Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
Psychosom Med ; 85(8): 727-735, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37363967

ABSTRACT

OBJECTIVE: This study assessed the associations of binge eating, compensatory behaviors, and dietary restraint with the composition and diversity of the intestinal microbiota among participants with binge-eating disorder or bulimia nervosa. METHODS: We analyzed data from 265 participants aged 18 to 45 years with current binge-eating disorder or bulimia nervosa enrolled in the Binge Eating Genetics Initiative study. We evaluated the associations of binge-eating frequency; presence/absence and frequency of vomiting, laxative use, and compulsive exercise; and dietary restraint with abundances of gut microbial genera, species, and diversity (Shannon diversity, Faith phylogenetic diversity, and Peilou's evenness) from 16S rRNA gene sequencing. General linear regression models adjusted for potential confounders, including age and current body mass index, were used to test associations; p values were corrected for the false discovery rate. RESULTS: The normalized abundance of four genus- and species-level gut microbes and three diversity indices were lower among Binge Eating Genetics Initiative participants who reported any laxative use compared with those who reported no laxative use. Vomiting frequency was positively associated with the normalized abundance of the genus Escherichia-Shigella , a potential pathobiont, although the association was attenuated to nonsignificance after adjustment for age, body mass index, and binge-eating episodes. CONCLUSIONS: Laxative use was highly and uniformly predictive of a reduced gut microbial diversity including potential commensals and pathobionts, and should be assessed and accounted for in all future studies of eating disorders and the gut microbiota. Future studies should collect data on specific medications-particularly laxatives-and dietary intake to obtain unbiased estimates of the effect of eating disorders on the gut microbiota and identify potential downstream clinical implications.Trial Registration:ClinicalTrials.gov identifier: NCT04162574 .


Subject(s)
Binge-Eating Disorder , Bulimia Nervosa , Bulimia , Feeding and Eating Disorders , Microbiota , Male , Female , Humans , Laxatives , Phylogeny , RNA, Ribosomal, 16S , Vomiting
2.
J Nutr ; 153(4): 1178-1188, 2023 04.
Article in English | MEDLINE | ID: mdl-36841667

ABSTRACT

BACKGROUND: Diet, a key component of type 1 diabetes (T1D) management, modulates the intestinal microbiota and its metabolically active byproducts-including SCFA-through fermentation of dietary carbohydrates such as fiber. However, the diet-microbiome relationship remains largely unexplored in longstanding T1D. OBJECTIVES: We evaluated whether increased carbohydrate intake, including fiber, is associated with increased SCFA-producing gut microbes, SCFA, and intestinal microbial diversity among young adults with longstanding T1D and overweight or obesity. METHODS: Young adult men and women with T1D for ≥1 y, aged 19-30 y, and BMI of 27.0-39.9 kg/m2 at baseline provided stool samples at baseline and 3, 6, and 9 mo of a randomized dietary weight loss trial. Diet was assessed by 1-2 24-h recalls. The abundance of SCFA-producing microbes was measured using 16S rRNA gene sequencing. GC-MS measured fecal SCFA (acetate, butyrate, propionate, and total) concentrations. Adjusted and Bonferroni-corrected generalized estimating equations modeled associations of dietary fiber (total, soluble, and pectins) and carbohydrate (available carbohydrate, and fructose) with microbiome-related outcomes. Primary analyses were restricted to data collected before COVID-19 interruptions. RESULTS: Fiber (total and soluble) and carbohydrates (available and fructose) were positively associated with total SCFA and acetate concentrations (n = 40 participants, 52 visits). Each 10 g/d of total and soluble fiber intake was associated with an additional 8.8 µmol/g (95% CI: 4.5, 12.8 µmol/g; P = 0.006) and 24.0 µmol/g (95% CI: 12.9, 35.1 µmol/g; P = 0.003) of fecal acetate, respectively. Available carbohydrate intake was positively associated with SCFA producers Roseburia and Ruminococcus gnavus. All diet variables except pectin were inversely associated with normalized abundance of Bacteroides and Alistipes. Fructose was inversely associated with Akkermansia abundance. CONCLUSIONS: In young adults with longstanding T1D, fiber and carbohydrate intake were associated positively with fecal SCFA but had variable associations with SCFA-producing gut microbes. Controlled feeding studies should determine whether gut microbes and SCFA can be directly manipulated in T1D.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Female , Humans , Male , Young Adult , Acetates , Dietary Fiber/analysis , Eating , Fatty Acids, Volatile/analysis , Feces/chemistry , Fructose , Obesity , Overweight , RNA, Ribosomal, 16S/genetics
3.
Nutr Metab Cardiovasc Dis ; 33(2): 388-398, 2023 02.
Article in English | MEDLINE | ID: mdl-36586772

ABSTRACT

BACKGROUND AND AIMS: Disordered eating (DE) in type 1 diabetes (T1D) includes insulin restriction for weight loss with serious complications. Gut microbiota-derived short chain fatty acids (SCFA) may benefit host metabolism but are reduced in T1D. We evaluated the hypothesis that DE and insulin restriction were associated with reduced SCFA-producing gut microbes, SCFA, and intestinal microbial diversity in adults with T1D. METHODS AND RESULTS: We collected stool samples at four timepoints in a hypothesis-generating gut microbiome pilot study ancillary to a weight management pilot in young adults with T1D. 16S ribosomal RNA gene sequencing measured the normalized abundance of SCFA-producing intestinal microbes. Gas-chromatography mass-spectrometry measured SCFA (total, acetate, butyrate, and propionate). The Diabetes Eating Problem Survey-Revised (DEPS-R) assessed DE and insulin restriction. Covariate-adjusted and Bonferroni-corrected generalized estimating equations modeled the associations. COVID-19 interrupted data collection, so models were repeated restricted to pre-COVID-19 data. Data were available for 45 participants at 109 visits, which included 42 participants at 65 visits pre-COVID-19. Participants reported restricting insulin "At least sometimes" at 53.3% of visits. Pre-COVID-19, each 5-point DEPS-R increase was associated with a -0.34 (95% CI -0.56, -0.13, p = 0.07) lower normalized abundance of genus Anaerostipes; and the normalized abundance of Lachnospira genus was -0.94 (95% CI -1.5, -0.42), p = 0.02 lower when insulin restriction was reported "At least sometimes" compared to "Rarely or Never". CONCLUSION: DE and insulin restriction were associated with a reduced abundance of SCFA-producing gut microbes pre-COVID-19. Additional studies are needed to confirm these associations to inform microbiota-based therapies in T1D.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 1 , Feeding and Eating Disorders , Gastrointestinal Microbiome , Humans , Young Adult , Diabetes Mellitus, Type 1/diagnosis , Pilot Projects , Fatty Acids, Volatile/metabolism , Insulin , Feces
4.
Arch Womens Ment Health ; 26(2): 227-234, 2023 04.
Article in English | MEDLINE | ID: mdl-36897389

ABSTRACT

Perinatal perceived stress can contribute to worse health outcomes for the parent-child dyad. Given the emerging relationship between the microbiota-gut-brain axis and stress, this study sought to elucidate connections between bowel symptoms and the gut microbiome in relation to perceived stress at three time points in the perinatal period: two during pregnancy and one postpartum. Ninety-five pregnant individuals participated in a prospective cohort study from April 2017 to November 2019. Researchers assessed Perceived Stress Scale-10 (PSS); bowel symptoms (according to the IBS Questionnaire); psychiatrist assessment of new onset or exacerbated depression and anxiety; and fecal samples analyzed for alpha diversity (measures of gut microbiome diversity utilizing Shannon, Observed OTUs, and Faith's PD) at each timepoint. Covariates included weeks of gestation and weeks postpartum. PSS scores were divided into "Perceived Self-Efficacy" and "Perceived Helplessness." Increased gut microbial diversity was associated with decreased bowel symptoms, decreased overall perceived stress, increased ability to cope with adversity, and decreased distress in the postpartum period. This study found a significant association between a less diverse microbial community, lower self-efficacy early in pregnancy, and greater bowel symptoms and perceived helplessness later in the perinatal period, relationships that may ultimately point to novel diagnostic methods and interventions for perceived stress based on the microbiota-gut-brain axis.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Pregnancy , Female , Humans , Brain-Gut Axis , Prospective Studies , Stress, Psychological
5.
BMC Psychiatry ; 20(1): 307, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546136

ABSTRACT

BACKGROUND: The Binge Eating Genetics Initiative (BEGIN) is a multipronged investigation examining the interplay of genomic, gut microbiota, and behavioral factors in bulimia nervosa and binge-eating disorder. METHODS: 1000 individuals who meet current diagnostic criteria for bulimia nervosa or binge-eating disorder are being recruited to collect saliva samples for genotyping, fecal sampling for microbiota characterization, and recording of 30 days of passive data and behavioral phenotyping related to eating disorders using the app Recovery Record adapted for the Apple Watch. DISCUSSION: BEGIN examines the interplay of genomic, gut microbiota, and behavioral factors to explore etiology and develop predictors of risk, course of illness, and response to treatment in bulimia nervosa and binge-eating disorder. We will optimize the richness and longitudinal structure of deep passive and active phenotypic data to lay the foundation for a personalized precision medicine approach enabling just-in-time interventions that will allow individuals to disrupt eating disorder behaviors in real time before they occur. TRIAL REGISTRATION: The ClinicalTrials.gov identifier is NCT04162574. November 14, 2019, Retrospectively Registered.


Subject(s)
Binge-Eating Disorder , Bulimia Nervosa , Bulimia , Feeding and Eating Disorders , Binge-Eating Disorder/genetics , Bulimia/genetics , Bulimia Nervosa/genetics , Feeding Behavior , Humans
6.
PLoS Pathog ; 13(7): e1006471, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28750066

ABSTRACT

Diverse enteropathogen exposures associate with childhood malnutrition. To elucidate mechanistic pathways whereby enteric microbes interact during malnutrition, we used protein deficiency in mice to develop a new model of co-enteropathogen enteropathy. Focusing on common enteropathogens in malnourished children, Giardia lamblia and enteroaggregative Escherichia coli (EAEC), we provide new insights into intersecting pathogen-specific mechanisms that enhance malnutrition. We show for the first time that during protein malnutrition, the intestinal microbiota permits persistent Giardia colonization and simultaneously contributes to growth impairment. Despite signals of intestinal injury, such as IL1α, Giardia-infected mice lack pro-inflammatory intestinal responses, similar to endemic pediatric Giardia infections. Rather, Giardia perturbs microbial host co-metabolites of proteolysis during growth impairment, whereas host nicotinamide utilization adaptations that correspond with growth recovery increase. EAEC promotes intestinal inflammation and markers of myeloid cell activation. During co-infection, intestinal inflammatory signaling and cellular recruitment responses to EAEC are preserved together with a Giardia-mediated diminishment in myeloid cell activation. Conversely, EAEC extinguishes markers of host energy expenditure regulatory responses to Giardia, as host metabolic adaptations appear exhausted. Integrating immunologic and metabolic profiles during co-pathogen infection and malnutrition, we develop a working mechanistic model of how cumulative diet-induced and pathogen-triggered microbial perturbations result in an increasingly wasted host.


Subject(s)
Coinfection/microbiology , Coinfection/parasitology , Escherichia coli Infections/microbiology , Escherichia coli/physiology , Giardia lamblia/physiology , Giardiasis/parasitology , Malnutrition/microbiology , Malnutrition/parasitology , Animals , Child , Coinfection/immunology , Cytokines/immunology , Disease Models, Animal , Escherichia coli Infections/immunology , Giardiasis/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Male , Malnutrition/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology
7.
Am J Hum Genet ; 96(5): 797-807, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957468

ABSTRACT

High-throughput sequencing technology has enabled population-based studies of the role of the human microbiome in disease etiology and exposure response. Distance-based analysis is a popular strategy for evaluating the overall association between microbiome diversity and outcome, wherein the phylogenetic distance between individuals' microbiome profiles is computed and tested for association via permutation. Despite their practical popularity, distance-based approaches suffer from important challenges, especially in selecting the best distance and extending the methods to alternative outcomes, such as survival outcomes. We propose the microbiome regression-based kernel association test (MiRKAT), which directly regresses the outcome on the microbiome profiles via the semi-parametric kernel machine regression framework. MiRKAT allows for easy covariate adjustment and extension to alternative outcomes while non-parametrically modeling the microbiome through a kernel that incorporates phylogenetic distance. It uses a variance-component score statistic to test for the association with analytical p value calculation. The model also allows simultaneous examination of multiple distances, alleviating the problem of choosing the best distance. Our simulations demonstrated that MiRKAT provides correctly controlled type I error and adequate power in detecting overall association. "Optimal" MiRKAT, which considers multiple candidate distances, is robust in that it suffers from little power loss in comparison to when the best distance is used and can achieve tremendous power gain in comparison to when a poor distance is chosen. Finally, we applied MiRKAT to real microbiome datasets to show that microbial communities are associated with smoking and with fecal protease levels after confounders are controlled for.


Subject(s)
Genetics, Population , Microbiota/genetics , Models, Statistical , Computer Simulation , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Polymorphism, Single Nucleotide , Software
8.
J Pediatr Gastroenterol Nutr ; 67(4): 483-487, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29901551

ABSTRACT

Fecal microbiota transplantation (FMT) involves the transfer of stool from a healthy individual into the intestinal tract of a diseased recipient. Although used primarily for recurrent Clostridium difficile infection, FMT is increasingly being attempted as an experimental therapy for other illnesses, including metabolic disorders. D-lactic acidosis (D-LA) is a metabolic disorder that may occur in individuals with short bowel syndrome when lactate-producing bacteria in the colon overproduce D-lactate. This results in elevated systemic levels of D-lactate, metabolic acidosis, and encephalopathy. In this study, we report the successful use of FMT for the treatment of recurrent D-LA in a child who was unresponsive to conventional therapies. Importantly, we also present profiles of the enteric microbiota, as well as fecal D-/L-lactic acid metabolites, before and longitudinally after FMT. These data provide valuable insight into the putative mechanisms of D-LA pathogenesis and its treatment.


Subject(s)
Acidosis, Lactic/therapy , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome , Lactic Acid/blood , Short Bowel Syndrome/complications , Acidosis, Lactic/blood , Acidosis, Lactic/microbiology , Child , Female , Humans , Short Bowel Syndrome/blood , Short Bowel Syndrome/microbiology , Treatment Outcome
9.
Dig Dis Sci ; 63(7): 1890-1899, 2018 07.
Article in English | MEDLINE | ID: mdl-29777439

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) has been associated with changes in the intestinal microbiota. Only a few studies have explored differences in the mucosa-associated microbiota between IBS patients and healthy controls (HC). AIMS: To characterize and compare the microbiota in mucosal and fecal samples from carefully selected patients with IBS-D and HC. METHODS: The cohort was composed of 23 diarrhea-predominant IBS (IBS-D) patients and 24 HC. Fresh stool samples were collected from participants prior to the collection of colonic mucosal samples from an unprepped bowel. After DNA extraction, 16S rRNA genes were sequenced by 454 pyrosequencing and analyzed using the QIIME pipeline. RESULTS: The fecal microbiota (luminal niche) of IBS-D patients was found to have reduced enteric richness compared to HC (P < 0.05), whereas no differences were observed between the two groups within the mucosal microbiota. Within the luminal niche, the relative proportions of Faecalibacterium genus were found to be lower in IBS-D than in HC and the Dorea genus was higher in IBS-D. None of the taxa proportions were significantly different in IBS-D patients versus HC using an FDR of ≤ 0.1 when analyzing samples that appeared in > 25% samples of either niche. CONCLUSION: Fecal and mucosal microbiota of IBS-D patients and HC are very similar and are not sufficient to explain the reported altered physiology and symptomatology of IBS-D. Future studies should investigate intestinal microbiome-dependent functional activity in addition to the fecal and mucosal-associated microbial composition.


Subject(s)
Bacteria/isolation & purification , Diarrhea/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Intestines/microbiology , Irritable Bowel Syndrome/microbiology , Bacteria/classification , Bacteria/genetics , Case-Control Studies , Diarrhea/diagnosis , Humans , Irritable Bowel Syndrome/diagnosis , Ribotyping
10.
PLoS Pathog ; 11(6): e1004911, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26067254

ABSTRACT

The commensal Enterococcus faecalis is among the most common causes of nosocomial infections. Recent findings regarding increased abundance of enterococci in the intestinal microbiota of patients with inflammatory bowel diseases and induction of colitis in IL-10-deficient (IL-10-/-) mice put a new perspective on the contribution of E. faecalis to chronic intestinal inflammation. Based on the expression of virulence-related genes in the inflammatory milieu of IL-10-/- mice using RNA-sequencing analysis, we characterized the colitogenic role of two bacterial structures that substantially impact on E. faecalis virulence by different mechanisms: the enterococcal polysaccharide antigen and cell surface-associated lipoproteins. Germ-free wild type and IL-10-/- mice were monoassociated with E. faecalis wild type OG1RF or the respective isogenic mutants for 16 weeks. Intestinal tissue and mesenteric lymph nodes (MLN) were collected to characterize tissue pathology, loss of intestinal barrier function, bacterial adhesion to intestinal epithelium and immune cell activation. Bone marrow-derived dendritic cells (BMDC) were stimulated with bacterial lysates and E. faecalis virulence was additionally investigated in three invertebrate models. Colitogenic activity of wild type E. faecalis (OG1RF score: 7.2±1.2) in monoassociated IL-10-/- mice was partially impaired in E. faecalis lacking enterococcal polysaccharide antigen (ΔepaB score: 4.7±2.3; p<0.05) and was almost completely abrogated in E. faecalis deficient for lipoproteins (Δlgt score: 2.3±2.3; p<0.0001). Consistently both E. faecalis mutants showed significantly impaired virulence in Galleria mellonella and Caenorhabditis elegans. Loss of E-cadherin in the epithelium was shown for all bacterial strains in inflamed IL-10-/- but not wild type mice. Inactivation of epaB in E. faecalis reduced microcolony and biofilm formation in vitro, altered bacterial adhesion to intestinal epithelium of germ-free Manduca sexta larvae and impaired penetration into the colonic mucus layer of IL-10-/- mice. Lipoprotein-deficient E. faecalis exhibited an impaired TLR2-mediated activation of BMDCs in vitro despite their ability to fully reactivate MLN cells as well as MLN-derived colitogenic T cells ex vivo. E. faecalis virulence factors accounting for bacterial adhesion to mucosal surfaces as well as intestinal barrier disruption partially contribute to colitogenic activity of E. faecalis. Beyond their well-known role in infections, cell surface-associated lipoproteins are essential structures for colitogenic activity of E. faecalis by mediating innate immune cell activation.


Subject(s)
Colitis/microbiology , Enterococcus faecalis/immunology , Enterococcus faecalis/pathogenicity , Gram-Positive Bacterial Infections/metabolism , Interleukin-10/metabolism , Animals , Blotting, Western , Enterococcus faecalis/metabolism , Fluorescent Antibody Technique , Gram-Positive Bacterial Infections/immunology , In Situ Hybridization, Fluorescence , Interleukin-10/deficiency , Lipoproteins/metabolism , Mice , Mice, Knockout , Virulence
11.
Curr Psychiatry Rep ; 19(8): 51, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28676966

ABSTRACT

PURPOSE OF REVIEW: We reviewed and evaluated recently published scientific studies that explored the role of the intestinal microbiota in eating disorders. RECENT FINDINGS: Studies have demonstrated that the intestinal microbiota is a contributing factor to both host energy homeostasis and behavior-two traits commonly disrupted in patients with eating disorders. To date, intestinal microbiota research in eating disorders has focused solely on anorexia nervosa (AN). Initial studies have reported an atypical intestinal microbial composition in patients with AN compared to healthy controls. However, the impact of these AN-associated microbial communities on host metabolism and behavior remains unknown. The intriguing pattern of findings in patients with AN encourages further investigation of the intestinal microbiota in eating disorders. Elucidating the specific role(s) of these microbial communities may yield novel ideas for augmenting current clinical therapies to promote weight gain, decrease gastrointestinal distress, and even reduce psychological symptomatology.


Subject(s)
Feeding and Eating Disorders/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Feeding and Eating Disorders/psychology , Gastrointestinal Diseases/psychology , Homeostasis/physiology , Humans
12.
Eur Eat Disord Rev ; 25(5): 423-427, 2017 09.
Article in English | MEDLINE | ID: mdl-28586130

ABSTRACT

Anorexia nervosa, a severe psychiatric illness, is associated with an intestinal microbial dysbiosis. Individual microbial signatures dominate in healthy samples, even over time and under controlled conditions, but whether microbial markers of the disorder overcome inter-individual variation during the acute stage of illness or renourishment is unknown. We characterized daily changes in the intestinal microbiota in three acutely ill patients with anorexia nervosa over the entire course of hospital-based renourishment and found significant, patient-specific changes in microbial composition and diversity. This preliminary case series suggests that even in a state of pathology, individual microbial signatures persist in accounting for the majority of intestinal microbial variation. Copyright © 2017 John Wiley & Sons, Ltd and Eating Disorders Association.


Subject(s)
Anorexia Nervosa/microbiology , Gastrointestinal Microbiome , Adolescent , Adult , Anorexia Nervosa/diagnosis , Anorexia Nervosa/therapy , Female , Humans , Middle Aged , Young Adult
13.
Am J Physiol Gastrointest Liver Physiol ; 310(6): G417-26, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26702134

ABSTRACT

Recent studies have demonstrated differences in the intestinal microbiota between patients with irritable bowel syndrome (IBS) and healthy controls (HC), suggesting a role for the intestinal microbiota in the pathogenesis of IBS. Alterations in the microbiota have also been implicated in the pathogenesis of abdominal bloating, a commonly reported symptom in IBS. We investigated the relationship between the intestinal microbiota, abdominal bloating, and altered bowel patterns in a cohort of patients with IBS and HC. The 16S rRNA gene from fresh fecal samples was amplified and pyrosequenced by using Roche-454 Titanium chemistry. A Core Measurable Microbiome (CMM) was generated for Operational Taxonomic Unit (OTU) detected in >75% of all samples and compositional features of CMM were compared between groups by Linear Discriminant Analysis (LDA). IBS differentiated from HC by LDA using continuous variation in the species/OTUs or the CMM genera. When subcategorized based on bloating symptoms and bowel characteristics, the same subjects were also well differentiated from one another and from HC. ANOVA analysis showed quantitative species/OTU differences between the subgroups including IBS with and without bloating, and subtypes based on bowel characteristics. The clear LDA differentiation and the significant microbial taxa differences between the groups imply a significant association of the microbiota with bloating symptoms and bowel characteristics in IBS. These changes in the microbiota may serve as a biomarker for IBS and its clinical subtypes and suggest a role for the intestinal microbiota in the pathogenesis of the main symptoms of the disorder.


Subject(s)
Abdominal Cavity , Gastrointestinal Microbiome , Intestines/microbiology , Intestines/physiopathology , Adult , Cohort Studies , DNA, Bacterial/genetics , Dilatation, Pathologic , Feces/chemistry , Feces/microbiology , Female , Humans , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/physiopathology , Male , Microbiota , Middle Aged , RNA, Ribosomal, 16S/biosynthesis , RNA, Ribosomal, 16S/genetics , Young Adult
14.
Infect Immun ; 83(10): 4068-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216423

ABSTRACT

Adherent-invasive Escherichia coli (AIEC), a functionally distinct subset of resident intestinal E. coli associated with Crohn's disease, is characterized by enhanced epithelial adhesion and invasion, survival within macrophages, and biofilm formation. Environmental factors, such as iron, modulate E. coli production of extracellular structures, which in turn influence the formation of multicellular communities, such as biofilms, and bacterial interactions with host cells. However, the physiological and functional responses of AIEC to variable iron availability have not been thoroughly investigated. We therefore characterized the impact of iron on the physiology of AIEC strain NC101 and subsequent interactions with macrophages. Iron promoted the cellulose-dependent aggregation of NC101. Bacterial cells recovered from the aggregates were more susceptible to phagocytosis than planktonic cells, which corresponded with the decreased macrophage production of the proinflammatory cytokine interleukin-12 (IL-12) p40. Prevention of aggregate formation through the disruption of cellulose production reduced the phagocytosis of iron-exposed NC101. In contrast, under iron-limiting conditions, where NC101 aggregation is not induced, the disruption of cellulose production enhanced NC101 phagocytosis and decreased macrophage secretion of IL-12 p40. Finally, abrogation of cellulose production reduced NC101 induction of colitis when NC101 was monoassociated in inflammation-prone Il10(-/-) mice. Taken together, our results introduce cellulose as a novel physiological factor that impacts host-microbe-environment interactions and alters the proinflammatory potential of AIEC.


Subject(s)
Bacterial Adhesion , Cellulose/metabolism , Colitis/microbiology , Escherichia coli Infections/microbiology , Escherichia coli/physiology , Iron/metabolism , Phagocytosis , Animals , Colitis/immunology , Escherichia coli/genetics , Escherichia coli Infections/immunology , Female , Humans , Interleukin-12/immunology , Macrophages/immunology , Macrophages/microbiology , Male , Mice
15.
Infect Immun ; 83(7): 2762-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25916983

ABSTRACT

Microbial protease-mediated disruption of the intestinal epithelium is a potential mechanism whereby a dysbiotic enteric microbiota can lead to disease. This mechanism was investigated using the colitogenic, protease-secreting enteric microbe Enterococcus faecalis. Caco-2 and T-84 epithelial cell monolayers and the mouse colonic epithelium were exposed to concentrated conditioned media (CCM) from E. faecalis V583 and E. faecalis lacking the gelatinase gene (gelE). The flux of fluorescein isothiocyanate (FITC)-labeled dextran across monolayers or the mouse epithelium following exposure to CCM from parental or mutant E. faecalis strains indicated paracellular permeability. A protease-activated receptor 2 (PAR2) antagonist and PAR2-deficient (PAR2(-/-)) mice were used to investigate the role of this receptor in E. faecalis-induced permeability. Gelatinase (GelE) purified from E. faecalis V583 was used to confirm the ability of this protease to induce epithelial cell permeability and activate PAR2. The protease-mediated permeability of colonic epithelia from wild-type (WT) and PAR2(-/-) mice by fecal supernatants from ulcerative colitis patients was assessed. Secreted E. faecalis proteins induced permeability in epithelial cell monolayers, which was reduced in the absence of gelE or by blocking PAR2 activity. Secreted E. faecalis proteins induced permeability in the colonic epithelia of WT mice that was absent in tissues from PAR2(-/-) mice. Purified GelE confirmed the ability of this protease to induce epithelial cell permeability via PAR2 activation. Fecal supernatants from ulcerative colitis patients induced permeability in the colonic epithelia of WT mice that was reduced in tissues from PAR2(-/-) mice. Our investigations demonstrate that GelE from E. faecalis can regulate enteric epithelial permeability via PAR2.


Subject(s)
Enterococcus faecalis/enzymology , Gelatinases/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Permeability , Receptor, PAR-2/metabolism , Animals , Cell Line , Colon/microbiology , Colon/physiology , Culture Media, Conditioned , Epithelial Cells/microbiology , Epithelial Cells/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-2/deficiency
16.
Psychosom Med ; 77(9): 969-81, 2015.
Article in English | MEDLINE | ID: mdl-26428446

ABSTRACT

OBJECTIVE: The relevance of the microbe-gut-brain axis to psychopathology is of interest in anorexia nervosa (AN), as the intestinal microbiota plays a critical role in metabolic function and weight regulation. METHODS: We characterized the composition and diversity of the intestinal microbiota in AN, using stool samples collected at inpatient admission (T1; n = 16) and discharge (T2; n = 10). At T1, participants completed the Beck Depression and Anxiety Inventories and the Eating Disorder Examination-Questionnaire. Patients with AN were compared with healthy individuals who participated in a previous study (healthy comparison group; HCG). Genomic DNA was isolated from stool samples, and bacterial composition was characterized by 454 pyrosequencing of the 16S rRNA gene. Sequencing results were processed by the Quantitative Insights Into Microbial Ecology pipeline. We compared T1 versus T2 samples, samples from both points were compared with HCG (n = 12), and associations between psychopathology and T1 samples were explored. RESULTS: In patients with AN, significant changes emerged between T1 and T2 in taxa abundance and beta (between-sample) diversity. Patients with AN had significantly lower alpha (within-sample) diversity than did HCG at both T1 (p = .0001) and T2 (p = .016), and differences in taxa abundance were found between AN patients and HCG. Levels of depression, anxiety, and eating disorder psychopathology at T1 were associated with composition and diversity of the intestinal microbiota. CONCLUSIONS: We provide evidence of an intestinal dysbiosis in AN and an association between mood and the enteric microbiota in this patient population. Future directions include mechanistic investigations of the microbe-gut-brain axis in animal models and association of microbial measures with metabolic changes and recovery indices.


Subject(s)
Anorexia Nervosa/microbiology , Gastrointestinal Microbiome/physiology , Adolescent , Adult , Affect , Anorexia Nervosa/psychology , Anorexia Nervosa/therapy , Anxiety/microbiology , Bacteroidetes/isolation & purification , Body Composition , Case-Control Studies , Convalescence , DNA, Bacterial/genetics , Depression/microbiology , Feces/microbiology , Feeding Behavior , Female , Firmicutes/isolation & purification , Humans , Lactobacillus/isolation & purification , Methanobrevibacter/isolation & purification , Ribotyping , Ruminococcus/isolation & purification , Surveys and Questionnaires , Young Adult
17.
Microbiol Immunol ; 59(8): 452-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26146866

ABSTRACT

Induction of mammalian heme oxygenase (HO)-1 and exposure of animals to carbon monoxide (CO) ameliorates experimental colitis. When enteric bacteria, including Escherichia coli, are exposed to low iron conditions, they express an HO-like enzyme, chuS, and metabolize heme into iron, biliverdin and CO. Given the abundance of enteric bacteria residing in the intestinal lumen, our postulate was that commensal intestinal bacteria may be a significant source of CO and those that express chuS and other Ho-like molecules suppress inflammatory immune responses through release of CO. According to real-time PCR, exposure of mice to CO results in changes in enteric bacterial composition and increases E. coli 16S and chuS DNA. Moreover, the severity of experimental colitis correlates positively with E. coli chuS expression in IL-10 deficient mice. To explore functional roles, E. coli were genetically modified to overexpress chuS or the chuS gene was deleted. Co-culture of chuS-overexpressing E. coli with bone marrow-derived macrophages resulted in less IL-12p40 and greater IL-10 secretion than in wild-type or chuS-deficient E. coli. Mice infected with chuS-overexpressing E. coli have more hepatic CO and less serum IL-12 p40 than mice infected with chuS-deficient E. coli. Thus, CO alters the composition of the commensal intestinal microbiota and expands populations of E. coli that harbor the chuS gene. These bacteria are capable of attenuating innate immune responses through expression of chuS. Bacterial HO-like molecules and bacteria-derived CO may represent novel targets for therapeutic intervention in inflammatory conditions.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/immunology , Heme Oxygenase (Decyclizing)/immunology , Heme Oxygenase (Decyclizing)/metabolism , Immune Evasion , Immunity, Innate , Animals , Carbon Monoxide/metabolism , Cells, Cultured , Coculture Techniques , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Escherichia coli/metabolism , Gene Deletion , Gene Expression , Heme Oxygenase (Decyclizing)/genetics , Interleukin-10/metabolism , Interleukin-12 Subunit p40/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics
18.
Eur Eat Disord Rev ; 23(6): 496-503, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26426680

ABSTRACT

With nearly 39% of the worldwide adult population classified as obese, much of the globe is facing a serious public health challenge. Increasing rates of obesity, coupled with the failure of many behavioural and pharmacological interventions, have contributed to a rise in popularity of bariatric surgery as a treatment for obesity. Surgery-mediated weight loss was initially thought to be a direct result of mechanical alterations causing restriction and calorie malabsorption. However, the mounting evidence suggests that indirect factors influence the accumulation and storage of fat in patients that have undergone this procedure. Given the established impact the intestinal microbiota has on adiposity, it is likely that this complex enteric microbial community contributes to surgery-mediated weight loss and maintenance of weight loss postsurgery. In this review, we discuss the physiological and psychological traits exhibited by bariatric surgery candidates that can be influenced by the intestinal microbiota. Additionally, we detail the studies that investigated the impact of bariatric surgery on the intestinal microbiota in humans and mouse models of this procedure.


Subject(s)
Bariatric Surgery , Gastrointestinal Microbiome/physiology , Obesity/surgery , Weight Loss/physiology , Animals , Bariatric Surgery/psychology , Humans , Mice , Models, Animal , Treatment Outcome
19.
Gut ; 63(6): 903-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23512834

ABSTRACT

OBJECTIVE: Although polymorphisms of the NOD2 gene predispose to the development of ileal Crohn's disease, the precise mechanisms of this increased susceptibility remain unclear. Previous work has shown that transcript expression of the Paneth cell (PC) antimicrobial peptides (AMPs) α-defensin 4 and α-defensin-related sequence 10 are selectively decreased in Nod2(-/-) mice. However, the specific mouse background used in this previous study is unclear. In light of recent evidence suggesting that mouse strain strongly influences PC antimicrobial activity, we sought to characterise PC AMP function in commercially available Nod2(-/-) mice on a C57BL/6 (B6) background. Specifically, we hypothesised that Nod2(-/-) B6 mice would display reduced AMP expression and activity. DESIGN: Wild-type (WT) and Nod2(-/-) B6 ileal AMP expression was assessed via real-time PCR, acid urea polyacrylamide gel electrophoresis and mass spectrometry. PCs were enumerated using flow cytometry. Functionally, α-defensin bactericidal activity was evaluated using a gel-overlay antimicrobial assay. Faecal microbial composition was determined using 454-sequencing of the bacterial 16S gene in cohoused WT and Nod2(-/-) littermates. RESULTS: WT and Nod2(-/-) B6 mice displayed similar PC AMP expression patterns, equivalent α-defensin profiles, and identical antimicrobial activity against commensal and pathogenic bacterial strains. Furthermore, minimal differences in gut microbial composition were detected between the two cohoused, littermate mouse groups. CONCLUSIONS: Our data reveal that Nod2 does not directly regulate PC antimicrobial activity in B6 mice. Moreover, we demonstrate that previously reported Nod2-dependent influences on gut microbial composition may be overcome by environmental factors, such as cohousing with WT littermates.


Subject(s)
Feces/microbiology , Ileum/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Paneth Cells/metabolism , RNA, Messenger/metabolism , alpha-Defensins/metabolism , Animals , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Defensins/genetics , Defensins/metabolism , Escherichia coli/drug effects , Ileum/cytology , Lectins, C-Type/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microbial Sensitivity Tests , Muramidase/metabolism , Nod2 Signaling Adaptor Protein/genetics , Pancreatitis-Associated Proteins , Paneth Cells/cytology , Peptides/genetics , Peptides/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Salmonella enterica/drug effects , Transcription, Genetic , alpha-Defensins/genetics , alpha-Defensins/pharmacology
20.
Infect Immun ; 82(6): 2239-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24643531

ABSTRACT

We evaluated whether a simplified human microbiota consortium (SIHUMI) induces colitis in germfree (GF) 129S6/SvEv (129) and C57BL/6 (B6) interleukin-10-deficient (IL-10(-/-)) mice, determined mouse strain effects on colitis and the microbiota, examined the effects of inflammation on relative bacterial composition, and identified immunodominant bacterial species in "humanized" IL-10(-/-) mice. GF wild-type (WT) and IL-10(-/-) 129 and B6 mice were colonized with 7 human-derived inflammatory bowel disease (IBD)-related intestinal bacteria and maintained under gnotobiotic conditions. Quantification of bacteria in feces, ileal and colonic contents, and tissues was performed using 16S rRNA gene selective quantitative PCR. Colonic segments were scored histologically, and gamma interferon (IFN-γ), IL-12p40, and IL-17 levels were measured in supernatants of unstimulated colonic tissue explants and of mesenteric lymph node (MLN) cells stimulated by lysates of individual or aggregate bacterial strains. Relative bacterial species abundances changed over time and differed between 129 and B6 mice, WT and IL-10(-/-) mice, luminal and mucosal samples, and ileal and colonic or fecal samples. SIHUMI induced colitis in all IL-10(-/-) mice, with more aggressive colitis and MLN cell activation in 129 mice. Escherichia coli LF82 and Ruminococcus gnavus lysates induced dominant effector ex vivo MLN TH1 and TH17 responses, although the bacterial mucosal concentrations were low. In summary, this study shows that a simplified human bacterial consortium induces colitis in ex-GF 129 and B6 IL-10(-/-) mice. Relative concentrations of individual SIHUMI species are determined by host genotype, the presence of inflammation, and anatomical location. A subset of IBD-relevant human enteric bacterial species preferentially stimulates bacterial antigen-specific TH1 and TH17 immune responses in this model, independent of luminal and mucosal bacterial concentrations.


Subject(s)
Colitis/microbiology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae , Interleukin-10/deficiency , Microbiota , Animals , Cells, Cultured , Colitis/immunology , Colon/metabolism , Colon/microbiology , Colony Count, Microbial , Cytokines/metabolism , Enterobacteriaceae/growth & development , Enterobacteriaceae/immunology , Enterobacteriaceae Infections/immunology , Feces/microbiology , Gastric Mucosa/microbiology , Germ-Free Life , Humans , Ileum/microbiology , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL