunomodulation, vol.21, pp.37-44, 2014.

A. Zager, J. P. Peron, G. Mennecier, S. C. Rodrigues, T. P. Aloia et al., Maternal immune activation in late gestation increases neuroinflammation and aggravates experimental autoimmune encephalomyelitis in the offspring, Brain. Behav. Immun, vol.43, pp.159-171, 2015.

Z. Zakostelska, M. Kverka, K. Klimesova, P. Rossmann, J. Mrazek et al., Lysate of probiotic Lactobacillus casei DN-114 001 ameliorates colitis by strengthening the gut barrier function and changing the gut microenvironment, PloS One, vol.6, p.27961, 2011.

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, Mol. Psychiatry, vol.21, pp.786-796, 2016.

X. Zhou, M. Li, X. Li, X. Long, X. Zuo et al., Visceral hypersensitive rats share common dysbiosis features with irritable bowel syndrome patients, World J. Gastroenterol, vol.22, pp.5211-5227, 2016.

F. Zhu, Y. Zheng, Y. Liu, X. Zhang, and J. Zhao, Minocycline alleviates behavioral deficits and inhibits microglial activation in the offspring of pregnant mice after administration of polyriboinosinic-polyribocytidilic acid, Psychiatry Res, vol.219, pp.680-686, 2014.

O. Agid, B. Shapira, J. Zislin, M. Ritsner, B. Hanin et al., Environment and vulnerability to major psychiatric illness: a case control study of early parental loss in major depression, bipolar disorder and schizophrenia, Mol. Psychiatry, vol.4, pp.163-172, 1999.

J. P. Aguggia, M. M. Suárez, and M. A. Rivarola, Early maternal separation: Neurobehavioral consequences in mother rats, Behav. Brain Res, vol.248, pp.25-31, 2013.

B. Aisa, R. Tordera, B. Lasheras, J. Del-río, and M. J. Ramírez, Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats, Psychoneuroendocrinology, vol.32, pp.256-266, 2007.

B. Aisa, N. Elizalde, R. Tordera, B. Lasheras, J. Del-rã-o et al., Effects of neonatal stress on markers of synaptic plasticity in the hippocampus: Implications for spatial memory, Hippocampus, vol.19, pp.1222-1231, 2009.

A. Ait-belgnaoui, S. Bradesi, J. Fioramonti, V. Theodorou, and L. Bueno, Acute stress-induced hypersensitivity to colonic distension depends upon increase in paracellular permeability: role of myosin light chain kinase, Pain, vol.113, pp.141-147, 2005.

A. Ait-belgnaoui, H. Eutamene, E. Houdeau, L. Bueno, J. Fioramonti et al., Lactobacillus farciminis treatment attenuates stress-induced overexpression of Fos protein in spinal and supraspinal sites after colorectal distension in rats, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.21, pp.18-19, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01600974

A. Ait-belgnaoui, H. Durand, C. Cartier, G. Chaumaz, H. Eutamene et al., Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats, Psychoneuroendocrinology, vol.37, pp.1885-1895, 2012.

A. Ait-belgnaoui, A. Colom, V. Braniste, L. Ramalho, A. Marrot et al., Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.510-520, 2014.

G. Akkasheh, Z. Kashani-poor, M. Tajabadi-ebrahimi, P. Jafari, H. Akbari et al., Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial, Nutr. Burbank Los Angel. Cty. Calif, vol.32, pp.315-320, 2016.

T. Alenghat, L. C. Osborne, S. A. Saenz, D. Kobuley, C. G. Ziegler et al., Histone deacetylase 3 coordinates commensal-bacteriadependent intestinal homeostasis, Nature, vol.504, pp.153-157, 2013.

M. Altemus, Sex differences in depression and anxiety disorders: potential biological determinants, Horm. Behav, vol.50, pp.534-538, 2006.

H. Amini-khoei, A. Mohammadi-asl, S. Amiri, M. Hosseini, M. Momeny et al., Oxytocin mitigated the depressivelike behaviors of maternal separation stress through modulating mitochondrial function and neuroinflammation, Prog. Neuropsychopharmacol. Biol. Psychiatry, vol.76, pp.169-178, 2017.

S. Amiri, H. Amini-khoei, A. Mohammadi-asl, S. Alijanpour, A. Haj-mirzaian et al., Involvement of D1 and D2 dopamine receptors in the antidepressant-like effects of selegiline in maternal separation model of mouse, Physiol. Behav, vol.163, pp.107-114, 2016.

R. F. Anda, D. W. Brown, V. J. Felitti, J. D. Bremner, S. R. Dube et al., Adverse childhood experiences and prescribed psychotropic medications in adults, Am. J. Prev. Med, vol.32, pp.389-394, 2007.

R. H. Anderberg, J. E. Richard, C. Hansson, H. Nissbrandt, F. Bergquist et al., GLP-1 is both anxiogenic and antidepressant; divergent effects of acute and chronic GLP-1 on emotionality, Psychoneuroendocrinology, vol.65, pp.54-66, 2016.

S. L. Andersen and M. H. Teicher, Delayed Effects of Early Stress on Hippocampal Development, Neuropsychopharmacology, vol.29, pp.1988-1993, 2004.

S. L. Andersen and M. H. Teicher, Stress, sensitive periods and maturational events in adolescent depression, Trends Neurosci, vol.31, pp.183-191, 2008.

V. Andresen, D. R. Bach, A. Poellinger, C. Tsrouya, A. Stroh et al., Brain activation responses to subliminal or supraliminal rectal stimuli and to auditory stimuli in irritable bowel syndrome, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.17, pp.827-837, 2005.

H. Anisman, M. D. Zaharia, M. J. Meaney, and Z. Merali, Do early-life events permanently alter behavioral and hormonal responses to stressors?, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.16, pp.149-164, 1998.

L. Arborelius and M. B. Eklund, Both long and brief maternal separation produces persistent changes in tissue levels of brain monoamines in middle-aged female rats, Neuroscience, vol.145, pp.738-750, 2007.

T. Arentsen, H. Raith, Y. Qian, H. Forssberg, D. Heijtz et al., Host microbiota modulates development of social preference in mice, Microb. Ecol. Health Dis, vol.26, p.29719, 2015.

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

K. Atarashi, T. Tanoue, M. Ando, N. Kamada, Y. Nagano et al., Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells, Cell, vol.163, pp.367-380, 2015.

A. E. Autry and L. M. Monteggia, Brain-Derived Neurotrophic Factor and Neuropsychiatric Disorders, Pharmacol. Rev, vol.64, pp.238-258, 2012.

L. Aya-ramos, C. Contreras-vargas, J. L. Rico, and Z. Dueñas, Early maternal separation induces preference for sucrose and aspartame associated with increased blood glucose and hyperactivity, Food Funct, vol.8, pp.2592-2600, 2017.

M. V. Baciu, B. L. Bonaz, E. Papillon, R. A. Bost, J. F. Le-bas et al., Central processing of rectal pain: a functional MR imaging study, AJNR Am. J. Neuroradiol, vol.20, pp.1920-1924, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00798650

M. Bai, X. Zhu, Y. Zhang, S. Zhang, L. Zhang et al., Abnormal hippocampal BDNF and miR-16 expression is associated with depression-like behaviors induced by stress during early life, PloS One, vol.7, p.46921, 2012.

M. Bambling, S. C. Edwards, S. Hall, and L. Vitetta, A combination of probiotics and magnesium orotate attenuate depression in a small SSRI resistant cohort: an intestinal anti-inflammatory response is suggested, Inflammopharmacology, vol.25, pp.271-274, 2017.

A. Barbazanges, M. Vallée, W. Mayo, J. Day, H. Simon et al., Early and later adoptions have different long-term effects on male rat offspring, J. Neurosci. Off. J. Soc. Neurosci, vol.16, pp.7783-7790, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02323986

C. Barboza-solís, M. Kelly-irving, R. Fantin, M. Darnaudéry, J. Torrisani et al., Adverse childhood experiences and physiological wear-and-tear in midlife: Findings from the 1958 British birth cohort, Proc. Natl. Acad. Sci. U. S. A, vol.112, pp.738-746, 2015.

D. J. Barker, In utero programming of chronic disease, Clin. Sci. Lond. Engl, vol.95, pp.115-128, 1979.

I. Barna, E. Bálint, J. Baranyi, N. Bakos, G. B. Makara et al., Gender-specific effect of maternal deprivation on anxiety and corticotropin-releasing hormone mRNA expression in rats, Brain Res. Bull, vol.62, pp.85-91, 2003.

I. Barna, E. Bálint, J. Baranyi, N. Bakos, G. B. Makara et al., Gender-specific effect of maternal deprivation on anxiety and corticotropin-releasing hormone mRNA expression in rats, Brain Res. Bull, vol.62, pp.85-91, 2003.

J. Barouei, M. Moussavi, and D. M. Hodgson, Effect of maternal probiotic intervention on HPA axis, immunity and gut microbiota in a rat model of irritable bowel syndrome, PloS One, vol.7, p.46051, 2012.

J. Barouei, M. Moussavi, and D. M. Hodgson, Perinatal maternal probiotic intervention impacts immune responses and ileal mucin gene expression in a rat model of irritable bowel syndrome, Benef. Microbes, vol.6, pp.83-95, 2015.

F. Barreau, L. Ferrier, J. Fioramonti, and L. Bueno, Neonatal maternal deprivation triggers long term alterations in colonic epithelial barrier and mucosal immunity in rats, Gut, vol.53, pp.501-506, 2004.

F. Barreau, C. Cartier, L. Ferrier, J. Fioramonti, and L. Bueno, Nerve growth factor mediates alterations of colonic sensitivity and mucosal barrier induced by neonatal stress in rats, Gastroenterology, vol.127, pp.524-534, 2004.

F. Barreau, C. Cartier, M. Leveque, L. Ferrier, R. Moriez et al., Pathways involved in gut mucosal barrier dysfunction induced in adult rats by maternal deprivation: corticotrophin-releasing factor and nerve growth factor interplay, J. Physiol, vol.580, pp.347-356, 2007.

F. Barreau, L. Ferrier, J. Fioramonti, and L. Bueno, New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models, Pediatr. Res, vol.62, pp.240-245, 2007.

F. Barreau, C. Salvador-cartier, E. Houdeau, L. Bueno, and J. Fioramonti, Long-term alterations of colonic nerve-mast cell interactions induced by neonatal maternal deprivation in rats, Gut, vol.57, pp.582-590, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01604741

K. Baruch, N. Rosenzweig, A. Kertser, A. Deczkowska, A. M. Sharif et al., Breaking immune tolerance by targeting Foxp3+ regulatory T cells mitigates Alzheimer's disease pathology, Nat. Commun, vol.6, 2015.

A. Baudin, K. Blot, C. Verney, L. Estevez, J. Santamaria et al., Maternal deprivation induces deficits in temporal memory and cognitive flexibility and exaggerates synaptic plasticity in the rat medial prefrontal cortex, Neurobiol. Learn. Mem, vol.98, pp.207-214, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01542337

F. Benetti, P. B. Mello, J. S. Bonini, S. Monteiro, M. Cammarota et al., Early postnatal maternal deprivation in rats induces memory deficits in adult life that can be reversed by donepezil and galantamine, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.27, pp.59-64, 2009.

D. Benton, C. Williams, and A. Brown, Impact of consuming a milk drink containing a probiotic on mood and cognition, Eur. J. Clin. Nutr, vol.61, pp.355-361, 2007.

P. Bercik, E. F. Verdu, J. A. Foster, J. Macri, M. Potter et al., Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice, Gastroenterology, vol.139, pp.2102-2112, 2010.

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, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.23, pp.1132-1139, 2011.

P. Bercik, E. Denou, J. Collins, W. Jackson, J. Lu et al., The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice, Gastroenterology, vol.141, pp.599-609, 2011.

G. Biagini, E. M. Pich, C. Carani, P. Marrama, and L. F. Agnati, Postnatal maternal separation during the stress hyporesponsive period enhances the adrenocortical response to novelty in adult rats by affecting feedback regulation in the CA1 hippocampal field, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.16, pp.187-197, 1998.

J. Bick and C. A. Nelson, Early Adverse Experiences and the Developing Brain, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.41, pp.177-196, 2016.

S. C. Bischoff, G. Barbara, W. Buurman, T. Ockhuizen, J. Schulzke et al., Intestinal permeability--a new target for disease prevention and therapy, BMC Gastroenterol, vol.14, p.189, 2014.

M. L. Boccia, M. Razzoli, S. Prasad-vadlamudi, W. Trumbull, C. Caleffie et al., Repeated long separations from pups produce depression-like behavior in rat mothers, Psychoneuroendocrinology, vol.32, pp.65-71, 2007.

J. Bock, M. Gruss, S. Becker, and K. Braun, Experience-induced changes of dendritic spine densities in the prefrontal and sensory cortex: correlation with developmental time windows, Cereb. Cortex, vol.15, pp.802-808, 2005.

S. R. Bodnoff, B. Suranyi-cadotte, R. Quirion, and M. J. Meaney, Postnatal handling reduces noveltyinduced fear and increases [3H]flunitrazepam binding in rat brain, Eur. J. Pharmacol, vol.144, pp.105-107, 1987.

G. J. Boersma, T. L. Bale, P. Casanello, H. E. Lara, A. B. Lucion et al., Long-Term Impact of Early Life Events on Physiology and Behaviour, J. Neuroendocrinol, vol.26, pp.587-602, 2014.

J. Bohacek and I. M. Mansuy, Epigenetic inheritance of disease and disease risk, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.38, pp.220-236, 2013.

J. Bollrath and F. M. Powrie, Controlling the frontier: regulatory T-cells and intestinal homeostasis, Semin. Immunol, vol.25, pp.352-357, 2013.

B. L. Bonaz and C. N. Bernstein, Brain-Gut Interactions in Inflammatory Bowel Disease, Gastroenterology, vol.144, pp.36-49, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00745108

T. J. Borody and A. Khoruts, Fecal microbiota transplantation and emerging applications, Nat. Rev. Gastroenterol. Hepatol, vol.9, pp.88-96, 2011.

L. V. Borovikova, S. Ivanova, M. Zhang, H. Yang, G. I. Botchkina et al., Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin, Nature, vol.405, pp.458-462, 2000.

Y. E. Borre, G. W. O'keeffe, G. Clarke, C. Stanton, T. G. Dinan et al., Microbiota and neurodevelopmental windows: implications for brain disorders, Trends Mol. Med, vol.20, pp.509-518, 2014.

S. Botschuijver, G. Roeselers, E. Levin, D. M. Jonkers, O. Welting et al., Intestinal Fungal Dysbiosis Is Associated With Visceral Hypersensitivity in Patients With Irritable Bowel Syndrome and Rats, Gastroenterology, vol.153, pp.1026-1039, 2017.

M. W. Bourassa, I. Alim, S. J. Bultman, and R. R. Ratan, Butyrate, neuroepigenetics and the gut microbiome: Can a high fiber diet improve brain health?, Neurosci. Lett, vol.625, pp.56-63, 2016.

D. Bouskra, C. Brézillon, M. Bérard, C. Werts, R. Varona et al., Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis, Nature, vol.456, pp.507-510, 2008.
URL : https://hal.archives-ouvertes.fr/pasteur-01402759

M. E. Bowers, D. C. Choi, and K. J. Ressler, Neuropeptide regulation of fear and anxiety: Implications of cholecystokinin, endogenous opioids, and neuropeptide Y, Physiol. Behav, vol.107, pp.699-710, 2012.

K. Bradford, W. Shih, E. J. Videlock, A. P. Presson, B. D. Naliboff et al., Association Between Early Adverse Life Events and Irritable Bowel Syndrome, Clin. Gastroenterol. Hepatol, vol.10, pp.385-390, 2012.

W. G. Brake, T. Y. Zhang, J. Diorio, M. J. Meaney, and A. Gratton, Influence of early postnatal rearing conditions on mesocorticolimbic dopamine and behavioural responses to psychostimulants and stressors in adult rats, Eur. J. Neurosci, vol.19, pp.1863-1874, 2004.

I. Branchi, D. Santucci, and E. Alleva, Ultrasonic vocalisation emitted by infant rodents: a tool for assessment of neurobehavioural development, Behav. Brain Res, vol.125, pp.49-56, 2001.

L. J. Brandt and O. C. Aroniadis, An overview of fecal microbiota transplantation: techniques, indications, and outcomes, Gastrointest. Endosc, vol.78, pp.240-249, 2013.

V. Braniste, M. Al-asmakh, C. Kowal, F. Anuar, A. Abbaspour et al., The gut microbiota influences blood-brain barrier permeability in mice, Sci. Transl. Med, vol.6, pp.263-158, 2014.

J. A. Bravo, P. Forsythe, M. V. Chew, E. Escaravage, H. M. Savignac et al., Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve, Proc. Natl. Acad. Sci, vol.108, pp.16050-16055, 2011.

J. A. Bravo, T. G. Dinan, and J. F. Cryan, Early-life stress induces persistent alterations in 5-HT1A receptor and serotonin transporter mRNA expression in the adult rat brain, Front. Mol. Neurosci, vol.7, 2014.

A. S. Brown, The environment and susceptibility to schizophrenia, Prog. Neurobiol, vol.93, pp.23-58, 2011.

L. C. Bruce, R. G. Heimberg, P. R. Goldin, and J. J. Gross, Childhood Maltreatment and Response to Cognitive Behavioral Therapy Among Individuals with Social Anxiety Disorder, Depress. Anxiety, vol.30, pp.662-669, 2013.

A. J. Bruce-keller, J. M. Salbaum, M. Luo, E. Blanchard, C. M. Taylor et al., Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity, Biol. Psychiatry, vol.77, pp.607-615, 2015.

S. A. Brunelli, J. P. Curley, K. Gudsnuk, F. A. Champagne, M. M. Myers et al., Variations in maternal behavior in rats selected for infant ultrasonic vocalization in isolation, Horm. Behav, vol.75, pp.78-83, 2015.

S. A. Buffington, G. V. Di-prisco, T. A. Auchtung, N. J. Ajami, J. F. Petrosino et al., Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring, Cell, vol.165, pp.1762-1775, 2016.

T. Buie, D. B. Campbell, G. J. Fuchs, G. T. Furuta, J. Levy et al., Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report, Pediatrics, vol.125, issue.1, pp.1-18, 2010.

A. Burokas, R. D. Moloney, T. G. Dinan, and J. F. Cryan, Microbiota regulation of the Mammalian gutbrain axis, Adv. Appl. Microbiol, vol.91, pp.1-62, 2015.

A. Burokas, S. Arboleya, R. D. Moloney, V. L. Peterson, K. Murphy et al., Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice, Biol. Psychiatry, vol.82, pp.472-487, 2017.

C. Caldji, B. Tannenbaum, S. Sharma, D. Francis, P. M. Plotsky et al., Maternal care during infancy regulates the development of neural systems mediating the expression of fearfulness in the rat, Proc. Natl. Acad. Sci, vol.95, pp.5335-5340, 1998.

C. Caldji, D. Francis, S. Sharma, P. M. Plotsky, and M. J. Meaney, The effects of early rearing environment on the development of GABAA and central benzodiazepine receptor levels and novelty-induced fearfulness in the rat, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.22, pp.219-229, 2000.

R. Callard, A. J. George, and J. Stark, Cytokines, chaos, and complexity. Immunity, vol.11, pp.507-513, 1999.

N. M. Cameron, F. A. Champagne, C. Parent, E. W. Fish, K. Ozaki-kuroda et al., The programming of individual differences in defensive responses and reproductive strategies in the rat through variations in maternal care, Neurosci. Biobehav. Rev, vol.29, pp.843-865, 2005.

M. Camilleri, K. Madsen, R. Spiller, B. Greenwood-van-meerveld, B. G. Van-meerveld et al., Intestinal barrier function in health and gastrointestinal disease, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.24, pp.503-512, 2012.

E. P. Candido, R. Reeves, D. , and J. R. , Sodium butyrate inhibits histone deacetylation in cultured cells, Cell, vol.14, pp.105-113, 1978.

X. Cao, S. Huang, J. Cao, T. Chen, P. Zhu et al., The timing of maternal separation affects morris water maze performance and long-term potentiation in male rats: Timing of Maternal Separation on Rats, Dev. Psychobiol, vol.56, pp.1102-1109, 2014.

L. Capuron and A. H. Miller, Immune system to brain signaling: neuropsychopharmacological implications, Pharmacol. Ther, vol.130, pp.226-238, 2011.

T. Carpenter, S. M. Grecian, and R. M. Reynolds, Sex differences in early-life programming of the hypothalamic-pituitary-adrenal axis in humans suggest increased vulnerability in females: a systematic review, J. Dev. Orig. Health Dis, vol.8, pp.244-255, 2017.

A. Castillo-ruiz, M. Mosley, A. J. George, L. F. Mussaji, E. F. Fullerton et al., The microbiota influences cell death and microglial colonization in the perinatal mouse brain, Brain. Behav. Immun, 2017.

N. Cerf-bensussan and V. Gaboriau-routhiau, The immune system and the gut microbiota: friends or foes?, Nat. Rev. Immunol, vol.10, pp.735-744, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01204303

A. Chaloner, G. Meerveld, and B. , Sexually dimorphic effects of unpredictable early life adversity on visceral pain behavior in a rodent model, J. Pain Off. J. Am. Pain Soc, vol.14, pp.270-280, 2013.

D. L. Champagne, R. C. Bagot, F. Van-hasselt, G. Ramakers, M. J. Meaney et al., Maternal Care and Hippocampal Plasticity: Evidence for Experience-Dependent Structural Plasticity, Altered Synaptic Functioning, and Differential Responsiveness to Glucocorticoids and Stress, J. Neurosci, vol.28, pp.6037-6045, 2008.

F. A. Champagne, D. D. Francis, A. Mar, and M. J. Meaney, Variations in maternal care in the rat as a mediating influence for the effects of environment on development, Physiol. Behav, vol.79, pp.359-371, 2003.

P. Chapillon, V. Patin, V. Roy, A. Vincent, C. et al., Effects of pre-and postnatal stimulation on developmental, emotional, and cognitive aspects in rodents: a review, Dev. Psychobiol, vol.41, pp.373-387, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01576712

D. P. Chapman, C. L. Whitfield, V. J. Felitti, S. R. Dube, V. J. Edwards et al., Adverse childhood experiences and the risk of depressive disorders in adulthood, J. Affect. Disord, vol.82, pp.217-225, 2004.

D. S. Charney and A. Deutch, A functional neuroanatomy of anxiety and fear: implications for the pathophysiology and treatment of anxiety disorders, Crit. Rev. Neurobiol, vol.10, pp.419-446, 1996.

J. Chen, J. H. Winston, and S. K. Sarna, Neurological and cellular regulation of visceral hypersensitivity induced by chronic stress and colonic inflammation in rats, Neuroscience, vol.248, pp.469-478, 2013.

D. K. Chitkara, M. A. Van-tilburg, N. Blois-martin, and W. E. Whitehead, Early life risk factors that contribute to irritable bowel syndrome in adults: a systematic review, Am. J. Gastroenterol, vol.103, pp.765-774, 2008.

A. Chocyk, B. Bobula, D. Dudys, A. Przyborowska, I. Majcher-ma?lanka et al., Early-life stress affects the structural and functional plasticity of the medial prefrontal cortex in adolescent rats, Eur. J. Neurosci, vol.38, pp.2089-2107, 2013.

A. Chocyk, I. Majcher-ma?lanka, A. Przyborowska, M. Ma?kowiak, and K. W?dzony, Early-life stress increases the survival of midbrain neurons during postnatal development and enhances reward-related and anxiolytic-like behaviors in a sex-dependent fashion, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.44, pp.33-47, 2015.

G. P. Chrousos and T. Kino, Glucocorticoid signaling in the cell. Expanding clinical implications to complex human behavioral and somatic disorders, Ann. N. Y. Acad. Sci, vol.1179, pp.153-166, 2009.

F. Cirulli, E. Alleva, A. Antonelli, A. , and L. , NGF expression in the developing rat brain: effects of maternal separation, Brain Res. Dev. Brain Res, vol.123, pp.129-134, 2000.

F. Cirulli, A. Berry, and E. Alleva, Early disruption of the mother-infant relationship: effects on brain plasticity and implications for psychopathology, Neurosci. Biobehav. Rev, vol.27, pp.73-82, 2003.

F. Cirulli, N. Francia, A. Berry, L. Aloe, E. Alleva et al., Early life stress as a risk factor for mental health: role of neurotrophins from rodents to non-human primates, Neurosci. Biobehav. Rev, vol.33, pp.573-585, 2009.

L. L. Clarke, A guide to Ussing chamber studies of mouse intestine, Am. J. Physiol. Gastrointest. Liver Physiol, vol.296, pp.1151-1166, 2009.

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, pp.666-673, 2013.

D. R. Clayburgh, T. A. Barrett, Y. Tang, J. B. Meddings, L. J. Van-eldik et al., Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo, J. Clin. Invest, vol.115, pp.2702-2715, 2005.

M. C. Collado, S. Rautava, J. Aakko, E. Isolauri, and S. Salminen, Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid, Sci. Rep, vol.6, p.23129, 2016.

S. M. Collins, A role for the gut microbiota in IBS, Nat. Rev. Gastroenterol. Hepatol, vol.11, pp.497-505, 2014.

J. Collins, R. Borojevic, E. F. Verdu, J. D. Huizinga, and E. M. Ratcliffe, Intestinal microbiota influence the early postnatal development of the enteric nervous system, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.98-107, 2014.

S. M. Collins, Z. Kassam, and P. Bercik, The adoptive transfer of behavioral phenotype via the intestinal microbiota: experimental evidence and clinical implications, Curr. Opin. Microbiol, vol.16, pp.240-245, 2013.

E. M. Cotella, I. Mestres-lascano, L. Franchioni, G. M. Levin, and M. M. Suárez, Long-term effects of maternal separation on chronic stress response suppressed by amitriptyline treatment, Stress Amst. Neth, vol.16, pp.477-481, 2013.

E. C. Cottrell and J. R. Seckl, Prenatal Stress, Glucocorticoids and the Programming of Adult Disease, Front. Behav. Neurosci, vol.3, 2009.

S. V. Coutinho, P. M. Plotsky, M. Sablad, J. C. Miller, H. Zhou et al., Neonatal maternal separation alters stress-induced responses to viscerosomatic nociceptive stimuli in rat, Am. J. Physiol. Gastrointest. Liver Physiol, vol.282, pp.307-316, 2002.

F. S. Couto, . Do, V. L. Batalha, J. S. Valadas, J. Data-franca et al., Escitalopram improves memory deficits induced by maternal separation in the rat, Eur. J. Pharmacol, vol.695, pp.71-75, 2012.

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. 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.

J. F. Cryan and A. Holmes, The ascent of mouse: advances in modelling human depression and anxiety, Nat. Rev. Drug Discov, vol.4, pp.775-790, 2005.

A. G. Cummins and F. M. Thompson, Effect of breast milk and weaning on epithelial growth of the small intestine in humans, Gut, vol.51, pp.748-754, 2002.

D. Silva, S. Robbe-masselot, C. Ait-belgnaoui, A. Mancuso, A. Mercade-loubière et al., Stress disrupts intestinal mucus barrier in rats via mucin O-glycosylation shift: prevention by a probiotic treatment, Am. J. Physiol. Gastrointest. Liver Physiol, vol.307, pp.420-429, 2014.

M. F. Dallman, S. F. Akana, N. Levin, C. D. Walker, M. J. Bradbury et al., Corticosteroids and the control of function in the hypothalamo-pituitary-adrenal (HPA) axis, Ann. N. Y. Acad. Sci, vol.746, pp.64-67, 1994.

F. R. D'amato, E. Scalera, C. Sarli, and A. Moles, Pups call, mothers rush: does maternal responsiveness affect the amount of ultrasonic vocalizations in mouse pups?, Behav. Genet, vol.35, pp.103-112, 2005.

A. Danese and B. S. Mcewen, Adverse childhood experiences, allostasis, allostatic load, and age-related disease, Physiol. Behav, vol.106, pp.29-39, 2012.

J. Danielewicz and G. Hess, Early life stress alters synaptic modification range in the rat lateral amygdala, Behav. Brain Res, vol.265, pp.32-37, 2014.

W. M. Daniels, C. Y. Pietersen, M. E. Carstens, and D. J. Stein, Maternal Separation in Rats Leads to Anxiety-Like Behavior and a Blunted ACTH Response and Altered Neurotransmitter Levels in Response to a Subsequent Stressor, Metab. Brain Dis, vol.19, pp.3-14, 2004.

R. Dantzer, Cytokine-induced sickness behavior: mechanisms and implications, Ann. N. Y. Acad. Sci, vol.933, pp.222-234, 2001.

R. Dantzer, J. P. Konsman, R. M. Bluthé, and K. W. Kelley, Neural and humoral pathways of communication from the immune system to the brain: parallel or convergent?, Auton. Neurosci. Basic Clin, vol.85, pp.60-65, 2000.

M. Darnaudéry, M. Koehl, A. Barbazanges, S. Cabib, M. Le-moal et al., Early and later adoptions differently modify mother-pup interactions, Behav. Neurosci, vol.118, pp.590-596, 2004.

N. A. Datson, M. C. Morsink, O. C. Meijer, and E. R. De-kloet, Central corticosteroid actions: Search for gene targets, Eur. J. Pharmacol, vol.583, pp.272-289, 2008.

E. A. Daubert and B. G. Condron, Serotonin: a regulator of neuronal morphology and circuitry, Trends Neurosci, vol.33, pp.424-434, 2010.

C. P. Davis and D. C. Savage, Habitat, succession, attachment, and morphology of segmented, filamentous microbes indigenous to the murine gastrointestinal tract, Infect. Immun, vol.10, pp.948-956, 1974.

D. Bellis, M. D. Keshavan, M. S. Shifflett, H. Iyengar, S. Beers et al., Brain structures in pediatric maltreatment-related posttraumatic stress disorder: a sociodemographically matched study, Biol. Psychiatry, vol.52, pp.1066-1078, 2002.

M. N. De-lima, J. Presti-torres, G. Vedana, L. A. Alcalde, L. Stertz et al., Early life stress decreases hippocampal BDNF content and exacerbates recognition memory deficits induced by repeated D-amphetamine exposure, Behav. Brain Res, vol.224, pp.100-106, 2011.

G. De-palma, P. Blennerhassett, J. Lu, Y. Deng, A. J. Park et al., Microbiota and host determinants of behavioural phenotype in maternally separated mice, Nat. Commun, vol.6, p.7735, 2015.

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, 2017.

D. Vadder, F. Kovatcheva-datchary, P. Goncalves, D. Vinera, J. Zitoun et al., Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits, Cell, vol.156, pp.84-96, 2014.

R. M. Deacon, Digging and marble burying in mice: simple methods for in vivo identification of biological impacts, Nat. Protoc, vol.1, pp.122-124, 2006.

V. H. Denenberg, D. R. Ottinger, and M. W. Stephens, Effects of maternal factors upon growth and behavior of the rat, Child Dev, vol.33, pp.65-71, 1962.

W. Deng, J. B. Aimone, and F. H. Gage, New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory?, Nat. Rev. Neurosci, vol.11, pp.339-350, 2010.

A. M. Depino, Early prenatal exposure to LPS results in anxiety-and depression-related behaviors in adulthood, Neuroscience, vol.299, pp.56-65, 2015.

B. Deplancke and H. R. Gaskins, Microbial modulation of innate defense: goblet cells and the intestinal mucus layer, Am. J. Clin. Nutr, vol.73, pp.1131-1141, 2001.

L. Desbonnet, L. Garrett, G. Clarke, J. Bienenstock, and T. G. Dinan, The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat, J. Psychiatr. Res, vol.43, pp.164-174, 2008.

L. Desbonnet, L. Garrett, G. Clarke, B. Kiely, J. F. Cryan et al., Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression, Neuroscience, vol.170, pp.1179-1188, 2010.

L. Desbonnet, G. Clarke, F. Shanahan, T. G. Dinan, and J. F. Cryan, Microbiota is essential for social development in the mouse, Mol. Psychiatry, vol.19, pp.146-148, 2014.

L. Desbonnet, G. Clarke, A. Traplin, O. O'sullivan, F. Crispie et al., Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour, Brain. Behav. Immun, vol.48, pp.165-173, 2015.

L. A. Diehl, N. Pereira, S. C. De, D. P. Laureano, A. N. Benitz et al., Contextual Fear Conditioning in Maternal Separated Rats: The Amygdala as a Site for Alterations, Neurochem. Res, vol.39, pp.384-393, 2014.

D. G. Dillon, A. J. Holmes, J. L. Birk, N. Brooks, K. Lyons-ruth et al., Childhood adversity is associated with left basal ganglia dysfunction during reward anticipation in adulthood, Biol. Psychiatry, vol.66, pp.206-213, 2009.

J. J. Dimatelis, I. M. Vermeulen, K. Bugarith, D. J. Stein, and V. A. Russell, Female rats are resistant to developing the depressive phenotype induced by maternal separation stress, Metab. Brain Dis, vol.31, pp.109-119, 2016.

T. G. Dinan, C. Stanton, and J. F. Cryan, Psychobiotics: a novel class of psychotropic, Biol. Psychiatry, vol.74, pp.720-726, 2013.

S. Diwakarla, L. J. Fothergill, J. Fakhry, B. Callaghan, and J. B. Furness, Heterogeneity of enterochromaffin cells within the gastrointestinal tract, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.29, 2017.

D. Prado, C. H. Narahari, T. Holland, F. H. Lee, H. Murthy et al., Effects of early adolescent environmental enrichment on cognitive dysfunction, prefrontal cortex development, and inflammatory cytokines after early life stress, Dev. Psychobiol, 2015.

M. G. Dominguez-bello, E. K. Costello, M. Contreras, M. Magris, G. Hidalgo et al., Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.11971-11975, 2010.

C. Dominianni, R. Sinha, J. J. Goedert, Z. Pei, L. Yang et al., Sex, body mass index, and dietary fiber intake influence the human gut microbiome, PloS One, vol.10, p.124599, 2015.

G. P. Donaldson, S. M. Lee, and S. K. Mazmanian, Gut biogeography of the bacterial microbiota, Nat. Rev. Microbiol, vol.14, pp.20-32, 2016.

M. J. Dörfel and O. Huber, Modulation of tight junction structure and function by kinases and phosphatases targeting occludin, J. Biomed. Biotechnol, p.807356, 2012.

S. H. Duncan, P. Louis, J. M. Thomson, and H. J. Flint, The role of pH in determining the species composition of the human colonic microbiota, Environ. Microbiol, vol.11, pp.2112-2122, 2009.

G. Eberl, A new vision of immunity: homeostasis of the superorganism, Mucosal Immunol, vol.3, pp.450-460, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-00509637

G. Eberl, Immunity by equilibrium, Nat. Rev. Immunol, vol.16, pp.524-532, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01402375

G. Eberl and M. Lochner, The development of intestinal lymphoid tissues at the interface of self and microbiota, Mucosal Immunol, vol.2, pp.478-485, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00509629

P. B. Eckburg, E. M. Bik, C. N. Bernstein, E. Purdom, L. Dethlefsen et al., Diversity of the human intestinal microbial flora, Science, vol.308, pp.1635-1638, 2005.

K. L. Edelblum, G. Sharon, G. Singh, M. A. Odenwald, A. Sailer et al., The Microbiome Activates CD4 T-cell-mediated Immunity to Compensate for Increased Intestinal Permeability, Cell. Mol. Gastroenterol. Hepatol, vol.4, pp.285-297, 2017.

L. Eiland and B. S. Mcewen, Early life stress followed by subsequent adult chronic stress potentiates anxiety and blunts hippocampal structural remodeling, Hippocampus, vol.22, pp.82-91, 2012.

A. J. Eisch and D. Petrik, Depression and hippocampal neurogenesis: a road to remission?, Science, vol.338, pp.72-75, 2012.

M. B. Eklund, A. , and L. , Twice daily long maternal separations in Wistar rats decreases anxietylike behaviour in females but does not affect males, Behav. Brain Res, vol.172, pp.278-285, 2006.

S. El-aidy, . Van-den, B. Bogert, and M. Kleerebezem, The small intestine microbiota, nutritional modulation and relevance for health, Curr. Opin. Biotechnol, vol.32, pp.14-20, 2015.

S. El-aidy, A. S. Ramsteijn, F. Dini-andreote, R. Van-eijk, D. J. Houwing et al., Serotonin Transporter Genotype Modulates the Gut Microbiota Composition in Young Rats, an Effect Augmented by Early Life Stress, Front. Cell. Neurosci, vol.11, p.222, 2017.

S. Elsenbruch, C. Rosenberger, U. Bingel, M. Forsting, M. Schedlowski et al., Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli, Gastroenterology, vol.139, pp.1310-1319, 2010.

J. R. Emge, K. Huynh, E. N. Miller, M. Kaur, C. Reardon et al., Modulation of the microbiota-gut-brain axis by probiotics in a murine model of inflammatory bowel disease, Am. J. Physiol. Gastrointest. Liver Physiol, vol.310, pp.989-998, 2016.

M. Enayati, J. Solati, M. Hosseini, H. Shahi, G. Saki et al., Maternal infection during late pregnancy increases anxiety-and depression-like behaviors with increasing age in male offspring, Brain Res. Bull, vol.87, pp.295-302, 2012.

B. Engelhardt and R. M. Ransohoff, Capture, crawl, cross: the T cell code to breach the blood-brain barriers, Trends Immunol, vol.33, pp.579-589, 2012.

S. Entringer, C. Buss, and P. D. Wadhwa, Prenatal stress, development, health and disease risk: A psychobiological perspective-2015 Curt Richter Award Paper, Psychoneuroendocrinology, vol.62, pp.366-375, 2015.

D. Erny, A. L. Hrab?-de-angelis, D. Jaitin, P. Wieghofer, O. Staszewski et al., Host microbiota constantly control maturation and function of microglia in the CNS, Nat. Neurosci, vol.18, pp.965-977, 2015.

M. Estienne, J. Claustre, G. Clain-gardechaux, A. Paquet, Y. Tache et al., Maternal deprivation alters epithelial secretory cell lineages in rat duodenum: role of CRF-related peptides, Gut, vol.59, pp.744-751, 2010.

R. Famularo, R. Kinscherff, and T. Fenton, Psychiatric diagnoses of maltreated children: preliminary findings, J. Am. Acad. Child Adolesc. Psychiatry, vol.31, pp.863-867, 1992.

C. Faravelli, Childhood stressful events, HPA axis and anxiety disorders, World J. Psychiatry, vol.2, p.13, 2012.

J. Farkas, D. Reglodi, B. Gaszner, D. Szogyi, G. Horvath et al., Effects of maternal separation on the neurobehavioral development of newborn Wistar rats, Brain Res. Bull, vol.79, pp.208-214, 2009.

M. R. Farrell, F. H. Holland, R. M. Shansky, and H. C. Brenhouse, Sex-specific effects of early life stress on social interaction and prefrontal cortex dendritic morphology in young rats, Behav. Brain Res, vol.310, pp.119-125, 2016.

A. J. Feifel, H. N. Shair, and C. Schmauss, Lasting effects of early life stress in mice: interaction of maternal environment and infant genes, Genes Brain Behav, 2017.

V. J. Felitti, R. F. Anda, D. Nordenberg, D. F. Williamson, A. M. Spitz et al., Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) Study, Am. J. Prev. Med, vol.14, pp.245-258, 1998.

C. F. Ferreira, J. R. Bernardi, R. Krolow, D. M. Arcego, G. R. Fries et al., Vulnerability to dietary n-3 polyunsaturated fatty acid deficiency after exposure to early stress in rats, Pharmacol. Biochem. Behav, vol.107, pp.11-19, 2013.

E. Flinkkilä, A. Keski-rahkonen, M. Marttunen, and A. Raevuori, Prenatal Inflammation, Infections and Mental Disorders, Psychopathology, vol.49, pp.317-333, 2016.

H. J. Flint, E. A. Bayer, M. T. Rincon, R. Lamed, and B. A. White, Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis, Nat. Rev. Microbiol, vol.6, pp.121-131, 2008.

K. A. Foley, D. F. Macfabe, M. Kavaliers, and K. Ossenkopp, Sexually dimorphic effects of prenatal exposure to lipopolysaccharide, and prenatal and postnatal exposure to propionic acid, on acoustic startle response and prepulse inhibition in adolescent rats: relevance to autism spectrum disorders, Behav. Brain Res, vol.278, pp.244-256, 2015.

D. G. Folks, The interface of psychiatry and irritable bowel syndrome, Curr. Psychiatry Rep, vol.6, pp.210-215, 2004.

E. Fombonne, The prevalence of autism, JAMA, vol.289, pp.87-89, 2003.

G. Fond, A. Loundou, N. Hamdani, W. Boukouaci, A. Dargel et al., Anxiety and depression comorbidities in irritable bowel syndrome (IBS): a systematic review and meta-analysis, Eur. Arch. Psychiatry Clin. Neurosci, vol.264, pp.651-660, 2014.

F. Fouhy, A. G. Clooney, C. Stanton, M. J. Claesson, and P. D. Cotter, 16S rRNA gene sequencing of mock microbial populations-impact of DNA extraction method, primer choice and sequencing platform, BMC Microbiol, vol.16, p.123, 2016.

D. D. Francis and M. J. Meaney, Maternal care and the development of stress responses, Curr. Opin. Neurobiol, vol.9, pp.128-134, 1999.

D. Francis, J. Diorio, D. Liu, and M. J. Meaney, Nongenomic transmission across generations of maternal behavior and stress responses in the rat, Science, vol.286, pp.1155-1158, 1999.

D. D. Francis, J. Diorio, P. M. Plotsky, and M. J. Meaney, Environmental enrichment reverses the effects of maternal separation on stress reactivity, J. Neurosci, vol.22, pp.7840-7843, 2002.

F. Fransen, A. A. Van-beek, T. Borghuis, B. Meijer, F. Hugenholtz et al., The Impact of, Gut Microbiota on Gender-Specific Differences in Immunity. Front. Immunol, vol.8, p.754, 2017.

T. Frodl, E. Reinhold, N. Koutsouleris, G. Donohoe, B. Bondy et al., Childhood stress, serotonin transporter gene and brain structures in major depression, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.35, pp.1383-1390, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00509101

T. Frodl, E. Reinhold, N. Koutsouleris, M. Reiser, and E. M. Meisenzahl, Interaction of childhood stress with hippocampus and prefrontal cortex volume reduction in major depression, J. Psychiatr. Res, vol.44, pp.799-807, 2010.

E. E. Fröhlich, A. Farzi, R. Mayerhofer, F. Reichmann, A. Ja?an et al., Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication, Brain. Behav. Immun, 2016.

K. E. Fujimura, N. A. Slusher, M. D. Cabana, and S. V. Lynch, Role of the gut microbiota in defining human health, Expert Rev. Anti Infect. Ther, vol.8, pp.435-454, 2010.

H. Furmaga, A. Shah, and A. Frazer, Serotonergic and noradrenergic pathways are required for the anxiolytic-like and antidepressant-like behavioral effects of repeated vagal nerve stimulation in rats, Biol. Psychiatry, vol.70, pp.937-945, 2011.

J. B. Furness, The enteric nervous system and neurogastroenterology, Nat. Rev. Gastroenterol. Hepatol, vol.9, pp.286-294, 2012.

V. Gaboriau-routhiau, S. Rakotobe, E. Lécuyer, I. Mulder, A. Lan et al., The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses, Immunity, vol.31, pp.677-689, 2009.

M. Gacias, S. Gaspari, P. G. Santos, S. Tamburini, M. Andrade et al., Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior, 2016.

I. Gárate, B. García-bueno, J. L. Madrigal, L. Bravo, E. Berrocoso et al., Origin and consequences of brain Toll-like receptor 4 pathway stimulation in an experimental model of depression, J. Neuroinflammation, vol.8, p.151, 2011.

C. L. García-ródenas, G. E. Bergonzelli, S. Nutten, A. Schumann, C. Cherbut et al., Nutritional approach to restore impaired intestinal barrier function and growth after neonatal stress in rats, J. Pediatr. Gastroenterol. Nutr, vol.43, pp.16-24, 2006.

M. G. Gareau, J. Jury, P. C. Yang, G. Macqueen, and M. H. Perdue, Neonatal maternal separation causes colonic dysfunction in rat pups including impaired host resistance, Pediatr. Res, vol.59, pp.83-88, 2006.

M. G. Gareau, J. Jury, G. Macqueen, P. M. Sherman, and M. H. Perdue, Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation, Gut, vol.56, pp.1522-1528, 2007.

M. G. Gareau, J. Jury, and M. H. Perdue, Neonatal maternal separation of rat pups results in abnormal cholinergic regulation of epithelial permeability, Am. J. Physiol. Gastrointest. Liver Physiol, vol.293, pp.198-203, 2007.

M. G. Gareau, E. Wine, D. M. Rodrigues, J. H. Cho, M. T. Whary et al., Bacterial infection causes stress-induced memory dysfunction in mice, Gut, vol.60, pp.307-317, 2011.

, Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study, GBD 2015 Disease and Injury Incidence and Prevalence Collaborators, vol.388, pp.1545-1602, 2015.

M. D. Gershon, The enteric nervous system: a second brain, Hosp. Pract, vol.34, pp.41-42, 1995.

J. Ghia, P. Blennerhassett, and S. M. Collins, Impaired parasympathetic function increases susceptibility to inflammatory bowel disease in a mouse model of depression, J. Clin. Invest, vol.118, pp.2209-2218, 2008.

G. R. Gibson and M. B. Roberfroid, Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics, J. Nutr, vol.125, pp.1401-1412, 1995.

S. Giovanoli, H. Engler, A. Engler, J. Richetto, J. Feldon et al., Preventive effects of minocycline in a neurodevelopmental two-hit model with relevance to schizophrenia, Transl. Psychiatry, vol.6, p.772, 2016.

V. Glover, Annual Research Review: Prenatal stress and the origins of psychopathology: an evolutionary perspective, J. Child Psychol. Psychiatry, vol.52, pp.356-367, 2011.

P. D. Gluckman and M. A. Hanson, Living with the Past: Evolution, Development, and Patterns of Disease, Science, vol.305, pp.1733-1736, 2004.

A. R. Gobinath, J. L. Workman, C. Chow, S. E. Lieblich, and L. A. Galea, Maternal postpartum corticosterone and fluoxetine differentially affect adult male and female offspring on anxiety-like behavior, stress reactivity, and hippocampal neurogenesis, Neuropharmacology, vol.101, pp.165-178, 2016.

A. D. Goldberg, C. D. Allis, and E. Bernstein, Epigenetics: a landscape takes shape, Cell, vol.128, pp.635-638, 2007.

A. V. Golubeva, S. Crampton, L. Desbonnet, D. Edge, O. O'sullivan et al., Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood, Psychoneuroendocrinology, vol.60, pp.58-74, 2015.

M. Gomez-de-agüero, S. C. Ganal-vonarburg, T. Fuhrer, S. Rupp, Y. Uchimura et al., The maternal microbiota drives early postnatal innate immune development, Science, vol.351, pp.1296-1302, 2016.

M. C. Gondré-lewis, P. J. Darius, H. Wang, and J. S. Allard, Stereological analyses of reward system nuclei in maternally deprived/separated alcohol drinking rats, J. Chem. Neuroanat, vol.76, pp.122-132, 2016.

T. Gos, J. Bock, G. Poeggel, and K. Braun, Stress-induced synaptic changes in the rat anterior cingulate cortex are dependent on endocrine developmental time windows, vol.62, pp.229-232, 2008.

R. Gosselin, R. M. O'connor, M. Tramullas, M. Julio-pieper, T. G. Dinan et al., Riluzole normalizes early-life stress-induced visceral hypersensitivity in rats: role of spinal glutamate reuptake mechanisms, Gastroenterology, vol.138, pp.2418-2425, 2010.

I. Gracia-rubio, M. Moscoso-castro, O. J. Pozo, J. Marcos, R. Nadal et al., Maternal separation induces neuroinflammation and long-lasting emotional alterations in mice, Prog. Neuropsychopharmacol. Biol. Psychiatry, vol.65, pp.104-117, 2016.

R. Grassi-oliveira, J. A. Honeycutt, F. H. Holland, P. Ganguly, and H. C. Brenhouse, Cognitive impairment effects of early life stress in adolescents can be predicted with early biomarkers: Impacts of sex, experience, and cytokines, Psychoneuroendocrinology, vol.71, pp.19-30, 2016.

S. Grenham, G. Clarke, J. F. Cryan, and T. G. Dinan, Brain-Gut-Microbe Communication in Health and Disease, Front. Physiol, vol.2, 2011.

N. Gröger, E. Matas, T. Gos, A. Lesse, G. Poeggel et al., The transgenerational transmission of childhood adversity: behavioral, cellular, and epigenetic correlates, J. Neural Transm, vol.123, pp.1037-1052, 1996.

D. F. Gros, M. M. Antony, R. E. Mccabe, and R. P. Swinson, Frequency and severity of the symptoms of irritable bowel syndrome across the anxiety disorders and depression, J. Anxiety Disord, vol.23, pp.290-296, 2009.

R. L. Grover, G. S. Ginsburg, and N. Ialongo, Childhood Predictors of Anxiety Symptoms: A Longitudinal Study, Child Psychiatry Hum. Dev, vol.36, pp.133-153, 2005.

M. Gruss, K. Braun, J. U. Frey, and V. Korz, Maternal separation during a specific postnatal time window prevents reinforcement of hippocampal long-term potentiation in adolescent rats, Neuroscience, vol.152, pp.1-7, 2008.

F. Guida, F. Turco, M. Iannotta, D. De-gregorio, I. Palumbo et al., Antibiotic-induced microbiota perturbation causes gut endocannabinoidome changes, hippocampal neuroglial reorganization and depression in mice, Brain. Behav. Immun, 2017.

A. Guleria, A. Karyampudi, R. Singh, C. L. Khetrapal, A. Verma et al., Mapping of Brain Activations to Rectal Balloon Distension Stimuli in Male Patients with Irritable Bowel Syndrome Using Functional Magnetic Resonance Imaging, J. Neurogastroenterol. Motil, vol.23, pp.415-427, 2017.

B. B. Gundersen and J. A. Blendy, Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety, Neuropharmacology, vol.57, pp.67-74, 2009.

B. E. Gustafsson, Lightweight stainless steel systems for rearing germfree animals, Ann. N. Y. Acad. Sci, vol.78, pp.17-28, 1959.

B. E. Gustafsson, Vitamin K deficiency in germfree rats, Ann. N. Y. Acad. Sci, vol.78, pp.166-174, 1959.

D. A. Gutman and C. B. Nemeroff, Neurobiology of early life stress: rodent studies, Semin. Clin. Neuropsychiatry, vol.7, pp.89-95, 2002.

D. A. Hackman, M. J. Farah, and M. J. Meaney, Socioeconomic status and the brain: mechanistic insights from human and animal research, Nat. Rev. Neurosci, vol.11, pp.651-659, 2010.

M. Halland, A. Almazar, R. Lee, E. Atkinson, J. Larson et al., A case-control study of childhood trauma in the development of irritable bowel syndrome, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.990-998, 2014.

C. Hammen, J. Davila, G. Brown, A. Ellicott, and M. Gitlin, Psychiatric history and stress: predictors of severity of unipolar depression, J. Abnorm. Psychol, vol.101, pp.45-52, 1992.

C. Hammen, R. Henry, and S. E. Daley, Depression and sensitization to stressors among young women as a function of childhood adversity, J. Consult. Clin. Psychol, vol.68, pp.782-787, 2000.

R. Hanamsagar and S. D. Bilbo, Sex differences in neurodevelopmental and neurodegenerative disorders: Focus on microglial function and neuroinflammation during development, J. Steroid Biochem. Mol. Biol, vol.160, pp.127-133, 2016.

S. L. Hays, R. J. Mcpherson, S. E. Juul, G. Wallace, A. G. Schindler et al., Long-term effects of neonatal stress on adult conditioned place preference (CPP) and hippocampal neurogenesis, Behav. Brain Res, vol.227, pp.7-11, 2012.

R. D. Heijtz, S. Wang, F. Anuar, Y. Qian, B. Bjorkholm et al., Normal gut microbiota modulates brain development and behavior, Proc. Natl. Acad. Sci, vol.108, pp.3047-3052, 2011.

C. Heim and E. B. Binder, Current research trends in early life stress and depression: Review of human studies on sensitive periods, gene-environment interactions, and epigenetics, Exp. Neurol, vol.233, pp.102-111, 2012.

C. Heim and C. B. Nemeroff, The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies, Biol. Psychiatry, vol.49, pp.1023-1039, 2001.

J. J. Heindel, J. Balbus, L. Birnbaum, M. N. Brune-drisse, P. Grandjean et al., Developmental Origins of Health and Disease: Integrating Environmental Influences, Endocrinology, vol.156, pp.3416-3421, 2015.

M. R. Herbert, Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders, Curr. Opin. Neurol, vol.23, pp.103-110, 2010.

J. P. Herman, Regulation of Hypothalamo-Pituitary-Adrenocortical Responses to Stressors by the Nucleus of the Solitary Tract/Dorsal Vagal Complex, Cell. Mol. Neurobiol, 2017.

J. P. Herman, H. Figueiredo, N. K. Mueller, Y. Ulrich-lai, M. M. Ostrander et al., Central mechanisms of stress integration: hierarchical circuitry controlling hypothalamo-pituitaryadrenocortical responsiveness, Front. Neuroendocrinol, vol.24, pp.151-180, 2003.

J. P. Herman, J. M. Mcklveen, S. Ghosal, B. Kopp, A. Wulsin et al., Regulation of the Hypothalamic-Pituitary-Adrenocortical Stress Response, Compr. Physiol, vol.6, pp.603-621, 2016.

M. Heyman, J. Abed, C. Lebreton, and N. Cerf-bensussan, Intestinal permeability in coeliac disease: insight into mechanisms and relevance to pathogenesis, Gut, vol.61, pp.1355-1364, 2012.

R. A. Hill, M. Klug, S. Kiss-von-soly, M. D. Binder, A. J. Hannan et al., Sexspecific disruptions in spatial memory and anhedonia in a "two hit" rat model correspond with alterations in hippocampal brain-derived neurotrophic factor expression and signaling: Sex-Specific Effects of Stress on BDNF, Cognition, and Anhedonia. Hippocampus, vol.24, pp.1197-1211, 2014.

I. G. Hislop, Childhood deprivation: an antecedent of the irritable bowel syndrome, Med. J. Aust, vol.1, pp.372-374, 1979.

A. E. Hoban, R. M. Stilling, F. J. Ryan, F. Shanahan, T. G. Dinan et al., Regulation of prefrontal cortex myelination by the microbiota, Transl. Psychiatry, vol.6, p.774, 2016.

A. E. Hoban, R. D. Moloney, A. V. Golubeva, K. A. Mcvey-neufeld, O. O'sullivan et al., Behavioural and neurochemical consequences of chronic gut microbiota depletion during adulthood in the rat, Neuroscience, vol.339, pp.463-477, 2016.

M. A. Hofer, Multiple regulators of ultrasonic vocalization in the infant rat, Psychoneuroendocrinology, vol.21, pp.203-217, 1996.

M. A. Hofer, H. N. Shair, and S. A. Brunelli, Ultrasonic vocalizations in rat and mouse pups, Curr. Protoc. Neurosci. Chapter, vol.8, 2002.

E. B. Hollister, C. Gao, and J. Versalovic, Compositional and functional features of the gastrointestinal microbiome and their effects on human health, Gastroenterology, vol.146, pp.1449-1458, 2014.

P. Holzer, Neuropeptides, Microbiota, and Behavior, Int. Rev. Neurobiol, vol.131, pp.67-89, 2016.

S. Hong, G. Zheng, and J. W. Wiley, Epigenetic regulation of genes that modulate chronic stressinduced visceral pain in the peripheral nervous system, Gastroenterology, vol.148, pp.148-157, 2015.

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.

J. Hui, Z. Zhang, S. Liu, G. Xi, X. Zhang et al., Hippocampal neurochemistry is involved in the behavioural effects of neonatal maternal separation and their reversal by post-weaning environmental enrichment: a magnetic resonance study, Behav. Brain Res, vol.217, pp.122-127, 2011.

H. J. Hulshof, A. Novati, A. Sgoifo, P. G. Luiten, J. A. Boer et al., Maternal separation decreases adult hippocampal cell proliferation and impairs cognitive performance but has little effect on stress sensitivity and anxiety in adult Wistar rats, Behav. Brain Res, vol.216, pp.552-560, 2011.

R. L. Huot, K. V. Thrivikraman, M. J. Meaney, and P. M. Plotsky, Development of adult ethanol preference and anxiety as a consequence of neonatal maternal separation in Long Evans rats and reversal with antidepressant treatment, Psychopharmacology (Berl.), vol.158, pp.366-373, 2001.

R. L. Huot, M. E. Gonzalez, C. O. Ladd, K. V. Thrivikraman, and P. M. Plotsky, Foster litters prevent hypothalamic-pituitary-adrenal axis sensitization mediated by neonatal maternal separation, Psychoneuroendocrinology, vol.29, pp.279-289, 2004.

E. Husebye, P. M. Hellström, and T. Midtvedt, Intestinal microflora stimulates myoelectric activity of rat small intestine by promoting cyclic initiation and aboral propagation of migrating myoelectric complex, Dig. Dis. Sci, vol.39, pp.946-956, 1994.

E. Husebye, P. M. Hellström, F. Sundler, J. Chen, and T. Midtvedt, Influence of microbial species on small intestinal myoelectric activity and transit in germ-free rats, Am. J. Physiol. Gastrointest. Liver Physiol, vol.280, pp.368-380, 2001.

N. P. Hyland, M. Julio-pieper, S. M. O'mahony, D. C. Bulmer, K. Lee et al., A distinct subset of submucosal mast cells undergoes hyperplasia following neonatal maternal separation: a role in visceral hypersensitivity?, Gut, vol.58, pp.1030-1031, 2009.

N. P. Hyland, S. M. O'mahony, D. O'malley, C. M. O'mahony, T. G. Dinan et al., Early-life stress selectively affects gastrointestinal but not behavioral responses in a genetic model of brain-gut axis dysfunction, Neurogastroenterol. Motil, vol.27, pp.105-113, 2015.

M. Iijima and S. Chaki, Separation-induced ultrasonic vocalization in rat pups: Further pharmacological characterization, Pharmacol. Biochem. Behav, vol.82, pp.652-657, 2005.

I. I. Ivanov, K. Atarashi, N. Manel, E. L. Brodie, T. Shima et al., Induction of intestinal Th17 cells by segmented filamentous bacteria, Cell, vol.139, pp.485-498, 2009.

R. Jaenisch and A. Bird, Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals, Nat. Genet, vol.33, pp.245-254, 2003.

N. Jain and W. A. Walker, Diet and host-microbial crosstalk in postnatal intestinal immune homeostasis, Nat. Rev. Gastroenterol. Hepatol, vol.12, pp.14-25, 2015.

H. E. Jakobsson, A. M. Rodríguez-piñeiro, A. Schütte, A. Ermund, P. Boysen et al., The composition of the gut microbiota shapes the colon mucus barrier, EMBO Rep, vol.16, pp.164-177, 2015.

S. M. Jandhyala, R. Talukdar, C. Subramanyam, H. Vuyyuru, M. Sasikala et al., Role of the normal gut microbiota, World J. Gastroenterol, vol.21, pp.8787-8803, 2015.

E. Ja?arevi?, K. E. Morrison, and T. L. Bale, Sex differences in the gut microbiome-brain axis across the lifespan, Philos. Trans. R. Soc. Lond. B. Biol. Sci, vol.371, 2016.

M. C. Jawahar, C. Murgatroyd, E. L. Harrison, and B. T. Baune, Epigenetic alterations following early postnatal stress: a review on novel aetiological mechanisms of common psychiatric disorders, Clin. Epigenetics, vol.7, p.122, 2015.

J. N. Jaworski, D. D. Francis, C. L. Brommer, E. T. Morgan, and M. J. Kuhar, Effects of early maternal separation on ethanol intake, GABA receptors and metabolizing enzymes in adult rats, Psychopharmacology (Berl.), vol.181, pp.8-15, 2005.

S. Jia, Z. Lu, Z. Gao, J. An, X. Wu et al., Chitosan oligosaccharides alleviate cognitive deficits in an amyloid-?1-42-induced rat model of Alzheimer's disease, Int. J. Biol. Macromol, vol.83, pp.416-425, 2016.

Q. Jiao, X. Du, Y. Li, B. Gong, L. Shi et al., The neurological effects of ghrelin in brain diseases: Beyond metabolic functions, Neurosci. Biobehav. Rev, vol.73, pp.98-111, 2017.

A. C. Johnson, L. Tran, J. Schulkin, G. Meerveld, and B. , Importance of stress receptormediated mechanisms in the amygdala on visceral pain perception in an intrinsically anxious rat, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.24, p.219, 2012.

M. Julio-pieper, J. A. Bravo, E. Aliaga, and M. Gotteland, Review article: intestinal barrier dysfunction and central nervous system disorders--a controversial association, Aliment. Pharmacol. Ther, vol.40, pp.1187-1201, 2014.

P. S. Kabouridis and V. Pachnis, Emerging roles of gut microbiota and the immune system in the development of the enteric nervous system, J. Clin. Invest, vol.125, pp.956-964, 2015.

O. Kaidanovich-beilin, T. Lipina, I. Vukobradovic, J. Roder, and J. R. Woodgett, Assessment of Social Interaction Behaviors, J. Vis. Exp. JoVE, 2011.

M. Kalinichev, K. W. Easterling, P. M. Plotsky, and S. G. Holtzman, Long-lasting changes in stressinduced corticosterone response and anxiety-like behaviors as a consequence of neonatal maternal separation in Long-Evans rats, Pharmacol. Biochem. Behav, vol.73, pp.131-140, 2002.

D. Kang, J. G. Park, Z. E. Ilhan, G. Wallstrom, J. Labaer et al., Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children, PloS One, vol.8, p.68322, 2013.

D. Kang, J. B. Adams, A. C. Gregory, T. Borody, L. Chittick et al., Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study, vol.5, p.10, 2017.

A. C. Kao, S. Harty, and P. W. Burnet, The Influence of Prebiotics on Neurobiology and Behavior, Int. Rev. Neurobiol, vol.131, pp.21-48, 2016.

S. Katsouli, A. Stamatakis, P. Giompres, E. D. Kouvelas, F. Stylianopoulou et al., Sexually dimorphic long-term effects of an early life experience on AMPA receptor subunit expression in rat brain, Neuroscience, vol.257, pp.49-64, 2014.

S. E. Kawakami, I. M. Quadros, R. B. Machado, and D. Suchecki, Sex-dependent effects of maternal separation on plasma corticosterone and brain monoamines in response to chronic ethanol administration, Neuroscience, vol.253, pp.55-66, 2013.

J. R. Kelly, P. J. Kennedy, J. F. Cryan, T. G. Dinan, G. Clarke et al., Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders, Front. Cell. Neurosci, vol.9, p.392, 2015.

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, J. Psychiatr. Res, vol.82, pp.109-118, 2016.

K. S. Kendler, M. C. Neale, R. C. Kessler, A. C. Heath, and L. J. Eaves, Childhood parental loss and adult psychopathology in women. A twin study perspective, Arch. Gen. Psychiatry, vol.49, pp.109-116, 1992.

K. S. Kendler, J. W. Kuhn, and C. A. Prescott, Childhood sexual abuse, stressful life events and risk for major depression in women, Psychol. Med, vol.34, pp.1475-1482, 2004.

H. Kettenmann, U. Hanisch, M. Noda, and A. Verkhratsky, Physiology of microglia, Physiol. Rev, vol.91, pp.461-553, 2011.

A. Khoruts, Faecal microbiota transplantation in 2013: developing human gut microbiota as a class of therapeutics, Nat. Rev. Gastroenterol. Hepatol, vol.11, pp.79-80, 2014.

D. Y. Kim and M. Camilleri, Serotonin: a mediator of the brain-gut connection, Am. J. Gastroenterol, vol.95, pp.2698-2709, 2000.

S. Kim, H. Kim, Y. S. Yim, S. Ha, K. Atarashi et al., Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring, Nature, vol.549, pp.528-532, 2017.

D. Klatzmann and A. K. Abbas, The promise of low-dose interleukin-2 therapy for autoimmune and inflammatory diseases, Nat. Rev. Immunol, vol.15, pp.283-294, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01617541

S. L. Klein, The effects of hormones on sex differences in infection: from genes to behavior, Neurosci. Biobehav. Rev, vol.24, pp.627-638, 2000.

T. K. Klooker, B. Braak, R. C. Painter, S. R. De-rooij, R. M. Van-elburg et al., Exposure to severe wartime conditions in early life is associated with an increased risk of irritable bowel syndrome: a population-based cohort study, Am. J. Gastroenterol, vol.104, pp.2250-2256, 2009.

M. Klug and M. Van-den-buuse, Chronic cannabinoid treatment during young adulthood induces sexspecific behavioural deficits in maternally separated rats, Behav. Brain Res, vol.233, pp.305-313, 2012.

I. Knuesel, L. Chicha, M. Britschgi, S. A. Schobel, M. Bodmer et al., Maternal immune activation and abnormal brain development across CNS disorders, Nat. Rev. Neurol, vol.10, pp.643-660, 2014.

A. S. Koe, A. Ashokan, M. , and R. , Short environmental enrichment in adulthood reverses anxiety and basolateral amygdala hypertrophy induced by maternal separation, Transl. Psychiatry, vol.6, p.729, 2016.

I. S. Kohane, A. Mcmurry, G. Weber, D. Macfadden, L. Rappaport et al., The co-morbidity burden of children and young adults with autism spectrum disorders, PloS One, vol.7, p.33224, 2012.

R. A. Kohman, T. K. Bhattacharya, C. Kilby, P. Bucko, and J. S. Rhodes, Effects of minocycline on spatial learning, hippocampal neurogenesis and microglia in aged and adult mice, Behav. Brain Res, vol.242, pp.17-24, 2013.

N. Kokras and C. Dalla, Sex differences in animal models of psychiatric disorders, Br. J. Pharmacol, vol.171, pp.4595-4619, 2014.

N. A. Koloski, M. Jones, J. Kalantar, M. Weltman, J. Zaguirre et al., The brain--gut pathway in functional gastrointestinal disorders is bidirectional: a 12-year prospective population-based study, Gut, vol.61, pp.1284-1290, 2012.

J. König, J. Wells, P. D. Cani, C. L. García-ródenas, T. Macdonald et al., Human Intestinal Barrier Function in Health and Disease, Clin. Transl. Gastroenterol, vol.7, p.196, 2016.

J. P. Konsman, G. N. Luheshi, R. M. Bluthé, and R. Dantzer, The vagus nerve mediates behavioural depression, but not fever, in response to peripheral immune signals; a functional anatomical analysis, Eur. J. Neurosci, vol.12, pp.4434-4446, 2000.

A. Korosi, The pathways from mother's love to baby's future, Front. Behav. Neurosci, vol.3, 2009.

A. Korosi and T. Z. Baram, Plasticity of the stress response early in life: Mechanisms and significance, Dev. Psychobiol, vol.52, pp.661-670, 2010.

M. Kosel and T. E. Schlaepfer, Mechanisms and state of the art of vagus nerve stimulation, J. ECT, vol.18, pp.189-192, 2002.

T. A. Kosten, M. J. Miserendino, and P. Kehoe, Enhanced acquisition of cocaine self-administration in adult rats with neonatal isolation stress experience, Brain Res, vol.875, pp.44-50, 2000.

T. A. Kosten, X. Y. Zhang, and P. Kehoe, Heightened cocaine and food self-administration in female rats with neonatal isolation experience, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.31, pp.70-76, 2006.

T. A. Kosten, H. J. Lee, and J. J. Kim, Early life stress impairs fear conditioning in adult male and female rats, Brain Res, vol.1087, pp.142-150, 2006.

T. A. Kosten, J. J. Kim, and H. J. Lee, Early life manipulations alter learning and memory in rats, Neurosci. Biobehav. Rev, vol.36, 1985.

N. Kratsman, D. Getselter, E. , and E. , Sodium butyrate attenuates social behavior deficits and modifies the transcription of inhibitory/excitatory genes in the frontal cortex of an autism model, Neuropharmacology, vol.102, pp.136-145, 2016.

C. R. Kuhlmann, R. Tamaki, M. Gamerdinger, V. Lessmann, C. Behl et al., Inhibition of the myosin light chain kinase prevents hypoxia-induced blood-brain barrier disruption: ML-7 prevents hypoxic brain edema formation, J. Neurochem, vol.102, pp.501-507, 2007.

H. Kuma, T. Miki, Y. Matsumoto, H. Gu, H. Li et al., Early maternal deprivation induces alterations in brain-derived neurotrophic factor expression in the developing rat hippocampus, Neurosci. Lett, vol.372, pp.68-73, 2004.

H. Kumar and B. Sharma, Minocycline ameliorates prenatal valproic acid induced autistic behaviour, biochemistry and blood brain barrier impairments in rats, Brain Res, vol.1630, pp.83-97, 2016.

M. Kundakovic, S. Lim, K. Gudsnuk, and F. A. Champagne, Sex-Specific and Strain-Dependent Effects of Early Life Adversity on Behavioral and Epigenetic Outcomes. Front, 2013.

J. S. Labus, B. N. Naliboff, J. Fallon, S. M. Berman, B. Suyenobu et al., Sex differences in brain activity during aversive visceral stimulation and its expectation in patients with chronic abdominal pain: a network analysis, NeuroImage, vol.41, pp.1032-1043, 2008.

C. O. Ladd, M. J. Owens, and C. B. Nemeroff, Persistent changes in corticotropin-releasing factor neuronal systems induced by maternal deprivation, Endocrinology, vol.137, pp.1212-1218, 1996.

C. O. Ladd, R. L. Huot, K. V. Thrivikraman, C. B. Nemeroff, M. J. Meaney et al., Longterm behavioral and neuroendocrine adaptations to adverse early experience, Prog. Brain Res, vol.122, pp.81-103, 2000.

L. Lambás-señas, O. Mnie-filali, V. Certin, C. Faure, L. Lemoine et al., Functional correlates for 5-HT1A receptors in maternally deprived rats displaying anxiety and depressionlike behaviors, Prog. Neuropsychopharmacol. Biol. Psychiatry, vol.33, pp.262-268, 2009.

M. B. Larsson, K. Tillisch, A. D. Craig, M. Engström, J. Labus et al., Brain responses to visceral stimuli reflect visceral sensitivity thresholds in patients with irritable bowel syndrome, Gastroenterology, vol.142, pp.463-472, 2012.

S. Leclercq, S. Matamoros, P. D. Cani, A. M. Neyrinck, F. Jamar et al., Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity, Proc. Natl. Acad. Sci. U. S. A, vol.111, pp.4485-4493, 2014.

S. Leclercq, F. M. Mian, A. M. Stanisz, L. B. Bindels, E. Cambier et al., Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior, Nat. Commun, vol.8, p.15062, 2017.

S. Y. Lee and I. Soltesz, Cholecystokinin: a multi-functional molecular switch of neuronal circuits, Dev. Neurobiol, vol.71, pp.83-91, 2011.

J. Lee, H. J. Kim, J. G. Kim, V. Ryu, B. Kim et al., Depressive behaviors and decreased expression of serotonin reuptake transporter in rats that experienced neonatal maternal separation, Neurosci. Res, vol.58, pp.32-39, 2007.

J. Lee, J. Y. Kim, and J. W. Jahng, Highly Palatable Food during Adolescence Improves Anxiety-Like Behaviors and Hypothalamic-Pituitary-Adrenal Axis Dysfunction in Rats that Experienced Neonatal Maternal Separation, Endocrinol. Metab, vol.29, p.169, 2014.

S. Y. Lee, Z. Wang, C. Lin, C. H. Contag, L. C. Olds et al., Regulation of intestine-specific spatiotemporal expression by the rat lactase promoter, J. Biol. Chem, vol.277, pp.13099-13105, 2002.

J. Lehmann, C. R. Pryce, D. Bettschen, and J. Feldon, The maternal separation paradigm and adult emotionality and cognition in male and female Wistar rats, Pharmacol. Biochem. Behav, vol.64, pp.705-715, 1999.

J. Lehmann, H. Russig, J. Feldon, P. , and C. R. , Effect of a single maternal separation at different pup ages on the corticosterone stress response in adult and aged rats, Pharmacol. Biochem. Behav, vol.73, pp.141-145, 2002.

S. Lejeune, N. Dourmap, M. Martres, B. Giros, V. Daugé et al., The dopamine D1 receptor agonist SKF 38393 improves temporal order memory performance in maternally deprived rats, Neurobiol. Learn. Mem, vol.106, pp.268-273, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01542333

K. P. Lemon, G. C. Armitage, D. A. Relman, and M. A. Fischbach, Microbiota-targeted therapies: an ecological perspective, Sci. Transl. Med, vol.4, pp.137-142, 2012.

K. M. Lenz, B. M. Nugent, R. Haliyur, and M. M. Mccarthy, Microglia are essential to masculinization of brain and behavior, J. Neurosci. Off. J. Soc. Neurosci, vol.33, pp.2761-2772, 2013.

D. A. León-rodríguez and Z. Dueñas, Maternal Separation during Breastfeeding Induces Gender-Dependent Changes in Anxiety and the GABA-A Receptor Alpha-Subunit in Adult Wistar Rats, PLoS ONE, vol.8, p.68010, 2013.

S. Levine, Infantile experience and resistance to physiological stress, Science, vol.126, p.405, 1957.

B. Li, A. Zani, C. Lee, E. Zani-ruttenstock, Z. Zhang et al., Endoplasmic reticulum stress is involved in the colonic epithelium damage induced by maternal separation, J. Pediatr. Surg, vol.51, pp.1001-1004, 2016.

B. Li, C. Lee, T. Filler, A. Hock, R. Y. Wu et al., Inhibition of corticotropin-releasing hormone receptor 1 and activation of receptor 2 protect against colonic injury and promote epithelium repair, Sci. Rep, vol.7, p.46616, 2017.

B. Li, C. Lee, Z. Martin, X. Li, Y. Koike et al., Intestinal epithelial injury induced by maternal separation is protected by hydrogen sulfide, J. Pediatr. Surg, vol.52, pp.40-44, 2017.

M. Li, X. Xue, S. Shao, F. Shao, W. et al., Cognitive, emotional and neurochemical effects of repeated maternal separation in adolescent rats, Brain Res, vol.1518, pp.82-90, 2013.

S. Liang, T. Wang, X. Hu, J. Luo, W. Li et al., Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress, Neuroscience, vol.310, pp.561-577, 2015.

P. V. Licciardi, K. Ververis, and T. C. Karagiannis, Histone deacetylase inhibition and dietary shortchain Fatty acids. ISRN Allergy, p.869647, 2011.

M. Lippmann, A. Bress, C. B. Nemeroff, P. M. Plotsky, and L. M. Monteggia, Long-term behavioural and molecular alterations associated with maternal separation in rats: Molecular adaptations after maternal separation, Eur. J. Neurosci, vol.25, pp.3091-3098, 2007.

R. G. Lister, Ethologically-based animal models of anxiety disorders, Pharmacol. Ther, vol.46, pp.321-340, 1990.

Y. Litvin, C. A. Turner, M. B. Rios, P. M. Maras, S. Chaudhury et al., Fibroblast growth factor 2 alters the oxytocin receptor in a developmental model of anxiety-like behavior in male rat pups, Horm. Behav, vol.86, pp.64-70, 2016.

D. Liu, Maternal Care, Hippocampal Glucocorticoid Receptors, and Hypothalamic-Pituitary-Adrenal Responses to Stress, Science, vol.277, pp.1659-1662, 1997.

X. Liu, A. Silverman, M. Kern, B. D. Ward, S. Li et al., Excessive coupling of the salience network with intrinsic neurocognitive brain networks during rectal distension in adolescents with irritable bowel syndrome: a preliminary report, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.28, pp.43-53, 2016.

D. Lotan, M. Cunningham, J. , and D. , Antibiotic treatment attenuates behavioral and neurochemical changes induced by exposure of rats to group a streptococcal antigen, PloS One, vol.9, 2014.

P. Louis and H. J. Flint, Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine, FEMS Microbiol. Lett, vol.294, pp.1-8, 2009.

P. Luczynski, K. Mcvey-neufeld, C. S. Oriach, G. Clarke, T. G. Dinan et al., Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior, Int. J. Neuropsychopharmacol. Off. Sci. J. Coll. Int. Neuropsychopharmacol. CINP, 2016.

P. Luczynski, S. O. Whelan, C. O'sullivan, G. Clarke, F. Shanahan et al., Adult microbiota-deficient mice have distinct dendritic morphological changes: differential effects in the amygdala and hippocampus, Eur. J. Neurosci, vol.44, pp.2654-2666, 2016.

P. Luczynski, M. Tramullas, M. Viola, F. Shanahan, G. Clarke et al., Microbiota regulates visceral pain in the mouse, 2017.

C. Luh, C. R. Kuhlmann, B. Ackermann, R. Timaru-kast, H. J. Luhmann et al., Inhibition of myosin light chain kinase reduces brain edema formation after traumatic brain injury, J. Neurochem, vol.112, pp.1015-1025, 2010.

W. J. Lukiw, Bacteroides fragilis Lipopolysaccharide and Inflammatory Signaling in Alzheimer's, Disease. Front. Microbiol, vol.7, p.1544, 2016.

R. Lundberg, M. F. Toft, B. August, A. K. Hansen, and C. H. Hansen, Antibiotic-treated versus germfree rodents for microbiota transplantation studies, 2016.

S. J. Lupien, S. Parent, A. C. Evans, R. E. Tremblay, P. D. Zelazo et al., Larger amygdala but no change in hippocampal volume in 10-year-old children exposed to maternal depressive symptomatology since birth, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.14324-14329, 2011.

F. S. Luppino, L. M. De-wit, P. F. Bouvy, T. Stijnen, P. Cuijpers et al., Overweight, obesity, and depression: a systematic review and meta-analysis of longitudinal studies, Arch. Gen. Psychiatry, vol.67, pp.220-229, 2010.

P. Lutz and G. Turecki, DNA methylation and childhood maltreatment: From animal models to human studies, Neuroscience, vol.264, pp.142-156, 2014.

W. J. Lynch, L. D. Mangini, and J. R. Taylor, Neonatal isolation stress potentiates cocaine seeking behavior in adult male and female rats, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.30, pp.322-329, 2005.

M. Lyte, Microbial endocrinology: Host-microbiota neuroendocrine interactions influencing brain and behavior, Gut Microbes, vol.5, pp.381-389, 2014.

S. Maccari, P. V. Piazza, M. Kabbaj, A. Barbazanges, H. Simon et al., Adoption reverses the long-term impairment in glucocorticoid feedback induced by prenatal stress, J. Neurosci. Off. J. Soc. Neurosci, vol.15, pp.110-116, 1995.

S. Maccari, H. J. Krugers, S. Morley-fletcher, M. Szyf, and P. J. Brunton, The Consequences of Early-Life Adversity: Neurobiological, Behavioural and Epigenetic Adaptations, J. Neuroendocrinol, vol.26, pp.707-723, 2014.

G. T. Macfarlane, G. R. Gibson, and J. H. Cummings, Comparison of fermentation reactions in different regions of the human colon, J. Appl. Bacteriol, vol.72, pp.57-64, 1992.

A. J. Macpherson and N. L. Harris, Interactions between commensal intestinal bacteria and the immune system, Nat. Rev. Immunol, vol.4, pp.478-485, 2004.

G. M. Macqueen, K. Ramakrishnan, R. Ratnasingan, B. Chen, Y. et al., Desipramine treatment reduces the long-term behavioural and neurochemical sequelae of early-life maternal separation, Int. J. Neuropsychopharmacol, vol.6, pp.391-396, 2003.

S. Macrí, G. J. Mason, and H. Würbel, Dissociation in the effects of neonatal maternal separations on maternal care and the offspring's HPA and fear responses in rats, Eur. J. Neurosci, vol.20, pp.1017-1024, 2004.

J. L. Madara, Sodium-glucose cotransport and epithelial permeability, Gastroenterology, vol.107, pp.319-320, 1994.

M. Maes, The cytokine hypothesis of depression: inflammation, oxidative & nitrosative stress (IO&NS) and leaky gut as new targets for adjunctive treatments in depression, Neuro Endocrinol. Lett, vol.29, pp.287-291, 2008.

M. Maes and J. Leunis, Normalization of leaky gut in chronic fatigue syndrome (CFS) is accompanied by a clinical improvement: effects of age, duration of illness and the translocation of LPS from gram-negative bacteria, Neuro Endocrinol. Lett, vol.29, pp.902-910, 2008.

M. Maes, M. Kubera, and J. Leunis, The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression, Neuro Endocrinol. Lett, vol.29, pp.117-124, 2008.

L. De-magistris, V. Familiari, A. Pascotto, A. Sapone, A. Frolli et al., Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives, J. Pediatr. Gastroenterol. Nutr, vol.51, pp.418-424, 2010.

V. Maletic, M. Robinson, T. Oakes, S. Iyengar, S. G. Ball et al., Neurobiology of depression: an integrated view of key findings, Int. J. Clin. Pract, vol.61, pp.2030-2040, 2007.

O. Malkesman, M. L. Scattoni, D. Paredes, T. Tragon, B. Pearson et al., The female urine sniffing test: a novel approach for assessing reward-seeking behavior in rodents, Biol. Psychiatry, vol.67, pp.864-871, 2010.

F. Mangiola, G. Ianiro, F. Franceschi, S. Fagiuoli, G. Gasbarrini et al., Gut microbiota in autism and mood disorders, World J. Gastroenterol, vol.22, pp.361-368, 2016.

J. Maniam and M. J. Morris, Voluntary exercise and palatable high-fat diet both improve behavioural profile and stress responses in male rats exposed to early life stress: Role of hippocampus, Psychoneuroendocrinology, vol.35, pp.1553-1564, 2010.

J. Maniam and M. J. Morris, Palatable cafeteria diet ameliorates anxiety and depression-like symptoms following an adverse early environment, Psychoneuroendocrinology, vol.35, pp.717-728, 2010.

J. Maniam and M. J. Morris, Long-term postpartum anxiety and depression-like behavior in mother rats subjected to maternal separation are ameliorated by palatable high fat diet, Behav. Brain Res, vol.208, pp.72-79, 2010.

L. Marais, S. J. Van-rensburg, J. M. Van-zyl, D. J. Stein, and W. M. Daniels, Maternal separation of rat pups increases the risk of developing depressive-like behavior after subsequent chronic stress by altering corticosterone and neurotrophin levels in the hippocampus, Neurosci. Res, vol.61, pp.106-112, 2008.

A. Marín-burgin and A. F. Schinder, Requirement of adult-born neurons for hippocampus-dependent learning, Behav. Brain Res, vol.227, pp.391-399, 2012.

J. G. Markle, D. N. Frank, S. Mortin-toth, C. E. Robertson, L. M. Feazel et al., Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity, Science, vol.339, pp.1084-1088, 2013.

M. Marmendal, E. Roman, C. J. Eriksson, I. Nylander, and C. Fahlke, Maternal separation alters maternal care, but has minor effects on behavior and brain opioid peptides in adult offspring, Dev. Psychobiol, vol.45, pp.140-152, 2004.

F. A. Marvel, C. Chen, N. Badr, R. P. Gaykema, and L. E. Goehler, Reversible inactivation of the dorsal vagal complex blocks lipopolysaccharide-induced social withdrawal and c-Fos expression in central autonomic nuclei, Brain. Behav. Immun, vol.18, pp.123-134, 2004.

O. Matcovitch-natan, D. R. Winter, A. Giladi, S. Vargas-aguilar, A. Spinrad et al., Microglia development follows a stepwise program to regulate brain homeostasis, Science, vol.353, p.8670, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01438174

M. Matsumoto, R. Kibe, T. Ooga, Y. Aiba, E. Sawaki et al., Cerebral lowmolecular metabolites influenced by intestinal microbiota: a pilot study, Front. Syst. Neurosci, vol.7, p.9, 2013.

D. Mattei, A. Djodari-irani, R. Hadar, A. Pelz, L. F. De-cossío et al., Minocycline rescues decrease in neurogenesis, increase in microglia cytokines and deficits in sensorimotor gating in an animal model of schizophrenia, Brain. Behav. Immun, vol.38, pp.175-184, 2014.

D. M. Matthews and S. M. Jenks, Ingestion of Mycobacterium vaccae decreases anxiety-related behavior and improves learning in mice, Behav. Processes, vol.96, pp.27-35, 2013.

K. Matthews, J. W. Dalley, C. Matthews, T. Hu-tsai, and T. W. Robbins, Periodic maternal separation of neonatal rats produces region-and gender-specific effects on biogenic amine content in postmortem adult brain, Synapse, vol.40, pp.1-10, 2001.

F. Mattioli, C. Fucile, V. Marini, L. Isola, F. Montanaro et al., Assessment of intestinal permeability using sugar probes: influence of urinary volume, Clin. Lab, vol.57, pp.909-918, 2011.

E. A. Mayer, Gut feelings: the emerging biology of gut-brain communication, Nat. Rev. Neurosci, vol.12, pp.453-466, 2011.

E. A. Mayer, B. Naliboff, O. Lee, J. Munakata, C. et al., Review article: gender-related differences in functional gastrointestinal disorders, Aliment. Pharmacol. Ther, vol.13, issue.2, pp.65-69, 1999.

E. A. Mayer, R. Knight, S. K. Mazmanian, J. F. Cryan, and K. Tillisch, Gut Microbes and the Brain: Paradigm Shift in Neuroscience, J. Neurosci, vol.34, pp.15490-15496, 2014.

E. A. Mayer, D. Padua, and K. Tillisch, Altered brain-gut axis in autism: comorbidity or causative mechanisms?, BioEssays News Rev. Mol. Cell. Dev. Biol, vol.36, pp.933-939, 2014.

T. M. Maynard, L. Sikich, J. A. Lieberman, and A. S. Lamantia, Neural development, cell-cell signaling, and the "two-hit" hypothesis of schizophrenia, Schizophr. Bull, vol.27, pp.457-476, 2001.

M. M. Mccarthy, B. M. Nugent, and K. M. Lenz, Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain, Nat. Rev. Neurosci, vol.18, pp.471-484, 2017.

J. Mccauley, D. E. Kern, K. Kolodner, L. Dill, A. F. Schroeder et al., Clinical characteristics of women with a history of childhood abuse: unhealed wounds, JAMA, vol.277, pp.1362-1368, 1997.

B. S. Mcewen, Stress, adaptation, and disease. Allostasis and allostatic load, Ann. N. Y. Acad. Sci, vol.840, pp.33-44, 1998.

P. O. Mcgowan, Epigenomic Mechanisms of Early Adversity and HPA Dysfunction: Considerations for PTSD Research. Front, 2013.

P. O. Mcgowan, A. Sasaki, A. C. D'alessio, S. Dymov, B. Labonté et al., Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse, Nat. Neurosci, vol.12, pp.342-348, 2009.

D. P. Mckernan, A. Nolan, E. K. Brint, S. M. O'mahony, N. P. Hyland et al., Toll-like receptor mRNA expression is selectively increased in the colonic mucosa of two animal models relevant to irritable bowel syndrome, PloS One, vol.4, p.8226, 2009.

D. B. Mckim, A. Niraula, A. J. Tarr, E. S. Wohleb, J. F. Sheridan et al., Neuroinflammatory Dynamics Underlie Memory Impairments after Repeated Social Defeat, J. Neurosci. Off. J. Soc. Neurosci, vol.36, pp.2590-2604, 2016.

K. A. Mclaughlin, L. D. Kubzansky, E. C. Dunn, R. Waldinger, G. Vaillant et al., Childhood social environment, emotional reactivity to stress, and mood and anxiety disorders across the life course, Depress. Anxiety, vol.27, pp.1087-1094, 2010.

M. J. Meaney, Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations, Annu. Rev. Neurosci, vol.24, pp.1161-1192, 2001.

M. J. Meaney and M. Szyf, Environmental programming of stress responses through DNA methylation: life at the interface between a dynamic environment and a fixed genome, Dialogues Clin. Neurosci, vol.7, pp.103-123, 2005.

M. J. Meaney, J. B. Mitchell, D. H. Aitken, S. Bhatnagar, S. R. Bodnoff et al., The effects of neonatal handling on the development of the adrenocortical response to stress: implications for neuropathology and cognitive deficits in later life, Psychoneuroendocrinology, vol.16, pp.85-103, 1991.

M. J. Meaney, V. Viau, S. Bhatnagar, K. Betito, L. J. Iny et al., Cellular mechanisms underlying the development and expression of individual differences in the hypothalamicpituitary-adrenal stress response, J. Steroid Biochem. Mol. Biol, vol.39, pp.265-274, 1991.

M. J. Meaney, D. H. Aitken, S. Bhatnagar, and R. M. Sapolsky, Postnatal handling attenuates certain neuroendocrine, anatomical, and cognitive dysfunctions associated with aging in female rats, Neurobiol. Aging, vol.12, pp.31-38, 1991.

J. B. Meddings and I. Gibbons, Discrimination of site-specific alterations in gastrointestinal permeability in the rat, Gastroenterology, vol.114, pp.83-92, 1998.

M. A. Mehta, N. I. Golembo, C. Nosarti, E. Colvert, A. Mota et al., Amygdala, hippocampal and corpus callosum size following severe early institutional deprivation: The English and Romanian Adoptees Study Pilot, J. Child Psychol. Psychiatry, vol.50, pp.943-951, 2009.

M. A. Mehta, E. Gore-langton, N. Golembo, E. Colvert, S. C. Williams et al., Hyporesponsive reward anticipation in the basal ganglia following severe institutional deprivation early in life, J. Cogn. Neurosci, vol.22, pp.2316-2325, 2010.

S. Ménard, N. Cerf-bensussan, and M. Heyman, Multiple facets of intestinal permeability and epithelial handling of dietary antigens, Mucosal Immunol, vol.3, pp.247-259, 2010.

J. Meng, G. M. Sindberg, R. , and S. , Disruption of gut homeostasis by opioids accelerates HIV disease progression, Front. Microbiol, vol.6, p.643, 2015.

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, Br. J. Nutr, vol.105, pp.755-764, 2011.

C. C. Michaels and S. G. Holtzman, Enhanced sensitivity to naltrexone-induced drinking suppression of fluid intake and sucrose consumption in maternally separated rats, Pharmacol. Biochem. Behav, vol.86, pp.784-796, 2007.

M. J. Millan, G. M. Goodwin, A. Meyer-lindenberg, O. Ögren, and S. , Learning from the past and looking to the future: Emerging perspectives for improving the treatment of psychiatric disorders, Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol, vol.25, pp.599-656, 2015.

B. R. Miller and R. Hen, The current state of the neurogenic theory of depression and anxiety, Curr. Opin. Neurobiol, vol.30, pp.51-58, 2015.

M. Million and M. Larauche, Stress, sex, and the enteric nervous system, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.28, pp.1283-1289, 2016.

M. Million, L. Wang, Y. Wang, D. W. Adelson, P. Yuan et al., CRF2 receptor activation prevents colorectal distension induced visceral pain and spinal ERK1/2 phosphorylation in rats, Gut, vol.55, pp.172-181, 2006.

R. A. Millstein and A. Holmes, Effects of repeated maternal separation on anxiety-and depressionrelated phenotypes in different mouse strains, Neurosci. Biobehav. Rev, vol.31, pp.3-17, 2007.

C. Mirescu, J. D. Peters, and E. Gould, Early life experience alters response of adult neurogenesis to stress, Nat. Neurosci, vol.7, pp.841-846, 2004.

K. M. Moench and C. L. Wellman, Stress-induced alterations in prefrontal dendritic spines: Implications for post-traumatic stress disorder, Neurosci. Lett, vol.601, pp.41-45, 2015.

M. Moffett, A. Vicentic, M. Kozel, P. Plotsky, D. Francis et al., Maternal Separation Alters Drug Intake Patterns in Adulthood in Rats, Biochem. Pharmacol, vol.73, pp.321-330, 2007.

L. Möhle, D. Mattei, M. M. Heimesaat, S. Bereswill, A. Fischer et al., Ly6C(hi) Monocytes Provide a Link between Antibiotic-Induced Changes in Gut Microbiota and Adult Hippocampal Neurogenesis, Cell Rep, vol.15, pp.1945-1956, 2016.

M. Moisan, L. Moal, and M. , Le stress dans tous ses états, Médecine/Sciences, vol.28, pp.612-617, 2012.

J. Molet, P. M. Maras, S. Avishai-eliner, and T. Z. Baram, Naturalistic rodent models of chronic earlylife stress, Dev. Psychobiol, vol.56, pp.1675-1688, 2014.

R. D. Moloney, A. C. Johnson, S. M. O'mahony, T. G. Dinan, B. Greenwood-van-meerveld et al., Stress and the Microbiota-Gut-Brain Axis in Visceral Pain: Relevance to Irritable Bowel Syndrome, CNS Neurosci. Ther, 2015.

R. D. Moloney, R. M. Stilling, T. G. Dinan, and J. F. Cryan, Early-life stress-induced visceral hypersensitivity and anxiety behavior is reversed by histone deacetylase inhibition, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.27, pp.1831-1836, 2015.

R. D. Moloney, T. G. Dinan, and J. F. Cryan, Strain-dependent variations in visceral sensitivity: relationship to stress, anxiety and spinal glutamate transporter expression, Genes Brain Behav, vol.14, pp.319-329, 2015.

E. Monroy, E. Hernández-torres, and G. Flores, Maternal separation disrupts dendritic morphology of neurons in prefrontal cortex, hippocampus, and nucleus accumbens in male rat offspring, J. Chem. Neuroanat, vol.40, pp.93-101, 2010.

R. K. Montgomery, A. E. Mulberg, and R. J. Grand, Development of the human gastrointestinal tract: twenty years of progress, Gastroenterology, vol.116, pp.702-731, 1999.

J. C. Morales-medina, Y. Dumont, C. Benoit, S. Bastianetto, G. Flores et al., Role of neuropeptide Y Y? and Y? receptors on behavioral despair in a rat model of depression with co-morbid anxiety, Neuropharmacology, vol.62, pp.200-208, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01001776

R. Moriez, C. Salvador-cartier, V. Theodorou, J. Fioramonti, H. Eutamene et al., Myosin light chain kinase is involved in lipopolysaccharide-induced disruption of colonic epithelial barrier and bacterial translocation in rats, Am. J. Pathol, vol.167, pp.1071-1079, 2005.

V. Mourlon, A. Baudin, O. Blanc, A. Lauber, B. Giros et al., Maternal deprivation induces depressive-like behaviours only in female rats, Behav. Brain Res, vol.213, pp.278-287, 2010.

V. Mourlon, L. Naudon, B. Giros, M. Crumeyrolle-arias, and V. Daugé, Early stress leads to effects on estrous cycle and differential responses to stress, Physiol. Behav, vol.102, pp.304-310, 2011.

N. Moussaoui, V. Braniste, A. Ait-belgnaoui, M. Gabanou, S. Sekkal et al., Changes in intestinal glucocorticoid sensitivity in early life shape the risk of epithelial barrier defect in maternal-deprived rats, PloS One, vol.9, p.88382, 2014.

N. Moussaoui, M. Larauche, M. Biraud, J. Molet, M. Million et al., Limited Nesting Stress Alters Maternal Behavior and In Vivo Intestinal Permeability in Male Wistar Pup Rats, PloS One, vol.11, 2016.

N. Moussaoui, J. P. Jacobs, M. Larauche, M. Biraud, M. Million et al., Chronic Early-life Stress in Rat Pups Alters Basal Corticosterone, Intestinal Permeability, and Fecal Microbiota at Weaning: Influence of Sex, J. Neurogastroenterol. Motil, vol.23, pp.135-143, 2017.

A. Moya-pérez, A. Perez-villalba, A. Benítez-páez, I. Campillo, and Y. Sanz, Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice, Brain. Behav. Immun, 2017.

A. Muhammad and B. Kolb, Maternal separation altered behavior and neuronal spine density without influencing amphetamine sensitization, Behav. Brain Res, vol.223, pp.7-16, 2011.

A. Muhammad, C. Carroll, and B. Kolb, Stress during development alters dendritic morphology in the nucleus accumbens and prefrontal cortex, Neuroscience, vol.216, pp.103-109, 2012.

J. G. Mulle, W. G. Sharp, and J. F. Cubells, The gut microbiome: a new frontier in autism research, Curr. Psychiatry Rep, vol.15, p.337, 2013.

P. E. Mullen, J. L. Martin, J. C. Anderson, S. E. Romans, and G. P. Herbison, The long-term impact of the physical, emotional, and sexual abuse of children: a community study, Child Abuse Negl, vol.20, pp.7-21, 1996.

T. Murakami, K. Kamada, K. Mizushima, Y. Higashimura, K. Katada et al., Changes in Intestinal Motility and Gut Microbiota Composition in a Rat Stress Model, Digestion, vol.95, pp.55-60, 2017.

C. Murgatroyd, A. V. Patchev, Y. Wu, V. Micale, Y. Bockmühl et al., Dynamic DNA methylation programs persistent adverse effects of early-life stress, Nat. Neurosci, vol.12, pp.1559-1566, 2009.

K. Musholt, G. Cirillo, C. Cavaliere, R. Bianco, M. Bock et al., Neonatal separation stress reduces glial fibrillary acidic protein-and S100beta-immunoreactive astrocytes in the rat medial precentral cortex, Dev. Neurobiol, vol.69, pp.203-211, 2009.

B. Myers, G. Meerveld, and B. , Differential involvement of amygdala corticosteroid receptors in visceral hyperalgesia following acute or repeated stress, Am. J. Physiol. Gastrointest. Liver Physiol, vol.302, pp.260-266, 2012.

N. Nagy and A. M. Goldstein, Enteric nervous system development: A crest cell's journey from neural tube to colon, Semin. Cell Dev. Biol, vol.66, pp.94-106, 2017.

A. Naseribafrouei, K. Hestad, E. Avershina, M. Sekelja, A. Linløkken et al., Correlation between the human fecal microbiota and depression, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.1155-1162, 2014.

L. Naudon, F. Piscitelli, B. Giros, V. Di-marzo, and V. Daugé, Possible involvement of endocannabinoids in the increase of morphine consumption in maternally deprived rat, Neuropharmacology, vol.65, pp.193-199, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01542325

E. Nederhof and M. V. Schmidt, Mismatch or cumulative stress: Toward an integrated hypothesis of programming effects, Physiol. Behav, vol.106, pp.691-700, 2012.

C. B. Nemeroff, Paradise Lost: The Neurobiological and Clinical Consequences of Child Abuse and Neglect, Neuron, vol.89, pp.892-909, 2016.

E. J. Nestler and S. E. Hyman, Animal models of neuropsychiatric disorders, Nat. Neurosci, vol.13, pp.1161-1169, 2010.

K. M. Neufeld, N. Kang, J. Bienenstock, and J. A. Foster, Effects of intestinal microbiota on anxiety-like behavior, Commun. Integr. Biol, vol.4, pp.492-494, 2011.

K. M. Neufeld, N. Kang, J. Bienenstock, and J. A. Foster, Reduced anxiety-like behavior and central neurochemical change in germ-free mice, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.23, p.119, 2011.

N. C. Nicolaides, E. Charmandari, G. P. Chrousos, and T. Kino, Circadian endocrine rhythms: the hypothalamic-pituitary-adrenal axis and its actions, Ann. N. Y. Acad. Sci, vol.1318, pp.71-80, 2014.

R. M. Norman and A. K. Malla, A prospective study of daily stressors and symptomatology in schizophrenic patients, Soc. Psychiatry Psychiatr. Epidemiol, vol.29, pp.244-249, 1994.

R. Nusslock and G. E. Miller, Early-Life Adversity and Physical and Emotional Health Across the Lifespan: A Neuroimmune Network Hypothesis, Biol. Psychiatry, vol.80, pp.23-32, 2016.

E. S. Ogbonnaya, G. Clarke, F. Shanahan, T. G. Dinan, J. F. Cryan et al., Adult Hippocampal Neurogenesis Is Regulated by the Microbiome, Biol. Psychiatry, vol.78, pp.7-9, 2015.

L. Öhman and M. Simrén, Pathogenesis of IBS: role of inflammation, immunity and neuroimmune interactions, Nat. Rev. Gastroenterol. 38 Hepatol, vol.7, pp.163-173, 2010.

K. Ohta, T. Miki, K. Warita, S. Suzuki, T. Kusaka et al., Prolonged maternal separation disturbs the serotonergic system during early brain development, Int. J. Dev. Neurosci, vol.33, pp.15-21, 2014.

E. Øines, R. Murison, J. Mrdalj, J. Grønli, and A. M. Milde, Neonatal maternal separation in male rats increases intestinal permeability and affects behavior after chronic social stress, Physiol. Behav, vol.105, pp.1058-1066, 2012.

B. D. Oladeji and O. Gureje, The comorbidity between depression and diabetes, Curr. Psychiatry Rep, vol.15, p.390, 2013.

E. Oliveros, M. Ramirez, E. Vazquez, A. Barranco, A. Gruart et al., Oral supplementation of 2'-fucosyllactose during lactation improves memory and learning in rats, J. Nutr. Biochem, vol.31, pp.20-27, 2016.

S. O'mahony, A. S. Chua, E. M. Quigley, G. Clarke, F. Shanahan et al., Evidence of an enhanced central 5HT response in irritable bowel syndrome and in the rat maternal separation model, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.20, pp.680-688, 2008.

S. M. O'mahony, J. R. Marchesi, P. Scully, C. Codling, A. Ceolho et al., Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses, Biol. Psychiatry, vol.65, pp.263-267, 2009.

S. M. O'mahony, N. P. Hyland, T. G. Dinan, and J. F. Cryan, Maternal separation as a model of brain-gut axis dysfunction, Psychopharmacology (Berl.), vol.214, pp.71-88, 2011.

S. M. O'mahony, V. D. Felice, K. Nally, H. M. Savignac, M. J. Claesson et al., Disturbance of the gut microbiota in early-life selectively affects visceral pain in adulthood without impacting cognitive or anxiety-related behaviors in male rats, Neuroscience, vol.277, pp.885-901, 2014.

D. O'malley, M. Julio-pieper, S. M. Gibney, T. G. Dinan, and J. F. Cryan, Distinct alterations in colonic morphology and physiology in two rat models of enhanced stress-induced anxiety and depression-like behaviour, Stress Amst. Neth, vol.13, pp.114-122, 2010.

J. J. O'shea, A. Ma, and P. Lipsky, Cytokines and autoimmunity, Nat. Rev. Immunol, vol.2, pp.37-45, 2002.

S. J. Otto, S. R. Mccorkle, J. Hover, C. Conaco, J. Han et al., A new binding motif for the transcriptional repressor REST uncovers large gene networks devoted to neuronal functions, J. Neurosci. Off. J. Soc. Neurosci, vol.27, pp.6729-6739, 2007.

H. Park and M. Poo, Neurotrophin regulation of neural circuit development and function, Nat. Rev. Neurosci, vol.14, pp.7-23, 2012.

H. Park, D. Yoo, S. Kwon, T. Yoo, H. Park et al., Acupuncture stimulation at HT7 alleviates depression-induced behavioral changes via regulation of the serotonin system in the prefrontal cortex of maternally-separated rat pups, J. Physiol. Sci. JPS, vol.62, pp.351-357, 2012.

H. J. Park, S. K. Kim, W. S. Kang, J. Woo, and J. W. Kim, Effects of essential oil from Chamaecyparis obtusa on cytokine genes in the hippocampus of maternal separation rats. Can, J. Physiol. Pharmacol, vol.92, pp.95-101, 2014.

S. W. Park, J. G. Lee, M. K. Seo, N. N. Ly, C. H. Lee et al., Epigenetic modification of glucocorticoid receptor promoter I7 in maternally separated and restraint-stressed rats, Neurosci. Lett, vol.650, pp.38-44, 2017.

V. K. Patchev, A. Montkowski, D. Rouskova, L. Koranyi, F. Holsboer et al., Neonatal treatment of rats with the neuroactive steroid tetrahydrodeoxycorticosterone (THDOC) abolishes the behavioral and neuroendocrine consequences of adverse early life events, J. Clin. Invest, vol.99, pp.962-966, 1997.

L. Paternain, E. Martisova, J. Campión, J. A. Martínez, M. J. Ramírez et al., Methyl donor supplementation in rats reverses the deleterious effect of maternal separation on depression-like behaviour, Behav. Brain Res, vol.299, pp.51-58, 2016.

A. M. Patterson, I. E. Mulder, A. J. Travis, A. Lan, N. Cerf-bensussan et al., Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity, Front. Immunol, vol.8, p.1166, 2017.

D. Pelcovitz, S. Kaplan, B. Goldenberg, F. Mandel, J. Lehane et al., Post-traumatic stress disorder in physically abused adolescents, J. Am. Acad. Child Adolesc. Psychiatry, vol.33, pp.305-312, 1994.

C. J. Peña, H. G. Kronman, D. M. Walker, H. M. Cates, R. C. Bagot et al., Early life stress confers lifelong stress susceptibility in mice via ventral tegmental area OTX2, Science, vol.356, pp.1185-1188, 2017.

J. Penders, C. Thijs, C. Vink, F. F. Stelma, B. Snijders et al., Factors influencing the composition of the intestinal microbiota in early infancy, Pediatrics, vol.118, pp.511-521, 2006.

L. Peng, Z. Li, R. S. Green, I. R. Holzman, L. et al., Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers, J. Nutr, vol.139, pp.1619-1625, 2009.

N. K. Phillips, C. L. Hammen, P. A. Brennan, J. M. Najman, and W. Bor, Early Adversity and the Prospective Prediction of Depressive and Anxiety Disorders in Adolescents, J. Abnorm. Child Psychol, vol.33, pp.13-24, 2005.

C. Pickering, L. Gustafsson, A. Cebere, I. Nylander, and S. Liljequist, Repeated maternal separation of male Wistar rats alters glutamate receptor expression in the hippocampus but not the prefrontal cortex, Brain Res, vol.1099, pp.101-108, 2006.

M. Pigrau, B. K. Rodiño-janeiro, M. Casado-bedmar, B. Lobo, M. Vicario et al., The joint power of sex and stress to modulate brain-gut-microbiota axis and intestinal barrier homeostasis: implications for irritable bowel syndrome, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.28, pp.463-486, 2016.

R. M. Pinheiro, M. N. De-lima, B. C. Portal, S. B. Busato, L. Falavigna et al., Long-lasting recognition memory impairment and alterations in brain levels of cytokines and BDNF induced by maternal deprivation: effects of valproic acid and topiramate, J. Neural Transm, 2014.

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.

K. Ploj, E. Roman, and I. Nylander, Long-term effects of maternal separation on ethanol intake and brain opioid and dopamine receptors in male Wistar rats, Neuroscience, vol.121, pp.787-799, 2003.

K. Ploj, E. Roman, and I. Nylander, Long-term effects of short and long periods of maternal separation on brain opioid peptide levels in male Wistar rats, Neuropeptides, vol.37, pp.149-156, 2003.

P. M. Plotsky and M. J. Meaney, Early, postnatal experience alters hypothalamic corticotropin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats, Brain Res. Mol. Brain Res, vol.18, pp.195-200, 1993.

J. Pohl, M. C. Olmstead, K. E. Wynne-edwards, K. Harkness, and J. L. Menard, Repeated exposure to stress across the childhood-adolescent period alters rats' anxiety-and depression-like behaviors in adulthood: The importance of stressor type and gender, Behav. Neurosci, vol.121, pp.462-474, 2007.

R. D. Porsolt, M. Le-pichon, J. , and M. , Depression: a new animal model sensitive to antidepressant treatments, Nature, vol.266, pp.730-732, 1977.

M. Prinz and J. Priller, Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease, Nat. Rev. Neurosci, vol.15, pp.300-312, 2014.

N. Provençal and E. B. Binder, The effects of early life stress on the epigenome: From the womb to adulthood and even before, Exp. Neurol, vol.268, pp.10-20, 2015.

D. K. Prusator, G. Meerveld, and B. , Sex differences in stress-induced visceral hypersensitivity following early life adversity: a two hit model, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, 2016.

D. K. Prusator, G. Meerveld, and B. , Sex-related differences in pain behaviors following three early life stress paradigms, Biol. Sex Differ, vol.7, p.29, 2016.

D. K. Prusator, G. Meerveld, and B. , Amygdala-mediated mechanisms regulate visceral hypersensitivity in adult females following early life stress: importance of the glucocorticoid receptor and corticotropin-releasing factor, Pain, vol.158, pp.296-305, 2017.

L. Prut and C. Belzung, The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: a review, Eur. J. Pharmacol, vol.463, pp.3-33, 2003.

C. R. Pryce and J. Feldon, Long-term neurobehavioural impact of the postnatal environment in rats: manipulations, effects and mediating mechanisms, Neurosci. Biobehav. Rev, vol.27, pp.57-71, 2003.

C. R. Pryce, D. Bettschen, N. I. Nanz-bahr, and J. Feldon, Comparison of the effects of early handling and early deprivation on conditioned stimulus, context, and spatial learning and memory in adult rats, Behav. Neurosci, vol.117, pp.883-893, 2003.

M. Pusalkar, D. Suri, A. Kelkar, A. Bhattacharya, S. Galande et al., Early stress evokes dysregulation of histone modifiers in the medial prefrontal cortex across the life span, Dev. Psychobiol, vol.58, pp.198-210, 2016.

M. M. Pusceddu, S. El-aidy, F. Crispie, O. O'sullivan, P. Cotter et al., N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota, PloS One, vol.10, 2015.

B. Pyndt-jørgensen, L. Krych, T. B. Pedersen, N. Plath, J. P. Redrobe et al., Investigating the long-term effect of subchronic phencyclidinetreatment on novel object recognition and the association between the gut microbiota and behavior in the animal model of schizophrenia, Physiol. Behav, vol.141, pp.32-39, 2015.

F. J. Raabe and D. Spengler, Epigenetic Risk Factors in PTSD and Depression. Front, 2013.

C. L. Raison, L. Capuron, and A. H. Miller, Cytokines sing the blues: inflammation and the pathogenesis of depression, Trends Immunol, vol.27, pp.24-31, 2006.

M. Rajili?-stojanovi?, E. Biagi, H. G. Heilig, K. Kajander, R. A. Kekkonen et al., Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome, Gastroenterology, vol.141, pp.1792-1801, 2011.

R. M. Ransohoff and V. H. Perry, Microglial physiology: unique stimuli, specialized responses, Annu. Rev. Immunol, vol.27, pp.119-145, 2009.

M. Rao, G. , and M. D. , The bowel and beyond: the enteric nervous system in neurological disorders, Nat. Rev. Gastroenterol. Hepatol, vol.13, pp.517-528, 2016.

A. V. Rao, A. C. Bested, T. M. Beaulne, M. A. Katzman, C. Iorio et al., A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome, Gut Pathog, vol.1, p.6, 2009.

U. Rao, L. Chen, A. S. Bidesi, M. U. Shad, M. A. Thomas et al., Hippocampal changes associated with early-life adversity and vulnerability to depression, Biol. Psychiatry, vol.67, pp.357-364, 2010.

S. J. Reiser, K. A. Mcmillan, K. D. Wright, A. , and G. J. , Adverse childhood experiences and health anxiety in adulthood, Child Abuse Negl, vol.38, pp.407-413, 2014.

J. Reunanen, V. Kainulainen, L. Huuskonen, N. Ottman, C. Belzer et al., Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer, Appl. Environ. Microbiol, vol.81, pp.3655-3662, 2015.

G. Z. Réus, R. B. Stringari, K. F. Ribeiro, A. L. Cipriano, B. S. Panizzutti et al., Maternal deprivation induces depressive-like behaviour and alters neurotrophin levels in the rat brain, Neurochem. Res, vol.36, pp.460-466, 2011.

G. Z. Réus, G. C. Fernandes, A. B. De-moura, R. H. Silva, A. C. Darabas et al., Early life experience contributes to the developmental programming of depressive-like behaviour, neuroinflammation and oxidative stress, J. Psychiatr. Res, vol.95, pp.196-207, 2017.

L. Rinaman, L. Banihashemi, and T. J. Koehnle, Early life experience shapes the functional organization of stress-responsive visceral circuits, Physiol. Behav, vol.104, pp.632-640, 2011.

M. Rincel, A. L. Lépinay, P. Delage, J. Fioramonti, V. S. Théodorou et al., Maternal high-fat diet prevents developmental programming by early-life stress, Transl. Psychiatry, vol.6, p.966, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602432

A. C. Rios, P. K. Maurya, M. Pedrini, M. Zeni-graiff, E. Asevedo et al., Microbiota abnormalities and the therapeutic potential of probiotics in the treatment of mood disorders, Rev. Neurosci, 2017.

M. A. Rivarola, R. , and G. M. , What we know about the long-term consequences of early maternal separation and neuroendocrine response to stress, Rev. De, vol.17, 2014.

M. A. Rivarola and M. M. Suárez, Early maternal separation and chronic variable stress in adulthood changes the neural activity and the expression of glucocorticoid receptor in limbic structures, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.27, pp.567-574, 2009.

M. Roceri, F. Cirulli, C. Pessina, P. Peretto, G. Racagni et al., Postnatal repeated maternal deprivation produces age-dependent changes of brain-derived neurotrophic factor expression in selected rat brain regions, Biol. Psychiatry, vol.55, pp.708-714, 2004.

G. B. Rogers, D. J. Keating, R. L. Young, M. Wong, J. Licinio et al., From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways, Mol. Psychiatry, vol.21, pp.738-748, 2016.

E. Roman and I. Nylander, The impact of emotional stress early in life on adult voluntary ethanol intake-results of maternal separation in rats, Stress Amst. Neth, vol.8, pp.157-174, 2005.

E. Roman, L. Gustafsson, P. Hyytiä, and I. Nylander, Short and prolonged periods of maternal separation and voluntary ethanol intake in male and female ethanol-preferring AA and ethanol-avoiding ANA rats, Alcohol. Clin. Exp. Res, vol.29, pp.591-601, 2005.

E. Roman, L. Gustafsson, M. Berg, and I. Nylander, Behavioral profiles and stress-induced corticosteroid secretion in male Wistar rats subjected to short and prolonged periods of maternal separation, Horm. Behav, vol.50, pp.736-747, 2006.

A. Romano-lópez, M. Méndez-díaz, F. G. García, C. Regalado-santiago, A. E. Ruiz-contreras et al., Maternal separation and early stress cause long-lasting effects on dopaminergic and endocannabinergic systems and alters dendritic morphology in the nucleus accumbens and frontal cortex in rats, Dev. Neurobiol, vol.76, pp.819-831, 2016.

M. G. Rooks and W. S. Garrett, Gut microbiota, metabolites and host immunity, Nat. Rev. Immunol, vol.16, pp.341-352, 2016.

A. Roque, A. Ochoa-zarzosa, and L. Torner, Maternal separation activates microglial cells and induces an inflammatory response in the hippocampus of male rat pups, independently of hypothalamic and peripheral cytokine levels, Brain. Behav. Immun, vol.55, pp.39-48, 2016.

S. Roque, A. R. Mesquita, J. A. Palha, N. Sousa, and M. Correia-neves, The Behavioral and Immunological Impact of Maternal Separation: A Matter of Timing, Front. Behav. Neurosci, vol.8, 2014.

C. S. Rosenfeld, Microbiome Disturbances and Autism Spectrum Disorders, Drug Metab. Dispos. Biol. Fate Chem, vol.43, pp.1557-1571, 2015.

P. Rosenfeld, J. B. Wetmore, and S. Levine, Effects of repeated maternal separations on the adrenocortical response to stress of preweanling rats, Physiol. Behav, vol.52, pp.787-791, 1992.

P. Rosenfeld, D. Suchecki, and S. Levine, Multifactorial regulation of the hypothalamic-pituitaryadrenal axis during development, Neurosci. Biobehav. Rev, vol.16, pp.553-568, 1992.

S. P. Rosshart, B. G. Vassallo, D. Angeletti, D. S. Hutchinson, A. P. Morgan et al., Wild Mouse Gut Microbiota Promotes Host Fitness and Improves Disease Resistance, Cell, 2017.

A. Rosztóczy, J. Fioramonti, K. Jármay, F. Barreau, T. Wittmann et al., Influence of sex and experimental protocol on the effect of maternal deprivation on rectal sensitivity to distension in the adult rat, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.15, pp.679-686, 2003.

T. L. Roth and J. D. Sweatt, Epigenetic marking of the BDNF gene by early-life adverse experiences, Horm. Behav, vol.59, pp.315-320, 2011.

T. L. Roth, F. D. Lubin, A. J. Funk, and J. D. Sweatt, Lasting Epigenetic Influence of Early-Life Adversity on the BDNF Gene, Biol. Psychiatry, vol.65, pp.760-769, 2009.

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

D. Rüedi-bettschen, E. Pedersen, J. Feldon, P. , and C. R. , Early deprivation under specific conditions leads to reduced interest in reward in adulthood in Wistar rats, Behav. Brain Res, vol.156, pp.297-310, 2005.

A. Ruggieri, S. Anticoli, A. D'ambrosio, L. Giordani, and M. Viora, The influence of sex and gender on immunity, infection and vaccination, Ann. Ist. Super. Sanita, vol.52, pp.198-204, 2016.

M. Rutter, How the environment affects mental health, Br. J. Psychiatry, vol.186, pp.4-6, 2005.

M. Sadeghi, M. Peeri, and M. Hosseini, Adolescent voluntary exercise attenuated hippocampal innate immunity responses and depressive-like behaviors following maternal separation stress in male rats, Physiol. Behav, vol.163, pp.177-183, 2016.

S. Sakaguchi, T. Yamaguchi, T. Nomura, and M. Ono, Regulatory T cells and immune tolerance, Cell, vol.133, pp.775-787, 2008.

A. Salonen, J. Nikkilä, J. Jalanka-tuovinen, O. Immonen, M. Rajili?-stojanovi? et al., Comparative analysis of fecal DNA extraction methods with phylogenetic microarray: effective recovery of bacterial and archaeal DNA using mechanical cell lysis, J. Microbiol. Methods, vol.81, pp.127-134, 2010.

T. R. Sampson and S. K. Mazmanian, Control of brain development, function, and behavior by the microbiome, Cell Host Microbe, vol.17, pp.565-576, 2015.

T. R. Sampson, J. W. Debelius, T. Thron, S. Janssen, G. G. Shastri et al., Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson's Disease, Cell, vol.167, pp.1469-1480, 2016.

M. M. Sánchez, C. O. Ladd, and P. M. Plotsky, Early adverse experience as a developmental risk factor for later psychopathology: evidence from rodent and primate models, Dev. Psychopathol, vol.13, pp.419-449, 2001.

R. H. Sandler, S. M. Finegold, E. R. Bolte, C. P. Buchanan, A. P. Maxwell et al., Short-term benefit from oral vancomycin treatment of regressive-onset autism, J. Child Neurol, vol.15, pp.429-435, 2000.

R. M. Sapolsky and M. J. Meaney, Maturation of the adrenocortical stress response: neuroendocrine control mechanisms and the stress hyporesponsive period, Brain Res, vol.396, pp.64-76, 1986.

A. Sarkar, S. M. Lehto, S. Harty, T. G. Dinan, J. F. Cryan et al., Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals, Trends Neurosci, vol.39, pp.763-781, 2016.

D. M. Sartor and A. J. Verberne, Abdominal vagal signalling: a novel role for cholecystokinin in circulatory control?, Brain Res. Rev, vol.59, pp.140-154, 2008.

V. Sasselli, V. Pachnis, and A. J. Burns, The enteric nervous system, Dev. Biol, vol.366, pp.64-73, 2012.

H. M. Savignac, B. Kiely, T. G. Dinan, and J. F. Cryan, Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.26, pp.1615-1627, 2014.

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. Behav. Immun, vol.52, pp.120-131, 2016.

M. L. Scattoni, J. Crawley, and L. Ricceri, Ultrasonic vocalizations: A tool for behavioural phenotyping of mouse models of neurodevelopmental disorders, Neurosci. Biobehav. Rev, vol.33, pp.508-515, 2009.

D. P. Schafer and B. Stevens, Phagocytic glial cells: sculpting synaptic circuits in the developing nervous system, Curr. Opin. Neurobiol, vol.23, pp.1034-1040, 2013.

M. Schmidt, L. Enthoven, J. H. Van-woezik, S. Levine, E. R. De-kloet et al., The dynamics of the hypothalamic-pituitary-adrenal axis during maternal deprivation, J. Neuroendocrinol, vol.16, pp.52-57, 2004.

M. V. Schmidt, M. Schmidt, L. Enthoven, M. Van-der-mark, S. Levine et al., The postnatal development of the hypothalamic-pituitary-adrenal axis in the mouse, Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci, vol.21, pp.125-132, 2003.

M. V. Schmidt, X. Wang, and O. C. Meijer, Early life stress paradigms in rodents: potential animal models of depression?, Psychopharmacology (Berl.), vol.214, pp.131-140, 2011.

F. A. Schroeder, C. L. Lin, W. E. Crusio, A. , and S. , Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse, Biol. Psychiatry, vol.62, pp.55-64, 2007.

J. M. Schwarz and S. D. Bilbo, Sex, glia, and development: interactions in health and disease, Horm. Behav, vol.62, pp.243-253, 2012.

I. Schwetz, J. A. Mcroberts, S. V. Coutinho, S. Bradesi, G. Gale et al., Corticotropin-releasing factor receptor 1 mediates acute and delayed stress-induced visceral hyperalgesia in maternally separated Long-Evans rats, Am. J. Physiol. Gastrointest. Liver Physiol, vol.289, pp.704-712, 2005.

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.

M. K. Seo, N. N. Ly, C. H. Lee, H. Y. Cho, C. M. Choi et al., Early life stress increases stress vulnerability through BDNF gene epigenetic changes in the rat hippocampus, Neuropharmacology, vol.105, pp.388-397, 2016.

A. P. Shah, F. R. Carreno, H. Wu, Y. A. Chung, and A. Frazer, Role of TrkB in the anxiolytic-like and antidepressant-like effects of vagal nerve stimulation: Comparison with desipramine, Neuroscience, vol.322, pp.273-286, 2016.

U. Shalev and N. Kafkafi, Repeated maternal separation does not alter sucrose-reinforced and openfield behaviors, Pharmacol. Biochem. Behav, vol.73, pp.115-122, 2002.

N. Shanks, S. Larocque, and M. J. Meaney, Neonatal endotoxin exposure alters the development of the hypothalamic-pituitary-adrenal axis: early illness and later responsivity to stress, J. Neurosci. Off. J. Soc. Neurosci, vol.15, pp.376-384, 1995.

K. A. Sharkey, Emerging roles for enteric glia in gastrointestinal disorders, J. Clin. Invest, vol.125, pp.918-925, 2015.

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.

L. Shen, E. D. Black, E. D. Witkowski, W. I. Lencer, V. Guerriero et al., Myosin light chain phosphorylation regulates barrier function by remodeling tight junction structure, J. Cell Sci, vol.119, pp.2095-2106, 2006.

B. A. Shenderov, Gut indigenous microbiota and epigenetics, Microb. Ecol. Health Dis, vol.23, 2012.

E. Sherwin, K. Rea, T. G. Dinan, and J. F. Cryan, A gut (microbiome) feeling about the brain, Curr. Opin. Gastroenterol, 2016.

S. Y. Shin, S. H. Han, R. Woo, S. H. Jang, M. et al., Adolescent mice show anxiety-and aggressive-like behavior and the reduction of long-term potentiation in mossy fiber-CA3 synapses after neonatal maternal separation, Neuroscience, vol.316, pp.221-231, 2016.

S. Yim, Y. Park, A. Berrios, J. Lafourcade, M. Pascual et al., Reversing behavioural abnormalities in mice exposed to maternal inflammation, Nature, vol.549, pp.482-487, 2017.

C. Shu, L. Xiao, J. Tang, G. Wang, X. Zhang et al., Blunted behavioral and molecular responses to chronic mild stress in adult rats with experience of infancy maternal separation, Tohoku J. Exp. Med, vol.235, pp.81-87, 2015.

D. M. Silberman, G. B. Acosta, Z. Zubilete, and M. A. , Long-term effects of early life stress exposure: Role of epigenetic mechanisms, Pharmacol. Res, 2016.

J. L. Silverman, M. Yang, C. Lord, and J. N. Crawley, Behavioural phenotyping assays for mouse models of autism, Nat. Rev. Neurosci, vol.11, pp.490-502, 2010.

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.

A. Slyepchenko, M. Maes, R. Machado-vieira, G. Anderson, M. Solmi et al., Intestinal Dysbiosis, Gut Hyperpermeability and Bacterial Translocation: Missing Links Between Depression, Obesity and Type 2 Diabetes, Curr. Pharm. Des, vol.22, pp.6087-6106, 2016.

R. F. Slykerman, F. Hood, K. Wickens, J. M. Thompson, C. Barthow et al., Effect of Lactobacillus rhamnosus HN001 in Pregnancy on Postpartum Symptoms of Depression and Anxiety: A Randomised Double-blind, vol.24, pp.159-165, 2017.

K. Smith, K. D. Mccoy, and A. J. Macpherson, Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota, Semin. Immunol, vol.19, pp.59-69, 2007.

J. Snel, P. P. Heinen, H. J. Blok, R. J. Carman, A. J. Duncan et al., Comparison of 16S rRNA sequences of segmented filamentous bacteria isolated from mice, rats, and chickens and proposal of "Candidatus Arthromitus, Int. J. Syst. Bacteriol, vol.45, pp.780-782, 1995.

J. S. Snyder, A. Soumier, M. Brewer, J. Pickel, C. et al., Adult hippocampal neurogenesis buffers stress responses and depressive behaviour, Nature, vol.476, pp.458-461, 2011.

J. D. Söderholm, D. A. Yates, M. G. Gareau, P. Yang, G. Macqueen et al., Neonatal maternal separation predisposes adult rats to colonic barrier dysfunction in response to mild stress, Am. J. Physiol.-Gastrointest. Liver Physiol, vol.283, pp.1257-1263, 2002.

C. Sommer, Serotonin in pain and analgesia: actions in the periphery, Mol. Neurobiol, vol.30, pp.117-125, 2004.

G. H. Son, D. Geum, S. Chung, E. J. Kim, J. Jo et al., Maternal stress produces learning deficits associated with impairment of NMDA receptor-mediated synaptic plasticity, J. Neurosci. Off. J. Soc. Neurosci, vol.26, pp.3309-3318, 2006.

V. C. Sousa, J. Vital, A. R. Costenla, V. L. Batalha, A. M. Sebastião et al., Maternal separation impairs long term-potentiation in CA1-CA3 synapses and hippocampal-dependent memory in old rats, Neurobiol. Aging, vol.35, pp.1680-1685, 2014.

E. Soztutar, E. Colak, and E. Ulupinar, Gender-and anxiety level-dependent effects of perinatal stress exposure on medial prefrontal cortex, Exp. Neurol. 275 Pt, vol.2, pp.274-284, 2016.

U. Stadlbauer, S. C. Woods, W. Langhans, and U. Meyer, PYY3-36: Beyond food intake, Front. Neuroendocrinol, vol.38, pp.1-11, 2015.

Z. Steel, C. Marnane, C. Iranpour, T. Chey, J. W. Jackson et al., The global prevalence of common mental disorders: a systematic review and meta-analysis 1980-2013, Int. J. Epidemiol, vol.43, pp.476-493, 2014.

L. Steenbergen, R. Sellaro, S. Van-hemert, J. A. Bosch, and L. S. Colzato, A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood, Brain. Behav. Immun, vol.48, pp.258-264, 2015.

M. B. Stein, J. R. Walker, G. Anderson, A. L. Hazen, C. A. Ross et al., Childhood physical and sexual abuse in patients with anxiety disorders and in a community sample, Am. J. Psychiatry, vol.153, pp.275-277, 1996.

A. Stengel and Y. Taché, Corticotropin-releasing factor signaling and visceral response to stress, Exp. Biol. Med. Maywood NJ, vol.235, pp.1168-1178, 2010.

B. R. Stevens, R. Goel, K. Seungbum, E. M. Richards, R. C. Holbert et al., Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression, 2017.

C. W. Stevenson, C. H. Spicer, R. Mason, and C. A. Marsden, Early life programming of fear conditioning and extinction in adult male rats, Behav. Brain Res, vol.205, pp.505-510, 2009.

R. M. Stilling, T. G. Dinan, and J. F. Cryan, Microbial genes, brain & behaviour -epigenetic regulation of the gut-brain axis, Genes Brain Behav, vol.13, pp.69-86, 2014.

R. M. Stilling, S. R. Bordenstein, T. G. Dinan, and J. F. Cryan, Friends with social benefits: hostmicrobe interactions as a driver of brain evolution and development? Front, Cell. Infect. Microbiol, vol.4, p.147, 2014.

R. M. Stilling, M. Van-de-wouw, G. Clarke, C. Stanton, T. G. Dinan et al., The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis?, Neurochem. Int, vol.99, pp.110-132, 2016.

R. H. Straub, R. Wiest, U. G. Strauch, P. Härle, and J. Schölmerich, The role of the sympathetic nervous system in intestinal inflammation, Gut, vol.55, pp.1640-1649, 2006.

L. Su, L. Shen, D. R. Clayburgh, S. C. Nalle, E. A. Sullivan et al., Targeted epithelial tight junction dysfunction causes immune activation and contributes to development of experimental colitis, Gastroenterology, vol.136, pp.551-563, 2009.

L. Su, L. Shen, D. R. Clayburgh, S. C. Nalle, E. A. Sullivan et al., Targeted epithelial tight junction dysfunction causes immune activation and contributes to development of experimental colitis, Gastroenterology, vol.136, pp.551-563, 2009.

D. Suchecki, P. Rosenfeld, and S. Levine, Maternal regulation of the hypothalamic-pituitary-adrenal axis in the infant rat: the roles of feeding and stroking, Brain Res. Dev. Brain Res, vol.75, pp.185-192, 1993.

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

Y. Sumi, M. Miyakawa, M. Kanzaki, and Y. Kotake, Vitamin B-6 deficiency in germfree rats, J. Nutr, vol.107, pp.1707-1714, 1977.

Y. Sung, M. Shin, S. Cho, H. Baik, B. Jin et al., Depression-like state in maternal rats induced by repeated separation of pups is accompanied by a decrease of cell proliferation and an increase of apoptosis in the hippocampus, Neurosci. Lett, vol.470, pp.86-90, 2010.

D. Suri, V. Veenit, A. Sarkar, D. Thiagarajan, A. Kumar et al., Early Stress Evokes Age-Dependent Biphasic Changes in Hippocampal Neurogenesis, Bdnf Expression, and Cognition, Biol. Psychiatry, vol.73, pp.658-666, 2013.

M. Surjit, K. P. Ganti, A. Mukherji, T. Ye, G. Hua et al., Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor, Cell, vol.145, pp.224-241, 2011.

T. Suzuki, S. Yoshida, and H. Hara, Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability, Br. J. Nutr, vol.100, pp.297-305, 2008.

Y. Taché and B. Bonaz, Corticotropin-releasing factor receptors and stress-related alterations of gut motor function, J. Clin. Invest, vol.117, pp.33-40, 2007.

Y. Taché and M. H. Perdue, Role of peripheral CRF signalling pathways in stress-related alterations of gut motility and mucosal function, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.16, issue.1, pp.137-142, 2004.

M. Takada, K. Nishida, A. Kataoka-kato, Y. Gondo, H. Ishikawa et al., Probiotic Lactobacillus casei strain Shirota relieves stress-associated symptoms by modulating the gut-brain interaction in human and animal models, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, 2016.

S. Talukdar, B. M. Owen, P. Song, G. Hernandez, Y. Zhang et al., FGF21 Regulates Sweet and Alcohol Preference, vol.23, pp.344-349, 2016.

A. T. Tang, J. P. Choi, J. J. Kotzin, Y. Yang, C. C. Hong et al., Endothelial TLR4 and the microbiome drive cerebral cavernous malformations, Nature, vol.545, pp.305-310, 2017.

H. Tang, G. Zhang, N. Ji, L. Du, B. Chen et al., Toll-Like Receptor 4 in Paraventricular Nucleus Mediates Visceral Hypersensitivity Induced by Maternal Separation, Front. Pharmacol, vol.8, p.309, 2017.

A. J. Tarr, J. D. Galley, S. E. Fisher, M. Chichlowski, B. M. Berg et al., The prebiotics 3'Sialyllactose and 6'Sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: Evidence for effects on the gut-brain axis, Brain. Behav. Immun, vol.50, pp.166-177, 2015.

N. Tennoune, R. Legrand, W. Ouelaa, J. Breton, N. Lucas et al., Sex-related effects of nutritional supplementation of Escherichia coli: relevance to eating disorders, Nutr. Burbank Los Angel. Cty. Calif, vol.31, pp.498-507, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01960157

M. S. Thion and S. Garel, On place and time: microglia in embryonic and perinatal brain development, Curr. Opin. Neurobiol, vol.47, pp.121-130, 2017.

A. W. Thomas, N. Caporale, C. Wu, and L. Wilbrecht, Early maternal separation impacts cognitive flexibility at the age of first independence in mice, Dev. Cogn. Neurosci, vol.18, pp.49-56, 2016.

C. L. Thompson, R. Vier, A. Mikaelyan, T. Wienemann, and A. Brune, Candidatus Arthromitus" revised: segmented filamentous bacteria in arthropod guts are members of Lachnospiraceae, Environ. Microbiol, vol.14, pp.1454-1465, 2012.

K. Tillisch, E. A. Mayer, and J. S. Labus, Quantitative Meta-analysis Identifies Brain Regions Activated During Rectal Distension in Irritable Bowel Syndrome, Gastroenterology, vol.140, pp.91-100, 2011.

K. Tillisch, J. Labus, L. Kilpatrick, Z. Jiang, J. Stains et al., Consumption of fermented milk product with probiotic modulates brain activity, Gastroenterology, vol.144, pp.1-4, 1401.

Y. Tjong, S. Ip, L. Lao, J. Wu, H. H. Fong et al., Role of neuronal nitric oxide synthase in colonic distension-induced hyperalgesia in distal colon of neonatal maternal separated male rats, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.23, pp.666-278, 2011.

H. Toda, S. Boku, S. Nakagawa, T. Inoue, A. Kato et al., Maternal separation enhances conditioned fear and decreases the mRNA levels of the neurotensin receptor 1 gene with hypermethylation of this gene in the rat amygdala, PloS One, vol.9, p.97421, 2014.

K. Tominaga, Y. Fujikawa, F. Tanaka, N. Kamata, H. Yamagami et al., Structural changes in gastric glial cells and delayed gastric emptying as responses to early life stress and acute adulthood stress in rats, Life Sci, vol.148, pp.254-259, 2016.

S. G. Tractenberg, M. L. Levandowski, L. A. De-azeredo, R. Orso, L. G. Roithmann et al., An overview of maternal separation effects on behavioural outcomes in mice: Evidence from a four-stage methodological systematic review, Neurosci. Biobehav. Rev, vol.68, pp.489-503, 2016.

C. Troakes and C. D. Ingram, Anxiety behaviour of the male rat on the elevated plus maze: associated regional increase in c-fos mRNA expression and modulation by early maternal separation, Stress Int. J. Biol. Stress, vol.12, pp.362-369, 2009.

S. W. Tsang, M. Zhao, J. Wu, J. J. Sung, and Z. Bian, Nerve growth factor-mediated neuronal plasticity in spinal cord contributes to neonatal maternal separation-induced visceral hypersensitivity in rats: Nerve growth factor-mediated neuronal plasticity in spinal cord contributes to hypersensitivity in rats, Eur. J. Pain, vol.16, pp.463-472, 2012.

N. M. Tsankova, O. Berton, W. Renthal, A. Kumar, R. L. Neve et al., Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action, Nat. Neurosci, vol.9, pp.519-525, 2006.

M. C. Tsuda, N. Yamaguchi, and S. Ogawa, Early life stress disrupts peripubertal development of aggression in male mice, Neuroreport, vol.22, pp.259-263, 2011.

M. C. Tsuda, N. Yamaguchi, M. Nakata, and S. Ogawa, Modification of female and male social behaviors in estrogen receptor beta knockout mice by neonatal maternal separation, Front. Neurosci, vol.8, p.274, 2014.

P. J. Turnbaugh, F. Bäckhed, L. Fulton, G. , and J. I. , Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome, Cell Host Microbe, vol.3, pp.213-223, 2008.

S. Uchida, K. Hara, A. Kobayashi, H. Funato, T. Hobara et al., Early Life Stress Enhances Behavioral Vulnerability to Stress through the Activation of REST4-Mediated Gene Transcription in the Medial Prefrontal Cortex of Rodents, J. Neurosci, vol.30, pp.15007-15018, 2010.

Y. M. Ulrich-lai and J. P. Herman, Neural regulation of endocrine and autonomic stress responses, Nat. Rev. Neurosci, vol.10, pp.397-409, 2009.

M. Vallée, W. Mayo, S. Maccari, M. Le-moal, and H. Simon, Long-term effects of prenatal stress and handling on metabolic parameters: relationship to corticosterone secretion response, Brain Res, vol.712, pp.287-292, 1996.

M. Vallée, W. Mayo, F. Dellu, M. Le-moal, H. Simon et al., Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion, J. Neurosci. Off. J. Soc. Neurosci, vol.17, pp.2626-2636, 1997.

M. Vallée, S. Maccari, F. Dellu, H. Simon, M. Le-moal et al., Long-term effects of prenatal stress and postnatal handling on age-related glucocorticoid secretion and cognitive performance: a longitudinal study in the rat, Eur. J. Neurosci, vol.11, pp.2906-2916, 1999.

J. H. Van-bilsen, E. Sienkiewicz-sz?apka, D. Lozano-ojalvo, L. E. Willemsen, C. M. Antunes et al., Application of the adverse outcome pathway (AOP) concept to structure the available in vivo and in vitro mechanistic data for allergic sensitization to food proteins, Clin. Transl. Allergy, vol.7, p.13, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01604242

P. Van-den-abbeele, C. Belzer, M. Goossens, M. Kleerebezem, W. M. De-vos et al., Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model, ISME J, vol.7, pp.949-961, 2013.

R. M. Van-den-wijngaard, O. I. Stanisor, S. A. Van-diest, O. Welting, M. M. Wouters et al., Susceptibility to stress induced visceral hypersensitivity in maternally separated rats is transferred across generations, Neurogastroenterol. Motil, vol.25, pp.780-790, 2013.

,. Van-harmelen, M. Van-tol, N. J. Van-der-wee, D. J. Veltman, A. Aleman et al., Reduced Medial Prefrontal Cortex Volume in Adults Reporting Childhood Emotional Maltreatment, Biol. Psychiatry, vol.68, pp.832-838, 2010.

H. J. Van-oers, E. R. De-kloet, T. Whelan, and S. Levine, Maternal deprivation effect on the infant's neural stress markers is reversed by tactile stimulation and feeding but not by suppressing corticosterone, J. Neurosci, vol.18, pp.10171-10179, 1998.

T. Vanuytsel, S. Van-wanrooy, H. Vanheel, C. Vanormelingen, S. Verschueren et al., Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism, Gut, vol.63, pp.1293-1299, 2014.

A. K. Varghese, E. F. Verdú, P. Bercik, W. I. Khan, P. A. Blennerhassett et al., Antidepressants attenuate increased susceptibility to colitis in a murine model of depression, Gastroenterology, vol.130, pp.1743-1753, 2006.

D. M. Vázquez, Stress and the developing limbic-hypothalamic-pituitary-adrenal axis, Psychoneuroendocrinology, vol.23, pp.663-700, 1998.

E. Vázquez, A. Barranco, M. Ramírez, A. Gruart, J. M. Delgado-garcía et al., Effects of a human milk oligosaccharide, 2'-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents, J. Nutr. Biochem, vol.26, pp.455-465, 2015.

V. Vazquez, J. Penit-soria, C. Durand, M. J. Besson, B. Giros et al., Maternal deprivation increases vulnerability to morphine dependence and disturbs the enkephalinergic system in adulthood, J. Neurosci. Off. J. Soc. Neurosci, vol.25, pp.4453-4462, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00085367

V. Vazquez, B. Giros, and V. Daugé, Maternal deprivation specifically enhances vulnerability to opiate dependence, Behav. Pharmacol, vol.17, pp.715-724, 2006.

A. H. Veenema, Toward understanding how early-life social experiences alter oxytocin-and vasopressinregulated social behaviors, Horm. Behav, vol.61, pp.304-312, 2012.

A. H. Veenema, S. O. Reber, S. Selch, F. Obermeier, and I. D. Neumann, Early life stress enhances the vulnerability to chronic psychosocial stress and experimental colitis in adult mice, Endocrinology, vol.149, pp.2727-2736, 2008.

J. Vetulani, Early maternal separation: a rodent model of depression and a prevailing human condition, Pharmacol. Rep, vol.65, pp.1451-1461, 2013.

M. P. Viveros, R. Llorente, M. López-gallardo, J. Suarez, F. Bermúdez-silva et al., Sex-dependent alterations in response to maternal deprivation in rats, Psychoneuroendocrinology, vol.34, issue.1, pp.217-226, 2009.

J. V. Vlaini?, J. ?uran, T. Vlaini?, and A. L. Vukorep, Probiotics as an Adjuvant Therapy in Major Depressive Disorder, Curr. Neuropharmacol, vol.14, pp.952-958, 2016.

N. Voreades, A. Kozil, and T. L. Weir, Diet and the development of the human intestinal microbiome, Front. Microbiol, vol.5, p.494, 2014.

M. Vythilingam, C. Heim, J. Newport, A. H. Miller, E. Anderson et al., Childhood trauma associated with smaller hippocampal volume in women with major depression, Am. J. Psychiatry, vol.159, pp.2072-2080, 2002.

B. Wang and J. Brand-miller, The role and potential of sialic acid in human nutrition, Eur. J. Clin. Nutr, vol.57, pp.1351-1369, 2003.

Y. Wang and L. H. Kasper, The role of microbiome in central nervous system disorders, Brain. Behav. Immun, vol.38, pp.1-12, 2014.

A. Wang, W. Nie, H. Li, Y. Hou, Z. Yu et al., Epigenetic Upregulation of Corticotrophin-Releasing Hormone Mediates Postnatal Maternal Separation-Induced Memory Deficiency, PLoS ONE, vol.9, p.94394, 2014.

B. Wang, P. Mcveagh, P. Petocz, and J. Brand-miller, Brain ganglioside and glycoprotein sialic acid in breastfed compared with formula-fed infants, Am. J. Clin. Nutr, vol.78, pp.1024-1029, 2003.

E. M. Wang, W. T. Li, X. J. Yan, X. Chen, Q. Liu et al., Vagal afferent-dependent cholecystokinin modulation of visceral pain requires central amygdala NMDA-NR2B receptors in rats, Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc, vol.27, pp.1333-1343, 2015.

H. Wang, P. Wang, X. Wang, Y. Wan, and Y. Liu, Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein Claudin-1 transcription, Dig. Dis. Sci, vol.57, pp.3126-3135, 2012.

L. Wang, J. Jiao, and S. C. Dulawa, Infant maternal separation impairs adult cognitive performance in BALB/cJ mice, Psychopharmacology (Berl.), vol.216, pp.207-218, 2011.

Z. Wang, C. Friedrich, S. C. Hagemann, W. H. Korte, N. Goharani et al., Regulatory T cells promote a protective Th17-associated immune response to intestinal bacterial infection with C. rodentium, Mucosal Immunol, vol.7, pp.1290-1301, 2014.

Z. Wang, D. Koonen, M. Hofker, and J. Fu, Gut microbiome and lipid metabolism: from associations to mechanisms, Curr. Opin. Lipidol, vol.27, pp.216-224, 2016.

I. C. Weaver, M. Szyf, and M. J. Meaney, From maternal care to gene expression: DNA methylation and the maternal programming of stress responses, Endocr. Res, vol.28, p.699, 2002.

I. C. Weaver, N. Cervoni, F. A. Champagne, A. C. D'alessio, S. Sharma et al., Epigenetic programming by maternal behavior, Nat. Neurosci, vol.7, pp.847-854, 2004.

S. Weich, J. Patterson, R. Shaw, and S. Stewart-brown, Family relationships in childhood and common psychiatric disorders in later life: systematic review of prospective studies, Br. J. Psychiatry, vol.194, pp.392-398, 2009.

M. Weinstock, Gender differences in the effects of prenatal stress on brain development and behaviour, Neurochem. Res, vol.32, pp.1730-1740, 2007.

I. C. Weiss, T. B. Franklin, S. Vizi, and I. M. Mansuy, Inheritable Effect of Unpredictable Maternal Separation on Behavioral Responses in Mice, Front. Behav. Neurosci, vol.5, 2011.

K. A. Wellmann, E. I. Varlinskaya, and S. M. Mooney, D-Cycloserine ameliorates social alterations that result from prenatal exposure to valproic acid, Brain Res. Bull, vol.108, pp.1-9, 2014.

D. M. Werling and D. H. Geschwind, Sex differences in autism spectrum disorders, Curr. Opin. Neurol, vol.26, pp.146-153, 2013.

R. Wheeler, G. Chevalier, G. Eberl, G. Boneca, and I. , The biology of bacterial peptidoglycans and their impact on host immunity and physiology, Cell. Microbiol, vol.16, pp.1014-1023, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01402716

A. E. Whimbey and V. E. Denenberg, Programming Life Histories : Creating Individual Differences By The Experimental Control Of Early Experiences, Multivar. Behav. Res, vol.1, pp.279-286, 1966.

A. Wieck, S. L. Andersen, and H. C. Brenhouse, Evidence for a neuroinflammatory mechanism in delayed effects of early life adversity in rats: Relationship to cortical NMDA receptor expression, Brain. Behav. Immun, vol.28, pp.218-226, 2013.

A. A. Wilber, C. J. Southwood, and C. L. Wellman, Brief neonatal maternal separation alters extinction of conditioned fear and corticolimbic glucocorticoid and NMDA receptor expression in adult rats, Dev. Neurobiol, vol.69, pp.73-87, 2009.

P. Willner, A. Towell, D. Sampson, S. Sophokleous, and R. Muscat, Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant, Psychopharmacology (Berl.), vol.93, pp.358-364, 1987.

J. H. Winston and S. K. Sarna, Enhanced sympathetic nerve activity induced by neonatal colon inflammation induces gastric hypersensitivity and anxiety-like behavior in adult rats, Am. J. Physiol. Gastrointest. Liver Physiol, vol.311, pp.32-39, 2016.

F. L. Woon and D. W. Hedges, Hippocampal and amygdala volumes in children and adults with childhood maltreatment-related posttraumatic stress disorder: a meta-analysis, Hippocampus, vol.18, pp.729-736, 2008.

B. S. Wostmann, C. Larkin, A. Moriarty, and E. Bruckner-kardoss, Dietary intake, energy metabolism, and excretory losses of adult male germfree Wistar rats, Lab. Anim. Sci, vol.33, pp.46-50, 1983.

R. L. Wu, M. I. Vazquez-roque, P. Carlson, D. Burton, M. Grover et al., , 2017.

, Gluten-induced symptoms in diarrhea-predominant irritable bowel syndrome are associated with increased myosin light chain kinase activity and claudin-15 expression, Lab. Investig. J. Tech. Methods Pathol, vol.97, pp.14-23

L. Yang, T. Xu, K. Zhang, Z. Wei, X. Li et al., The essential role of hippocampal alpha6 subunit-containing GABAA receptors in maternal separation stress-induced adolescent depressive behaviors, Behav. Brain Res, 2016.

Y. Yang, Z. Cheng, H. Tang, H. Jiao, X. Sun et al., Neonatal Maternal Separation Impairs Prefrontal Cortical Myelination and Cognitive Functions in Rats Through Activation of Wnt Signaling, 1991.

C. Yen, C. Wang, W. Wu, C. , and H. , Fructo-oligosaccharide improved brain ?amyloid, ?-secretase, cognitive function, and plasma antioxidant levels in D-galactose-treated Balb/cJ mice, Nutr. Neurosci, vol.20, pp.228-237, 2017.

L. Yi, H. Zhang, H. Sun, L. Zhou, Y. Chen et al., Maternal Separation Induced Visceral Hypersensitivity from Childhood to Adulthood, J. Neurogastroenterol. Motil, vol.23, pp.306-315, 2017.

Y. Yin, Y. Wang, L. Zhu, W. Liu, N. Liao et al., Comparative analysis of the distribution of segmented filamentous bacteria in humans, mice and chickens, ISME J, vol.7, pp.615-621, 2013.

J. M. York, N. A. Blevins, T. Baynard, and G. G. Freund, Mouse testing methods in psychoneuroimmunology: an overview of how to measure sickness, depressive/anxietal, cognitive, and physical activity behaviors, Methods Mol. Biol. Clifton NJ, vol.934, pp.243-276, 2012.

E. A. Young, J. L. Abelson, G. C. Curtis, and R. M. Nesse, Childhood adversity and vulnerability to mood and anxiety disorders, Depress. Anxiety, vol.5, pp.66-72, 1997.

Z. Zakostelska, M. Kverka, K. Klimesova, P. Rossmann, J. Mrazek et al., Lysate of probiotic Lactobacillus casei DN-114 001 ameliorates colitis by strengthening the gut barrier function and changing the gut microenvironment, PloS One, vol.6, p.27961, 2011.

L. Zhang, V. S. Hernández, B. Liu, M. P. Medina, A. T. Nava-kopp et al., Hypothalamic vasopressin system regulation by maternal separation: Its impact on anxiety in rats, Neuroscience, vol.215, pp.135-148, 2012.

L. Zhang, S. Levine, G. Dent, Y. Zhan, G. Xing et al., Maternal deprivation increases cell death in the infant rat brain, Dev. Brain Res, vol.133, pp.1-11, 2002.

L. Zhao, The gut microbiota and obesity: from correlation to causality, Nat. Rev. Microbiol, vol.11, pp.639-647, 2013.

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, Mol. Psychiatry, vol.21, pp.786-796, 2016.

X. Zhou, M. Li, X. Li, X. Long, X. Zuo et al., Visceral hypersensitive rats share common dysbiosis features with irritable bowel syndrome patients, World J. Gastroenterol, vol.22, pp.5211-5227, 2016.

F. Zhu, Y. Liu, J. Zhao, and Y. Zheng, Minocycline alleviates behavioral deficits and inhibits microglial activation induced by intrahippocampal administration of Granulocyte-Macrophage Colony-Stimulating Factor in adult rats, Neuroscience, vol.266, pp.275-281, 2014.

F. Zhu, Y. Zheng, Y. Ding, Y. Liu, X. Zhang et al., Minocycline and risperidone prevent microglia activation and rescue behavioral deficits induced by neonatal intrahippocampal injection of lipopolysaccharide in rats, PloS One, vol.9, p.93966, 2014.

Y. Zhu, Y. Wang, R. Yao, T. Hao, J. Cao et al., Enhanced neuroinflammation mediated by DNA methylation of the glucocorticoid receptor triggers cognitive dysfunction after sevoflurane anesthesia in adult rats subjected to maternal separation during the neonatal period, J. Neuroinflammation, vol.14, p.6, 2017.

I. Zilber-rosenberg and E. Rosenberg, Role of microorganisms in the evolution of animals and plants: the hologenome theory of evolution, FEMS Microbiol. Rev, vol.32, pp.723-735, 2008.

B. Zimmerberg and E. W. Kajunski, Sexually dimorphic effects of postnatal allopregnanolone on the development of anxiety behavior after early deprivation, Pharmacol. Biochem. Behav, vol.78, pp.465-471, 2004.

B. Zimmerberg and K. A. Sageser, Comparison of Two Rodent Models of Maternal Separation on, Juvenile Social Behavior. Front. Psychiatry, vol.2, 2011.

B. Zimmerberg, S. A. Brunelli, A. J. Fluty, and C. A. Frye, Differences in affective behaviors and hippocampal allopregnanolone levels in adult rats of lines selectively bred for infantile vocalizations, Behav. Brain Res, vol.159, pp.301-311, 2005.

Y. Zolotarevsky, G. Hecht, A. Koutsouris, D. E. Gonzalez, C. Quan et al., A membrane-permeant peptide that inhibits MLC kinase restores barrier function in in vitro models of intestinal disease, Gastroenterology, vol.123, pp.163-172, 2002.

S. Zonis, R. N. Pechnick, V. A. Ljubimov, M. Mahgerefteh, K. Wawrowsky et al., Chronic intestinal inflammation alters hippocampal neurogenesis, J. Neuroinflammation, vol.12, p.65, 2015.

, Here using an animal model in rats, we explored the combinatorial effects of a maternal HFD (40% of energy from fat without impact on maternal weight; during gestation and lactation) and maternal separation (MS) in offspring. In the prefrontal cortex (PFC) of pups, MS led to changes in the expression of several genes such as Bdnf (brain derived neurotrophic factor), 5HT-r1a (serotonin receptor 1a) and Rest4 (neuron-restrictive silencer element, repressor element 1, silencing transcription factor (Rest), splicing variant 4). Surprisingly, perinatal HFD strongly attenuated the developmental alterations induced by MS. Furthermore, maternal HFD totally prevented the endophenotypes (anxiety, spatial memory, social behavior, hypothalamic-pituitary-adrenal (HPA) axis response to stress, hippocampal neurogenesis and visceral pain) associated with MS at adulthood. Finally, we also demonstrated that HFD intake reduced anxiety and enhanced maternal care in stressed dams. Overall, our data suggest that a HFD restricted to gestation and lactation, which did not lead to overweight in dams, had limited effects in unstressed offspring, highlighting the role of maternal obesity, rather than fat exposure per se, 2016.

M. Rutter, How the environment affects mental health, Br J Psychiatry, vol.186, pp.4-6, 2005.

S. Weich, J. Patterson, R. Shaw, and S. Stewart-brown, Family relationships in childhood and common psychiatric disorders in later life: systematic review of prospective studies, Br J Psychiatry, vol.194, pp.392-398, 2009.

D. A. Hackman, M. J. Farah, and M. J. Meaney, Socioeconomic status and the brain: mechanistic insights from human and animal research, Nat Rev Neurosci, vol.11, pp.651-659, 2010.

K. A. Mclaughlin, L. D. Kubzansky, E. C. Dunn, R. Waldinger, G. Vaillant et al., Childhood social environment, emotional reactivity to stress, and mood and anxiety disorders across the life course, Depress Anxiety, vol.27, pp.1087-1094, 2010.

S. J. Reiser, K. A. Mcmillan, K. D. Wright, and G. Asmundson, Adverse childhood experiences and health anxiety in adulthood, Child Abuse Negl, vol.38, pp.407-413, 2014.

C. B. Nemeroff, Paradise Lost: The Neurobiological and Clinical Consequences of Child Abuse and Neglect, Neuron, vol.89, pp.892-909, 2016.

H. M. Rivera, K. J. Christiansen, and E. L. Sullivan, The role of maternal obesity in the risk of neuropsychiatric disorders, Front Neurosci, vol.9, p.194, 2015.

F. Cirulli, A. Berry, and E. Alleva, Early disruption of the mother-infant relationship: effects on brain plasticity and implications for psychopathology, Neurosci Biobehav Rev, vol.27, pp.73-82, 2003.

T. B. Franklin, B. J. Saab, and I. M. Mansuy, Neural mechanisms of stress resilience and vulnerability, Neuron, vol.75, pp.747-761, 2012.

M. M. Sánchez, C. O. Ladd, and P. M. Plotsky, Early adverse experience as a developmental risk factor for later psychopathology: evidence from rodent and primate models, Dev Psychopathol, vol.13, pp.419-449, 2001.

J. Maniam and M. J. Morris, Voluntary exercise and palatable high-fat diet both improve behavioural profile and stress responses in male rats exposed to early life stress: Role of hippocampus, Psychoneuroendocrinology, vol.35, pp.1553-1564, 2010.

T. A. Kosten, J. J. Kim, and H. J. Lee, Early life manipulations alter learning and memory in rats, Neurosci Biobehav Rev, vol.36, pp.1985-2006, 2012.

D. D. Francis, J. Diorio, P. M. Plotsky, and M. J. Meaney, Environmental enrichment reverses the effects of maternal separation on stress reactivity, J Neurosci, vol.22, pp.7840-7843, 2002.

F. Barreau, L. Ferrier, J. Fioramonti, and L. Bueno, New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models, Pediatr Res, vol.62, pp.240-245, 2007.

B. Aisa, R. Tordera, B. Lasheras, D. Río, J. Ramírez et al., Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats, Psychoneuroendocrinology, vol.32, pp.256-266, 2007.

M. Z. Alfaradhi and S. E. Ozanne, Developmental programming in response to maternal overnutrition, Front Genet, vol.2, p.27, 2011.

M. Li, D. M. Sloboda, and M. H. Vickers, Maternal obesity and developmental programming of metabolic disorders in offspring: evidence from animal models, Exp Diabetes Res, vol.2011, pp.1-9, 2011.

E. L. Sullivan, E. K. Nousen, K. A. Chamlou, and K. L. Grove, The impact of maternal high-fat diet consumption on neural development and behavior of offspring, Int J Obes Suppl, vol.2, pp.7-13, 2012.

Y. Tozuka, E. Wada, and K. Wada, Diet-induced obesity in female mice leads to peroxidized lipid accumulations and impairment of hippocampal neurogenesis during the early life of their offspring, FASEB J, vol.23, pp.1920-1934, 2009.

Y. Tozuka, M. Kumon, E. Wada, M. Onodera, H. Mochizuki et al., Maternal obesity impairs hippocampal BDNF production and spatial learning performance in young mouse offspring, Neurochem Int, vol.57, pp.235-247, 2010.

S. D. Bilbo and V. Tsang, Enduring consequences of maternal obesity for brain inflammation and behavior of offspring, FASEB J, vol.24, pp.2104-2115, 2010.

D. Peleg-raibstein, E. Luca, and C. Wolfrum, Maternal high-fat diet in mice programs emotional behavior in adulthood, Behav Brain Res, vol.233, pp.398-404, 2012.

A. Sasaki, W. C. De-vega, S. St-cyr, P. Pan, and P. O. Mcgowan, Perinatal high fat diet alters glucocorticoid signaling and anxiety behavior in adulthood, Neuroscience, vol.240, pp.1-12, 2013.

K. A. Mclaughlin, M. A. Sheridan, W. Winter, N. A. Fox, C. H. Zeanah et al., Widespread reductions in cortical thickness following severe early-life deprivation: A neurodevelopmental pathway to ADHD, Biol Psychiatry, vol.76, pp.629-638, 2014.

A. S. Hodel, R. H. Hunt, R. A. Cowell, S. E. Van-den-heuvel, M. R. Gunnar et al., Duration of early adversity and structural brain development in postinstitutionalized adolescents, NeuroImage, vol.105, pp.112-119, 2015.

N. M. Goodfellow, M. Benekareddy, V. A. Vaidya, and E. K. Lambe, Layer II/III of the prefrontal cortex: inhibition by the serotonin 5-ht1a receptor in development and stress, J Neurosci, vol.29, pp.10094-10103, 2009.

S. Uchida, K. Hara, A. Kobayashi, H. Funato, T. Hobara et al., Early life stress enhances behavioral vulnerability to stress through the activation of REST4-mediated gene transcription in the medial prefrontal cortex of rodents, J Neurosci, vol.30, pp.15007-15018, 2010.

A. L. Lépinay, T. Larrieu, C. Joffre, N. Acar, I. Gárate et al., Perinatal high-fat diet increases hippocampal vulnerability to the adverse effects of subsequent high-fat feeding, Psychoneuroendocrinology, vol.53, pp.82-93, 2015.

N. Moussaoui, V. Braniste, A. Ait-belgnaoui, M. Gabanou, S. Sekkal et al., Changes in intestinal glucocorticoid sensitivity in early life shape the risk of epithelial barrier defect in maternal-deprived rats, PLoS ONE, vol.9, p.88382, 2014.

R. H. Chapman and J. M. Stern, Failure of severe maternal stress or ACTH during pregnancy to affect emotionality of male rat offspring: implications of litter effects for prenatal studies, Dev Psychobiol, vol.12, pp.255-267, 1979.

H. Louvart, S. Maccari, F. Ducrocq, P. Thomas, and M. Darnaudéry, Long-term behavioural alterations in female rats after a single intense footshock followed by situational reminders, Psychoneuroendocrinology, vol.30, pp.316-324, 2005.

S. Agostini, M. Goubern, V. Tondereau, C. Salvador-cartier, V. Bezirard et al., A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in rats, Neurogastroenterol Motil, vol.24, pp.376-172, 2012.

M. M. Myers, S. A. Brunelli, J. M. Squire, R. D. Shindeldecker, and M. A. Hofer, Maternal behavior of SHR rats and its relationship to offspring blood pressures, Dev Psychobiol, vol.22, pp.29-53, 1989.

, Translational Psychiatry, pp.1-9, 2016.

V. F. Labrousse, A. Nadjar, C. Joffre, L. Costes, A. A. Grégoire et al., Short-term long chain omega3 diet protects from neuroinflammatory processes and memory impairment in aged mice, PLoS ONE, vol.7, p.36861, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01191175

A. M. Minni, R. Dorey, C. Piérard, G. Dominguez, J. Helbling et al., Critical role of plasma corticosteroid-binding-globulin during stress to promote glucocorticoid delivery to the brain: impact on memory retrieval, Endocrinology, vol.153, pp.4766-4774, 2012.

G. Paxinos, C. Watson, M. Pennisi, A. Topple, and . Bregma, lambda and the interaural midpoint in stereotaxic surgery with rats of different sex, strain and weight, J Neurosci Methods, vol.13, pp.139-143, 1985.

V. F. Labrousse, L. Costes, A. A. Darnaudéry, M. Ferreira, G. Amédée et al., Impaired interleukin-1beta and c-Fos expression in the hippocampus is associated with a spatial memory deficit in P2X(7) receptor-deficient mice, PLoS ONE, vol.4, p.6006, 2009.

V. Lemaire, S. Lamarque, L. Moal, M. Piazza, P. Abrous et al., Postnatal stimulation of the pups counteracts prenatal stress-induced deficits in hippocampal neurogenesis, Biol Psychiatry, vol.59, pp.786-792, 2006.

J. S. De-andrade, R. O. Abrão, I. C. Céspedes, M. C. Garcia, J. Nascimento et al., Acute restraint differently alters defensive responses and fos immunoreactivity in the rat brain, Behav Brain Res, vol.232, pp.20-29, 2012.

R. L. Huot, M. E. Gonzalez, C. O. Ladd, K. V. Thrivikraman, and P. M. Plotsky, Foster litters prevent hypothalamic-pituitary-adrenal axis sensitization mediated by neonatal maternal separation, Psychoneuroendocrinology, vol.29, pp.279-289, 2004.

G. Fond, A. Loundou, N. Hamdani, W. Boukouaci, A. Dargel et al., Anxiety and depression comorbidities in irritable bowel syndrome (IBS): a systematic review and meta-analysis, Eur Arch Psychiatry Clin Neurosci, vol.264, pp.651-660, 2014.

M. Camilleri, B. Coulie, and J. F. Tack, Visceral hypersensitivity: facts, speculations, and challenges, Gut, vol.48, pp.125-131, 2001.

M. L. Boccia, M. Razzoli, P. Vadlamudi, S. Trumbull, W. Caleffie et al., Repeated long separations from pups produce depression-like behavior in rat mothers, Psychoneuroendocrinology, vol.32, pp.65-71, 2007.

D. Liu, Maternal care, hippocampal glucocorticoid receptors, and hypothalamicpituitary-adrenal responses to stress, Science, vol.277, pp.1659-1662, 1997.

R. H. Purcell, B. Sun, L. L. Pass, M. L. Power, T. H. Moran et al., Maternal stress and high-fat diet effect on maternal behavior, milk composition, and pup ingestive behavior, Physiol Behav, vol.104, pp.474-479, 2011.

V. Bellisario, P. Panetta, G. Balsevich, V. Baumann, J. Noble et al., High-fat diet during pregnancy acts as a stressor increasing maternal glucocorticoids' signaling to the fetus and disrupting maternal behavior in a mouse model, Psychoneuroendocrinology, vol.61, p.10, 2015.

M. D. Niculescu and D. S. Lupu, High fat diet-induced maternal obesity alters fetal hippocampal development, Int J Dev Neurosci, vol.27, pp.627-633, 2009.

P. Gaspar, O. Cases, and L. Maroteaux, The developmental role of serotonin: news from mouse molecular genetics, Nat Rev Neurosci, vol.4, pp.1002-1012, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01274960

S. Cohen-cory, A. H. Kidane, N. J. Shirkey, and S. Marshak, Brain-derived neurotrophic factor and the development of structural neuronal connectivity, Dev Neurobiol, vol.70, pp.271-288, 2010.

S. Migliarini, G. Pacini, B. Pelosi, G. Lunardi, and M. Pasqualetti, Lack of brain serotonin affects postnatal development and serotonergic neuronal circuitry formation, Mol Psychiatry, vol.18, pp.1106-1118, 2013.

J. R. Homberg, R. Molteni, F. Calabrese, and M. A. Riva, The serotonin-BDNF duo: developmental implications for the vulnerability to psychopathology, Neurosci Biobehav Rev, vol.43, pp.35-47, 2014.

T. J. Rebello, Q. Yu, N. M. Goodfellow, C. Cagliostro, M. K. Teissier et al., Postnatal day 2 to 11 constitutes a 5-HT-sensitive period impacting adult mPFC function, J Neurosci, vol.34, pp.12379-12393, 2014.

C. Gross, X. Zhuang, K. Stark, S. Ramboz, R. Oosting et al., Serotonin1A receptor acts during development to establish normal anxiety-like behaviour in the adult, Nature, vol.416, pp.396-400, 2002.

E. S. Brown, A. J. Rush, and B. S. Mcewen, Hippocampal remodeling and damage by corticosteroids: implications for mood disorders, Neuropsychopharmacology, vol.21, pp.474-484, 1999.

M. A. Kheirbek, K. C. Klemenhagen, A. Sahay, and R. Hen, Neurogenesis and generalization: a new approach to stratify and treat anxiety disorders, Nat Neurosci, vol.15, pp.1613-1620, 2012.

J. Maniam and M. J. Morris, Palatable cafeteria diet ameliorates anxiety and depressionlike symptoms following an adverse early environment, Psychoneuroendocrinology, vol.35, pp.717-728, 2010.

D. M. Arcego, R. Krolow, C. Lampert, A. P. Toniazzo, C. Berlitz et al., Early life adversities or high fat diet intake reduce cognitive function and alter BDNF signaling in adult rats: Interplay of these factors changes these effects, Int J Dev Neurosci, vol.50, pp.16-25, 2016.

J. Maniam, C. P. Antoniadis, V. Le, and M. J. Morris, A diet high in fat and sugar reverses anxiety-like behaviour induced by limited nesting in male rats: Impacts on hippocampal markers, Psychoneuroendocrinology, vol.68, pp.202-209, 2016.

H. J. Van-oers, E. R. De-kloet, T. Whelan, and S. Levine, Maternal deprivation effect on the infant's neural stress markers is reversed by tactile stimulation and feeding but not by suppressing corticosterone, J Neurosci, vol.18, pp.10171-10179, 1998.

M. J. Meaney, Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations, Annu Rev Neurosci, vol.24, pp.1161-1192, 2001.

G. Oliver and J. Wardle, Perceived effects of stress on food choice, Physiol Behav, vol.66, pp.511-515, 1999.

M. F. Dallman, N. Pecoraro, S. F. Akana, L. Fleur, S. E. Gomez et al., Chronic stress and obesity: a new view of 'comfort food, Proc Natl Acad Sci, vol.100, pp.11696-11701, 2003.

J. P. Aguggia, M. M. Suárez, and M. A. Rivarola, Early maternal separation: neurobehavioral consequences in mother rats, Behav Brain Res, vol.248, pp.25-31, 2013.

R. Krolow, C. G. Noschang, D. Arcego, A. C. Andreazza, W. Peres et al., Consumption of a palatable diet by chronically stressed rats prevents effects on anxiety-like behavior but increases oxidative stress in a sex-specific manner, Appetite, vol.55, pp.108-116, 2010.

B. C. Finger, T. G. Dinan, and J. F. Cryan, High-fat diet selectively protects against the effects of chronic social stress in the mouse, Neuroscience, vol.192, pp.351-360, 2011.

J. Maniam and M. J. Morris, Long-term postpartum anxiety and depression-like behavior in mother rats subjected to maternal separation are ameliorated by palatable high fat diet, Behav Brain Res, vol.208, pp.72-79, 2010.

F. A. Champagne and M. J. Meaney, Stress During Gestation Alters Postpartum Maternal Care and the Development of the Offspring in a Rodent Model, Biol Psychiatry, vol.59, pp.1227-1235, 2006.

G. A. Dunn and T. L. Bale, Maternal High-Fat Diet Effects on Third-Generation Female Body Size via the Paternal Lineage, Endocrinology, vol.152, pp.2228-2236, 2011.

B. S. Mcewen, Understanding the potency of stressful early life experiences on brain and body function, Metabolism, vol.57, issue.2, pp.11-16, 2008.

D. A. Hackman, M. J. Farah, and M. J. Meaney, Socioeconomic status and the brain: mechanistic insights from human and animal research, Nat Rev Neurosci, vol.11, pp.651-660, 2010.

P. O. Mcgowan, A. Sasaki, D. 'alessio, and A. C. , Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse, Nat Neurosci, vol.12, pp.342-350, 2009.

N. Provençal and E. B. Binder, The effects of early life stress on the epigenome: From the womb to adulthood and even before, Exp Neurol, vol.268, pp.10-20, 2015.

S. Levine, Infantile experience and resistance to physiological stress, Science, vol.126, p.405, 1957.

D. Francis, J. Diorio, and D. Liu, Nongenomic transmission across generations of maternal behavior and stress responses in the rat, Science, vol.286, pp.1155-1163, 1999.

V. Daugé, Impact neurobiologique de la séparation mère/nouveau-né chez le rongeur, Med Sci (Paris), vol.19, pp.607-618, 2003.

V. Vazquez, J. Penit-soria, and C. Durand, Maternal deprivation increases vulnerability to morphine dependence and disturbs the enkephalinergic system in adulthood, J Neurosci, vol.25, pp.4453-62, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00085367

T. B. Franklin, B. J. Saab, and I. M. Mansuy, Neural mechanisms of stress resilience and vulnerability, Neuron, vol.75, pp.747-61, 2012.

F. Barreau, L. Ferrier, and J. Fioramonti, New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models, Pediatr Res, vol.62, pp.240-245, 2007.

N. M. Talge, C. Neal, and V. Glover, Antenatal maternal stress and long-term effects on child neurodevelopment: how and why?, J Child Psychol Psychiatry, vol.48, pp.245-61, 2007.

J. M. Joffe, Genotype and prenatal and premating stress interact to affect adult behavior in rats, Science, vol.150, pp.1844-1849, 1965.

M. Darnaudéry and S. Maccari, Epigenetic programming of the stress response in male and female rats by prenatal restraint stress, Brain Res Rev, vol.57, pp.571-85, 2008.

S. Maccari, H. J. Krugers, and S. Morley-fletcher, The consequences of earlylife adversity: neurobiological, behavioural and epigenetic adaptations, J Neuroendocrinol, vol.26, pp.707-730, 2014.

M. P. Moisan, L. Moal, and M. , Le stress dans tous ses états, Med Sci, vol.28, pp.612-619, 2012.

V. Lemaire, M. Koehl, L. Moal, and M. , Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus, Proc Natl Acad Sci USA, vol.97, pp.11032-11039, 2000.
URL : https://hal.archives-ouvertes.fr/hal-02326250

J. Marrocco, J. Mairesse, and R. T. Ngomba, Anxiety-like behavior of prenatally stressed rats is associated with a selective reduction of glutamate release in the ventral hippocampus, J Neurosci, vol.32, pp.17143-54, 2012.

M. Weinstock, Sex-dependent changes induced by prenatal stress in cortical and hippocampal morphology and behaviour in rats: an update, Stress Amst Neth, vol.14, pp.604-617, 2011.

B. Labonté, M. Suderman, and G. Maussion, Genome-wide epigenetic regulation by early-life trauma, Arch Gen Psychiatry, vol.69, pp.722-753, 2012.

J. Bock, T. Wainstock, and K. Braun, Stress in utero: prenatal programming of brain plasticity and cognition, Biol Psychiatry, vol.78, pp.315-341, 2015.

T. L. Bale, Epigenetic and transgenerational reprogramming of brain development, Nat Rev Neurosci, vol.16, pp.332-376, 2015.

S. S. Richardson, C. R. Daniels, and M. W. Gillman, Society: don't blame the mothers, Nature, vol.512, pp.131-133, 2014.

F. Campbell, G. Conti, and J. J. Heckman, Early childhood investments substantially boost adult health, Science, vol.343, pp.1478-85, 2014.

J. J. Heckman, Schools, skills, and synapses, Econ Inq, vol.46, p.289, 2008.

J. Marrocco, M. Reynaert, and E. Gatta, The effects of antidepressant treatment in prenatally stressed rats support the glutamatergic hypothesis of stress-related disorders, J Neurosci, vol.34, pp.2015-2039, 2014.

S. Morley-fletcher, M. Darnaudéry, and E. Mocaer, Chronic treatment with imipramine reverses immobility behaviour, hippocampal corticosteroid receptors and cortical 5-HT(1A) receptor mRNA in prenatally stressed rats, Neuropharmacology, vol.47, pp.841-848, 2004.

J. Mairesse, J. Lesage, and C. Breton, Maternal stress alters endocrine function of the fetoplacental unit in rats, Am J Physiol Endocrinol Metab, vol.292, pp.1526-1559, 2007.

A. Barbazanges, P. V. Piazza, L. Moal, and M. , Maternal glucocorticoid secretion mediates long-term effects of prenatal stress, J Neurosci, vol.16, pp.3943-3952, 1996.

S. Maccari, P. V. Piazza, and M. Kabbaj, Adoption reverses the long-term impairment in glucocorticoid feedback induced by prenatal stress, J Neurosci, vol.15, pp.110-116, 1995.

I. Weaver, N. Cervoni, and F. A. Champagne, Epigenetic programming by maternal behavior, Nat Neurosci, vol.7, pp.847-54, 2004.

C. Tesone-coelho, L. J. Morel, and J. Bhatt, Vulnerability to opiate intake in maternally deprived rats: implication of MeCP2 and of histone acetylation, Addict Biol, vol.20, pp.120-151, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01204477