Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 16(1): 2361660, 2024.
Article in English | MEDLINE | ID: mdl-38935764

ABSTRACT

The microbiota significantly impacts digestive epithelium functionality, especially in nutrient processing. Given the importance of iron for both the host and the microbiota, we hypothesized that host-microbiota interactions fluctuate with dietary iron levels. We compared germ-free (GF) and conventional mice (SPF) fed iron-containing (65 mg/Kg) or iron-depleted (<6 mg/Kg) diets. The efficacy of iron privation was validated by iron blood parameters. Ferritin and Dmt1, which represent cellular iron storage and transport respectively, were studied in tissues where they are abundant: the duodenum, liver and lung. When the mice were fed an iron-rich diet, the microbiota increased blood hemoglobin and hepcidin and the intestinal ferritin levels, suggesting that the microbiota helps iron storage. When iron was limiting, the microbiota inhibited the expression of the intestinal Dmt1 transporter, likely via the pathway triggered by Hif-2α. The microbiota assists the host in storing intestinal iron when it is abundant and competes with the host by inhibiting Dmt1 in conditions of iron scarcity. Comparison between duodenum, liver and lung indicates organ-specific responses to microbiota and iron availability. Iron depletion induced temporal changes in microbiota composition and activity, reduced α-diversity of microbiota, and led to Lactobacillaceae becoming particularly more abundant after 60 days of privation. By inoculating GF mice with a simplified bacterial mixture, we show that the iron-depleted host favors the gut fitness of Bifidobacterium longum.


Subject(s)
Cation Transport Proteins , Duodenum , Gastrointestinal Microbiome , Hepcidins , Iron, Dietary , Liver , Animals , Mice , Gastrointestinal Microbiome/physiology , Iron, Dietary/metabolism , Iron, Dietary/administration & dosage , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Liver/metabolism , Liver/microbiology , Duodenum/metabolism , Duodenum/microbiology , Hepcidins/metabolism , Ferritins/metabolism , Germ-Free Life , Host Microbial Interactions , Lung/microbiology , Lung/metabolism , Iron/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Mice, Inbred C57BL , Hemoglobins/metabolism , Male
2.
Nat Commun ; 13(1): 6834, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36400769

ABSTRACT

Defects in RNA splicing have been linked to human disorders, but remain poorly explored in inflammatory bowel disease (IBD). Here, we report that expression of the chromatin and alternative splicing regulator HP1γ is reduced in ulcerative colitis (UC). Accordingly, HP1γ gene inactivation in the mouse gut epithelium triggers IBD-like traits, including inflammation and dysbiosis. In parallel, we find that its loss of function broadly increases splicing noise, favoring the usage of cryptic splice sites at numerous genes with functions in gut biology. This results in the production of progerin, a toxic splice variant of prelamin A mRNA, responsible for the Hutchinson-Gilford Progeria Syndrome of premature aging. Splicing noise is also extensively detected in UC patients in association with inflammation, with progerin transcripts accumulating in the colon mucosa. We propose that monitoring HP1γ activity and RNA splicing precision can help in the management of IBD and, more generally, of accelerated aging.


Subject(s)
Colitis, Ulcerative , Progeria , Humans , Mice , Animals , Chromobox Protein Homolog 5 , Colitis, Ulcerative/genetics , RNA Splicing/genetics , Progeria/genetics , Progeria/metabolism , Inflammation
3.
Microbiome ; 9(1): 157, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238386

ABSTRACT

BACKGROUND: Autism spectrum disorders (ASD) are associated with dysregulation of the microbiota-gut-brain axis, changes in microbiota composition as well as in the fecal, serum, and urine levels of microbial metabolites. Yet a causal relationship between dysregulation of the microbiota-gut-brain axis and ASD remains to be demonstrated. Here, we hypothesized that the microbial metabolite p-Cresol, which is more abundant in ASD patients compared to neurotypical individuals, could induce ASD-like behavior in mice. RESULTS: Mice exposed to p-Cresol for 4 weeks in drinking water presented social behavior deficits, stereotypies, and perseverative behaviors, but no changes in anxiety, locomotion, or cognition. Abnormal social behavior induced by p-Cresol was associated with decreased activity of central dopamine neurons involved in the social reward circuit. Further, p-Cresol induced changes in microbiota composition and social behavior deficits could be transferred from p-Cresol-treated mice to control mice by fecal microbiota transplantation (FMT). We also showed that mice transplanted with the microbiota of p-Cresol-treated mice exhibited increased fecal p-Cresol excretion, compared to mice transplanted with the microbiota of control mice. In addition, we identified possible p-Cresol bacterial producers. Lastly, the microbiota of control mice rescued social interactions, dopamine neurons excitability, and fecal p-Cresol levels when transplanted to p-Cresol-treated mice. CONCLUSIONS: The microbial metabolite p-Cresol induces selectively ASD core behavioral symptoms in mice. Social behavior deficits induced by p-Cresol are dependant on changes in microbiota composition. Our study paves the way for therapeutic interventions targeting the microbiota and p-Cresol production to treat patients with ASD. Video abstract.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Gastrointestinal Microbiome , Animals , Autistic Disorder/etiology , Cresols , Fecal Microbiota Transplantation , Humans , Mice
4.
Front Immunol ; 12: 745535, 2021.
Article in English | MEDLINE | ID: mdl-35069524

ABSTRACT

Food allergy is associated with alterations in the gut microbiota, epithelial barrier, and immune tolerance. These dysfunctions are observed within the first months of life, indicating that early intervention is crucial for disease prevention. Preventive nutritional strategies with prebiotics are an attractive option, as prebiotics such as galacto-oligosaccharides and inulin can promote tolerance, epithelial barrier reinforcement, and gut microbiota modulation. Nonetheless, the ideal period for intervention remains unknown. Here, we investigated whether galacto-oligosaccharide/inulin supplementation during gestation could protect offspring from wheat allergy development in BALB/cJRj mice. We demonstrated that gestational prebiotic supplementation promoted the presence of beneficial strains in the fecal microbiota of dams during gestation and partially during mid-lactation. This specific microbiota was transferred to their offspring and maintained to adulthood. The presence of B and T regulatory immune cell subsets was also increased in the lymph nodes of offspring born from supplemented mothers, suggestive of a more tolerogenic immune environment. Indeed, antenatal prebiotic supplementation reduced the development of wheat allergy symptoms in offspring. Our study thus demonstrates that prebiotic supplementation during pregnancy induces, in the offspring, a tolerogenic environment and a microbial imprint that mitigates food allergy development.


Subject(s)
Dietary Supplements , Food Hypersensitivity , Gastrointestinal Microbiome , Inulin/pharmacology , Prebiotics , Prenatal Exposure Delayed Effects , Animals , Female , Food Hypersensitivity/immunology , Food Hypersensitivity/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/microbiology , Prenatal Exposure Delayed Effects/prevention & control
5.
Mol Nutr Food Res ; 64(15): e1900835, 2020 08.
Article in English | MEDLINE | ID: mdl-32579743

ABSTRACT

SCOPE: Protein malnutrition is characterized by stunted growth, hepatic steatosis and a damaged gut mucosal architecture. Since high-fat shaped gut microbiota (HFM) has an increased ability in providing nutrients and energy from food to the host, the aim of this study is to determine whether such a microbiota could beneficially impact on the consequences of malnutrition. METHODS AND RESULTS: The cecal content of specific pathogen free C57Bl/6J mice fed a high-fat diet or a low-protein diet is transplanted in two groups of germ-free C57Bl/6J recipient mice, which are subsequently fed a low-protein diet for 8 weeks. Body weight gain is comparable between the two groups of microbiota-recipient mice. The HFM led to a worsening of microvesicular steatosis and a decrease of plasma lipids compared to the low-protein shaped microbiota. In the small intestine of mice receiving the HFM, although significant histological differences are not observed, the expression of antimicrobial genes promoting oxidative stress and immune response at the ileal epithelium (Duox2, Duoxa2, Saa1, Ang4, Defa5) is increased. CONCLUSION: The transplant of HFM in mice fed a low-protein diet represents a noxious stimulus for the ileal mucosa and impairs hepatic lipoprotein secretion, favoring the occurrence of hepatic microvesicular steatosis.


Subject(s)
Diet, High-Fat , Diet, Protein-Restricted/adverse effects , Gastrointestinal Microbiome/physiology , Non-alcoholic Fatty Liver Disease/microbiology , Animals , Cecum/microbiology , Cholesterol/blood , Dysbiosis/genetics , Dysbiosis/microbiology , Eating , Feces/microbiology , Gene Expression , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Organ Size , Triglycerides/blood , Weight Gain
6.
Behav Brain Res ; 384: 112549, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32050097

ABSTRACT

We have previously provided the first evidence that the microbiota modulates the physiology of the olfactory epithelium using germfree mice. The extent to which changes to the olfactory system depend on the microbiota is still unknown. In the present work, we explored if different microbiota would differentially impact olfaction. We therefore studied the olfactory function of three groups of mice of the same genetic background, whose parents had been conventionalized before mating with microbiota from three different mouse strains. Caecal short chain fatty acids profiles and 16S rRNA gene sequencing ascertained that gut microbiota differed between the three groups. We then used a behavioural test to measure the attractiveness of various odorants and observed that the three groups of mice differed in their attraction towards odorants. Their olfactory epithelium properties, including electrophysiological responses recorded by electro-olfactograms and expression of genes related to the olfactory transduction pathway, also showed several differences. Overall, our data demonstrate that differences in gut microbiota profiles are associated with differences in olfactory preferences and in olfactory epithelium functioning.


Subject(s)
Behavior, Animal , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Olfactory Mucosa/physiology , Smell/physiology , Animals , Bacteroidetes , Cecum , Electrodiagnosis , Firmicutes , Gastrointestinal Contents/chemistry , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Odorants , RNA, Ribosomal, 16S/genetics
7.
mBio ; 11(1)2020 02 04.
Article in English | MEDLINE | ID: mdl-32019793

ABSTRACT

Gut microbiota-derived endotoxin has been linked to human nonalcoholic fatty liver disease (NAFLD), but the specific causative agents and their molecular mechanisms remain elusive. In this study, we investigated whether bacterial strains of endotoxin-producing pathogenic species overgrowing in obese human gut can work as causative agents for NAFLD. We further assessed the role of lipopolysaccharide (LPS)-Toll-like receptor 4 (TLR4) cross talk in this pathogenicity. Nonvirulent strains of Gram-negative pathobionts were isolated from obese human gut and monoassociated with C57BL/6J germfree (GF) mice fed a high-fat diet (HFD). Deletion of waaG in the bacterial endotoxin synthetic pathway and knockout of TLR4 in GF mice were used to further study the underlying mechanism for a causal relationship between these strains and the development of NAFLD. Three endotoxin-producing strains, Enterobacter cloacae B29, Escherichia coli PY102, and Klebsiella pneumoniae A7, overgrowing in the gut of morbidly obese volunteers with severe fatty liver, induced NAFLD when monoassociated with GF mice on HFD, while HFD alone did not induce the disease in GF mice. The commensal Bacteroides thetaiotaomicron (ATCC 29148), whose endotoxin activity was markedly lower than that of Enterobacteriaceae strains, did not induce NAFLD in GF mice. B29 lost its proinflammatory properties and NAFLD-inducing capacity upon deletion of the waaG gene. Moreover, E. cloacae B29 did not induce NAFLD in TLR4-deficient GF mice. These nonvirulent endotoxin-producing strains in pathobiont species overgrowing in human gut may work as causative agents, with LPS-TLR4 cross talk as the most upstream and essential molecular event for NAFLD.IMPORTANCE Recent studies have reported a link between gut microbiota and nonalcoholic fatty liver disease (NAFLD), showing that germfree (GF) mice do not develop metabolic syndromes, including NAFLD. However, the specific bacterial species causing NAFLD, as well as their molecular cross talk with the host for driving liver disease, remain elusive. Here, we found that nonvirulent endotoxin-producing strains of pathogenic species overgrowing in obese human gut can act as causative agents for induction of NAFLD and related metabolic disorders. The cross talk between endotoxin from these specific producers and the host's TLR4 receptor is the most upstream and essential molecular event for inducing all phenotypes in NAFLD and related metabolic disorders. These nonvirulent endotoxin-producing strains of gut pathogenic species overgrowing in human gut may collectively become a predictive biomarker or serve as a novel therapeutic target for NAFLD and related metabolic disorders.


Subject(s)
Endotoxins/biosynthesis , Enterobacteriaceae/metabolism , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/microbiology , Signal Transduction , Toll-Like Receptor 4/metabolism , Adult , Animals , Child, Preschool , Diet, High-Fat , Enterobacteriaceae/genetics , Female , Germ-Free Life , Humans , Male , Mice , Mice, Inbred C57BL , Obesity, Morbid
8.
Nutrients ; 12(2)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973214

ABSTRACT

Interactions of diet, gut microbiota, and host genetics play essential roles in the development of metabolic diseases. A/J and C57BL/6J (C57) are two mouse strains known to display different susceptibilities to metabolic disorders. In this context, we analyzed gut microbiota composition in A/J and C57 mice, and assessed its responses to high-fat diet (HFD) and antibiotic (AB) treatment. We also exchanged the gut microbiota between the two strains following AB treatment to evaluate its impact on the metabolism. We showed that A/J and C57 mice have different microbiome structure and composition at baseline. Moreover, A/J and C57 microbiomes responded differently to HFD and AB treatments. Exchange of the gut microbiota between the two strains was successful as recipients' microbiota resembled donor-strain microbiota. Seven weeks after inoculation, the differences between recipients persisted and were still closer from the donor-strain microbiota. Despite effective microbiota transplants, the response to HFD was not markedly modified in C57 and A/J mice. Particularly, body weight gain and glucose intolerance in response to HFD remained different in the two mouse strains whatever the changes in microbiome composition. This indicated that genetic background has a much stronger impact on metabolic responses to HFD than gut microbiome composition.


Subject(s)
Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/genetics , Genetic Background , Metabolic Diseases/genetics , Metabolic Diseases/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Genetic Predisposition to Disease/genetics , Metabolic Diseases/etiology , Mice , Mice, Inbred C57BL
9.
Front Neurosci ; 12: 216, 2018.
Article in English | MEDLINE | ID: mdl-29686603

ABSTRACT

Gut microbiota produces a wide and diverse array of metabolites that are an integral part of the host metabolome. The emergence of the gut microbiome-brain axis concept has prompted investigations on the role of gut microbiota dysbioses in the pathophysiology of brain diseases. Specifically, the search for microbe-related metabolomic signatures in human patients and animal models of psychiatric disorders has pointed out the importance of the microbial metabolism of aromatic amino acids. Here, we investigated the effect of indole on brain and behavior in rats. Indole is produced by gut microbiota from tryptophan, through the tryptophanase enzyme encoded by the tnaA gene. First, we mimicked an acute and high overproduction of indole by injecting this compound in the cecum of conventional rats. This treatment led to a dramatic decrease of motor activity. The neurodepressant oxidized derivatives of indole, oxindole and isatin, accumulated in the brain. In addition, increase in eye blinking frequency and in c-Fos protein expression in the dorsal vagal complex denoted a vagus nerve activation. Second, we mimicked a chronic and moderate overproduction of indole by colonizing germ-free rats with the indole-producing bacterial species Escherichia coli. We compared emotional behaviors of these rats with those of germ-free rats colonized with a genetically-engineered counterpart strain unable to produce indole. Rats overproducing indole displayed higher helplessness in the tail suspension test, and enhanced anxiety-like behavior in the novelty, elevated plus maze and open-field tests. Vagus nerve activation was suggested by an increase in eye blinking frequency. However, unlike the conventional rats dosed with a high amount of indole, the motor activity was not altered and neither oxindole nor isatin could be detected in the brain. Further studies are required for a comprehensive understanding of the mechanisms supporting indole effects on emotional behaviors. As our findings suggest that people whose gut microbiota is highly prone to produce indole could be more likely to develop anxiety and mood disorders, we addressed the issue of the inter-individual variability of indole producing potential in humans. An in silico investigation of metagenomic data focused on the tnaA gene products definitively proved this inter-individual variability.

10.
Article in English | MEDLINE | ID: mdl-27827942

ABSTRACT

The presence of pesticide residues in food is a public health problem. Exposure to these substances in daily life could have serious effects on the intestine-the first organ to come into contact with food contaminants. The present study investigated the impact of a low dose (1 mg/day in oil) of the pesticide chlorpyrifos (CPF) on the community structure, diversity and metabolic response of the human gut microbiota using the SHIME® model (six reactors, representing the different parts of the gastrointestinal tract). The last three reactors (representing the colon) were inoculated with a mixture of feces from human adults. Three time points were studied: immediately before the first dose of CPF, and then after 15 and 30 days of CPF-oil administration. By using conventional bacterial culture and molecular biology methods, we showed that CPF in oil can affect the gut microbiota. It had the greatest effects on counts of culturable bacteria (with an increase in Enterobacteria, Bacteroides spp. and clostridia counts, and a decrease in bifidobacterial counts) and fermentative activity, which were colon-segment-dependent. Our results suggest that: (i) CPF in oil treatment affects the gut microbiota (although there was some discordance between the culture-dependent and culture-independent analyses); (ii) the changes are "SHIME®-compartment" specific; and (iii) the changes are associated with minor alterations in the production of short-chain fatty acids and lactate.


Subject(s)
Chlorpyrifos/toxicity , Environmental Pollutants/toxicity , Gastrointestinal Microbiome/drug effects , Insecticides/toxicity , Humans , Models, Biological , Models, Theoretical
11.
Sci Rep ; 6: 32484, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27577172

ABSTRACT

The gut microbiota is involved in many aspects of host physiology but its role in body weight and glucose metabolism remains unclear. Here we studied the compositional changes of gut microbiota in diet-induced obesity mice that were conventionally raised or received microbiota transplantation. In conventional mice, the diversity of the faecal microbiota was weakly associated with 1(st) week weight gain but transferring the microbiota of mice with contrasting weight gain to germfree mice did not change obesity development or feed efficiency of recipients regardless whether the microbiota was taken before or after 10 weeks high fat (HF) feeding. Interestingly, HF-induced glucose intolerance was influenced by microbiota inoculation and improved glucose tolerance was associated with a low Firmicutes to Bacteroidetes ratio. Transplantation of Bacteroidetes rich microbiota compared to a control microbiota ameliorated glucose intolerance caused by HF feeding. Altogether, our results demonstrate that gut microbiota is involved in the regulation of glucose metabolism and the abundance of Bacteroidetes significantly modulates HF-induced glucose intolerance but has limited impact on obesity in mice. Our results suggest that gut microbiota is a part of complex aetiology of insulin resistance syndrome, individual microbiota composition may cause phenotypic variation associated with HF feeding in mice.


Subject(s)
Diet, High-Fat , Dietary Fats/adverse effects , Glucose Intolerance/metabolism , Obesity/metabolism , Animals , Bacteroidetes/classification , Bacteroidetes/growth & development , Fecal Microbiota Transplantation , Firmicutes/classification , Firmicutes/growth & development , Gastrointestinal Microbiome/physiology , Glucose Intolerance/etiology , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/microbiology , Obesity/pathology , Proteobacteria/classification , Proteobacteria/growth & development
12.
J Infect Dis ; 212(8): 1332-40, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-25838265

ABSTRACT

CCL20 is a chemokine with antimicrobial activity. We investigated its expression and role during neonatal cryptosporidiosis, a worldwide protozoan enteric disease leading to severe diarrhea. Surprisingly, during infection by Cryptosporidium parvum, CCL20 production by the intestine of neonatal mice is reduced by a mechanism independent both of the enteric flora and of interferon γ, a key cytokine for the resolution of this infection. However, oral administration of recombinant CCL20 to neonatal mice significantly reduced the parasite load by a mechanism that was independent of immune cell recruitment and occurred instead by direct cytolytic activity on free stages of the parasite. MiR21 functionally targets CCL20 and is upregulated during the infection, thus contributing to the downregulation of the chemokine. Our findings demonstrate for the first time the direct antiparasitic activity of CCL20 against an enteric protozoan and its downregulation during C. parvum infection, which is detrimental to parasite clearance.


Subject(s)
Anti-Infective Agents/metabolism , Chemokine CCL20/metabolism , Cryptosporidiosis/immunology , Cryptosporidium parvum/physiology , MicroRNAs/genetics , Animals , Animals, Newborn , Cell Line , Chemokine CCL20/genetics , Disease Models, Animal , Epithelial Cells , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intestines/immunology , Intestines/parasitology , Mice , Mice, Inbred C57BL , Recombinant Proteins , Specific Pathogen-Free Organisms , Sporozoites
13.
Psychoneuroendocrinology ; 42: 207-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24636517

ABSTRACT

BACKGROUND AND AIMS: Establishment of the gut microbiota is one of the most important events in early life and emerging evidence indicates that the gut microbiota influences several aspects of brain functioning, including reactivity to stress. To better understand how the gut microbiota contributes to a vulnerability to the stress-related psychiatric disorders, we investigated the relationship between the gut microbiota, anxiety-like behavior and HPA axis activity in stress-sensitive rodents. We also analyzed the monoamine neurotransmitters in the brain upper structures involved in the regulation of stress and anxiety. METHODS: Germfree (GF) and specific pathogen free (SPF) F344 male rats were first subjected to neurological tests to rule out sensorimotor impairments as confounding factors. Then, we examined the behavior responses of rats to social interaction and open-field tests. Serum corticosterone concentrations, CRF mRNA expression levels in the hypothalamus, glucocorticoid receptor (GR) mRNA expression levels in the hippocampus, and monoamine concentrations in the frontal cortex, hippocampus and striatum were compared in rats that were either exposed to the open-field stress or not. RESULTS: GF rats spent less time sniffing an unknown partner than SPF rats in the social interaction test, and displayed a lower number of visits to the aversive central area, and an increase in latency time, time spent in the corners and number of defecations in the open-field test. In response to the open-field stress, serum corticosterone concentrations were 2.8-fold higher in GF than in SPF rats. Compared to that of SPF rats, GF rats showed elevated CRF mRNA expression in the hypothalamus and reduced GR mRNA expression in the hippocampus. GF rats also had a lower dopaminergic turnover rate in the frontal cortex, hippocampus and striatum than SPF rats. CONCLUSIONS: In stress-sensitive F344 rats, absence of the gut microbiota exacerbates the neuroendocrine and behavioral responses to acute stress and the results coexist with alterations of the dopaminergic turnover rate in brain upper structures that are known to regulate reactivity to stress and anxiety-like behavior.


Subject(s)
Anxiety/microbiology , Behavior, Animal/physiology , Intestines/microbiology , Stress, Psychological/microbiology , Animals , Anxiety/etiology , Anxiety/metabolism , Brain/metabolism , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Microbiota , Pituitary-Adrenal System/metabolism , Rats , Rats, Inbred F344 , Receptors, Glucocorticoid/metabolism , Social Behavior , Stress, Psychological/complications
14.
Food Chem ; 141(4): 3291-300, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23993484

ABSTRACT

Ochratoxin A (OTA) is a mycotoxin frequently encountered in coffee. The relevance of this contaminant in the colon upon digestion necessitates a study on its interaction with colon microbiota. Here, the fate of OTA during colon digestion was investigated using a dynamic simulator of the human gut. The influence of coffee as a food matrix was taken into account, as it may affect the colonic microbial ecosystem and, consequently, the fate of OTA. Biodegradation was followed by measuring OTA concentration over time, and by screening for several possible metabolites, using LC-ESI-MS and HRMS. The descending colon was found to be the main site of OTA biodegradation. Two metabolites, ochratoxin α and ochratoxin B, were identified, suggesting that biodegradation by gut microbiota is beneficial for the host, as they are considered less toxic than OTA. The extent of biodegradation was reduced in the presence of the coffee matrix, possibly due to competition for available carbon sources. Effects of OTA and the coffee matrix on the microbial ecosystem were contrasting. While OTA caused a specific, but lasting loss, of the beneficial species Lactobacillus reuteri, coffee temporarily altered the fermentation pattern towards lower ammonia and higher acetate and propionate production, likely due to its dietary fibre content.


Subject(s)
Coffee/metabolism , Colon/metabolism , Food Contamination/analysis , Ochratoxins/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Coffea/chemistry , Coffea/metabolism , Coffee/chemistry , Colon/microbiology , Humans , Microbiota , Models, Biological , Ochratoxins/chemistry
15.
PLoS One ; 8(8): e71026, 2013.
Article in English | MEDLINE | ID: mdl-23951074

ABSTRACT

Prebiotic fibres like short-chain fructo-oligosaccharides (scFOS) are known to selectively modulate the composition of the intestinal microbiota and especially to stimulate Bifidobacteria. In parallel, the involvement of intestinal microbiota in host metabolic regulation has been recently highlighted. The objective of the study was to evaluate the effect of scFOS on the composition of the faecal microbiota and on metabolic parameters in an animal model of diet-induced obesity harbouring a human-type microbiota. Forty eight axenic C57BL/6J mice were inoculated with a sample of faecal human microbiota and randomly assigned to one of 3 diets for 7 weeks: a control diet, a high fat diet (HF, 60% of energy derived from fat)) or an isocaloric HF diet containing 10% of scFOS (HF-scFOS). Mice fed with the two HF gained at least 21% more weight than mice from the control group. Addition of scFOS partially abolished the deposition of fat mass but significantly increased the weight of the caecum. The analysis of the taxonomic composition of the faecal microbiota by FISH technique revealed that the addition of scFOS induced a significant increase of faecal Bifidobacteria and the Clostridium coccoides group whereas it decreased the Clostridium leptum group. In addition to modifying the composition of the faecal microbiota, scFOS most prominently affected the faecal metabolome (e.g. bile acids derivatives, hydroxyl monoenoic fatty acids) as well as urine, plasma hydrophilic and plasma lipid metabolomes. The increase in C. coccoides and the decrease in C. leptum, were highly correlated to these metabolic changes, including insulinaemia, as well as to the weight of the caecum (empty and full) but not the increase in Bifidobacteria. In conclusion scFOS induce profound metabolic changes by modulating the composition and the activity of the intestinal microbiota, that may partly explain their effect on the reduction of insulinaemia.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/microbiology , Metabolome , Microbiota/drug effects , Oligosaccharides/administration & dosage , Animals , Bifidobacterium/classification , Cluster Analysis , Diet , Feces/microbiology , Germ-Free Life , Glucose Tolerance Test , Humans , Male , Metabolomics , Mice , Mice, Obese , Obesity/etiology , Oligosaccharides/chemistry
16.
Gut ; 62(12): 1787-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23197411

ABSTRACT

OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD) is prevalent among obese people and is considered the hepatic manifestation of metabolic syndrome. However, not all obese individuals develop NAFLD. Our objective was to demonstrate the role of the gut microbiota in NAFLD development using transplantation experiments in mice. DESIGN: Two donor C57BL/6J mice were selected on the basis of their responses to a high-fat diet (HFD). Although both mice displayed similar body weight gain, one mouse, called the 'responder', developed hyperglycaemia and had a high plasma concentration of pro-inflammatory cytokines. The other, called a 'non-responder', was normoglycaemic and had a lower level of systemic inflammation. Germ-free mice were colonised with intestinal microbiota from either the responder or the non-responder and then fed the same HFD. RESULTS: Mice that received microbiota from different donors developed comparable obesity on the HFD. The responder-receiver (RR) group developed fasting hyperglycaemia and insulinaemia, whereas the non-responder-receiver (NRR) group remained normoglycaemic. In contrast to NRR mice, RR mice developed hepatic macrovesicular steatosis, which was confirmed by a higher liver concentration of triglycerides and increased expression of genes involved in de-novo lipogenesis. Pyrosequencing of the 16S ribosomal RNA genes revealed that RR and NRR mice had distinct gut microbiota including differences at the phylum, genera and species levels. CONCLUSIONS: Differences in microbiota composition can determine response to a HFD in mice. These results further demonstrate that the gut microbiota contributes to the development of NAFLD independently of obesity.


Subject(s)
Fatty Liver/microbiology , Intestines/microbiology , Animals , Blood Glucose/analysis , Dietary Fats/adverse effects , Fatty Acids, Volatile/blood , Fatty Liver/etiology , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Microbiota/genetics , Microbiota/physiology , Non-alcoholic Fatty Liver Disease , Polymerase Chain Reaction , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Triglycerides/analysis
17.
Environ Microbiol ; 13(10): 2667-80, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21883787

ABSTRACT

The endogenous gut microbiota affects the host in many ways. Prebiotics should favour beneficial intestinal microbes and thus improve host health. In this study, we investigated how a novel class of potential prebiotic long-chain arabinoxylans (LC-AX) and the well-established prebiotic inulin (IN) modulate the gut microbiota of humanized rats. Six weeks after axenic rats were inoculated with a human faecal microbiota, their colonic microbiota was similar to this inoculum (∼ 70%), whereas their caecal microbiota was enriched with Verrucomicrobia and Firmicutes concomitant with lower abundance of Bacteroidetes. Moreover, different Bifidobacterium species colonized the lumen (B. adolescentis) and mucus (B. longum and B. bifidum). Both LC-AX and IN increased SCFA levels and induced a shift from acetate towards health-promoting propionate and butyrate respectively. By applying a high-resolution phylogenetic micro-array (HITChip) at the site of fermentation (caecum), IN and LC-AX were shown to stimulate bacterial groups with known butyrate-producers (Roseburia intestinalis, Eubacterium rectale, Anaerostipes caccae) and bifidobacteria (B. longum) respectively. Prebiotic administration also resulted in lower caecal abundances of the mucin-degrading Akkermansia muciniphila and potentially more mucin production by the host. Both factors might explain the increased caecal mucin levels for LC-AX (threefold) and IN (sixfold). These mucins were degraded along the colon, resulting in high faecal abundances of Akkermansia muciniphila for LC-AX and especially IN-treated rats. Finally, the microbial changes caused an adaptation period for the host with less weight gain, after which the host fine-tuned the interaction with this altered microbiota. Our results demonstrate that next to IN, LC-AX are promising prebiotic compounds by stimulating production of health-promoting metabolites by specific microbes in the proximal regions. Further, prebiotic supplementation shifted mucin degradation to distal regions, where mucin-degraders may produce beneficial metabolites (e.g. propionate by Akkermansia muciniphila), so that prebiotics may potentially improve gut health along the entire length of the intestine.


Subject(s)
Cecum/microbiology , Colon/microbiology , Inulin/pharmacology , Metagenome/drug effects , Mucins/metabolism , Xylans/pharmacology , Animals , Bacteria/drug effects , Bacteria/genetics , Bacteria/metabolism , Feces/microbiology , Fermentation , Humans , Male , Oligonucleotide Array Sequence Analysis , Phylogeny , Prebiotics , RNA, Ribosomal, 16S/genetics , Rats , Rats, Inbred F344 , Young Adult
18.
FASEB J ; 24(12): 4948-59, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20724524

ABSTRACT

Recent studies showed that germ-free (GF) mice are resistant to obesity when consuming a high-fat, high-carbohydrate Western diet. However, it remains unclear what mechanisms are involved in the antiobesity phenotype and whether GF mice develop insulin resistance and dyslipidemia with high-fat (HF) feeding. In the present study, we compared the metabolic consequences of HF feeding on GF and conventional (conv) C57BL/6J mice. GF mice consumed fewer calories, excreted more fecal lipids, and weighed significantly less than conv mice. GF/HF animals also showed enhanced insulin sensitivity with improved glucose tolerance, reduced fasting and nonfasting insulinemia, and increased phospho-Akt((Ser-473)) in adipose tissue. In association with enhanced insulin sensitivity, GF/HF mice had reduced plasma TNF-α and total serum amyloid A concentrations. Reduced hypercholesterolemia, a moderate accretion of hepatic cholesterol, and an increase in fecal cholesterol excretion suggest an altered cholesterol metabolism in GF/HF mice. Pronounced nucleus SREBP2 proteins and up-regulation of cholesterol biosynthesis genes indicate that enhanced cholesterol biosynthesis contributed to the cholesterol homeostasis in GF/HF mice. Our results demonstrate that fewer calorie consumption and increased lipid excretion contributed to the obesity-resistant phenotype of GF/HF mice and reveal that insulin sensitivity and cholesterol metabolism are metabolic targets influenced by the gut microbiota.


Subject(s)
Cholesterol/metabolism , Dietary Fats/adverse effects , Insulin Resistance/physiology , Animals , Blotting, Western , Body Weight/physiology , Germ-Free Life , Glucose Tolerance Test , Glycogen/metabolism , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
19.
Appl Environ Microbiol ; 76(15): 5237-46, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20562281

ABSTRACT

Dynamic, multicompartment in vitro gastrointestinal simulators are often used to monitor gut microbial dynamics and activity. These reactors need to harbor a microbial community that is stable upon inoculation, colon region specific, and relevant to in vivo conditions. Together with the reproducibility of the colonization process, these criteria are often overlooked when the modulatory properties from different treatments are compared. We therefore investigated the microbial colonization process in two identical simulators of the human intestinal microbial ecosystem (SHIME), simultaneously inoculated with the same human fecal microbiota with a high-resolution phylogenetic microarray: the human intestinal tract chip (HITChip). Following inoculation of the in vitro colon compartments, microbial community composition reached steady state after 2 weeks, whereas 3 weeks were required to reach functional stability. This dynamic colonization process was reproducible in both SHIME units and resulted in highly diverse microbial communities which were colon region specific, with the proximal regions harboring saccharolytic microbes (e.g., Bacteroides spp. and Eubacterium spp.) and the distal regions harboring mucin-degrading microbes (e.g., Akkermansia spp.). Importantly, the shift from an in vivo to an in vitro environment resulted in an increased Bacteroidetes/Firmicutes ratio, whereas Clostridium cluster IX (propionate producers) was enriched compared to clusters IV and XIVa (butyrate producers). This was supported by proportionally higher in vitro propionate concentrations. In conclusion, high-resolution analysis of in vitro-cultured gut microbiota offers new insight on the microbial colonization process and indicates the importance of digestive parameters that may be crucial in the development of new in vitro models.


Subject(s)
Bacteroidetes/growth & development , Biodiversity , Clostridium/growth & development , Colon/microbiology , Metagenome , Selection, Genetic , Humans , Models, Biological , Models, Theoretical
20.
Am J Physiol Gastrointest Liver Physiol ; 299(2): G348-57, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466941

ABSTRACT

Previous studies have suggested that intestinal microbiota modulates colonic epithelium renewal. The objective of our work was to study the effects of microbiota on colonic epithelium structure and cell cycle-related proteins by using gnotobiotic rats. Colonic crypts and amount of cell cycle-related proteins were compared between germ-free (GF), conventional (CV), and conventionalized rats by histochemistry and Western blot. Ki67 and proliferating cell nuclear antigen (PCNA) were used as surrogates for proliferative cells; p21(cip1) and p27(kip1) were markers of cell cycle arrest; anti- and proapoptotic proteins, Bcl2 and Bax, respectively, were also studied. We observed 40% increase of the crypt proliferative area 2 days after inoculation of GF rats with a complex microbiota. This recruitment of proliferative cells may account for the 30% increase of crypt depth observed between CV and GF rats. The hyperproliferative boost induced by microbiota was compensated by a fourfold increase of p21(cip1) and p27(kip1) involved in cell cycle arrest and a 30% drop of antiapoptotic Bcl2 protein while Bax was unchanged. Inductions of p21(cip1), p27(kip1), and PCNA protein were not paralleled by an increase of the corresponding mRNA. We also showed that p21(cip1) induction by microbiota was partially restored by Bacteroides thetaiotaomicron, Ruminococcus gnavus, and Clostridium paraputrificum. Colonization of the colon by a complex microbiota increases the crypt depth of colon epithelium. This event takes place in conjunction with a multistep process: a hyperproliferative boost accompanied by compensatory events as induction of p21(cip1) and p27(kip1) and decrease of Bcl2.


Subject(s)
Cell Cycle Proteins/biosynthesis , Colon/growth & development , Colon/metabolism , Germ-Free Life , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Metagenome , Adaptation, Physiological , Animals , Blotting, Western , Cell Cycle , Cell Proliferation , Colon/microbiology , Colon/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Histocytochemistry , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...