Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiome ; 11(1): 66, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37004103

ABSTRACT

BACKGROUND: Crohn's disease (CD) is associated with changes in the microbiota, and murine models of CD-like ileo-colonic inflammation depend on the presence of microbial triggers. Increased abundance of unknown Clostridiales and the microscopic detection of filamentous structures close to the epithelium of Tnf ΔARE mice, a mouse model of CD-like ileitis pointed towards segmented filamentous bacteria (SFB), a commensal mucosal adherent bacterium involved in ileal inflammation. RESULTS: We show that the abundance of SFB strongly correlates with the severity of CD-like ileal inflammation in two mouse models of ileal inflammation, including Tnf ΔARE and SAMP/Yit mice. SFB mono-colonization of germ-free Tnf ΔARE mice confirmed the causal link and resulted in severe ileo-colonic inflammation, characterized by elevated tissue levels of Tnf and Il-17A, neutrophil infiltration and loss of Paneth and goblet cell function. Co-colonization of SFB in human-microbiota associated Tnf ΔARE mice confirmed that SFB presence is indispensable for disease development. Screening of 468 ileal and colonic mucosal biopsies from adult and pediatric IBD patients, using previously published and newly designed human SFB-specific primer sets, showed no presence of SFB in human tissue samples, suggesting a species-specific functionality of the pathobiont. Simulating the human relevant therapeutic effect of exclusive enteral nutrition (EEN), EEN-like purified diet antagonized SFB colonization and prevented disease development in Tnf ΔARE mice, providing functional evidence for the protective mechanism of diet in modulating microbiota-dependent inflammation in IBD. CONCLUSIONS: We identified a novel pathogenic role of SFB in driving severe CD-like ileo-colonic inflammation characterized by loss of Paneth and goblet cell functions in Tnf ΔARE mice. A purified diet antagonized SFB colonization and prevented disease development in Tnf ΔARE mice in contrast to a fiber-containing chow diet, clearly demonstrating the important role of diet in modulating a novel IBD-relevant pathobiont and supporting a direct link between diet and microbial communities in mediating protective functions. Video Abstract.


Subject(s)
Crohn Disease , Ileitis , Adult , Humans , Mice , Animals , Child , Crohn Disease/microbiology , Inflammation , Ileitis/microbiology , Ileitis/pathology , Diet , Bacteria/genetics , Disease Models, Animal
2.
PLoS One ; 17(10): e0276195, 2022.
Article in English | MEDLINE | ID: mdl-36301950

ABSTRACT

Ulcerative colitis and Crohn's disease are chronic inflammatory bowel diseases (IBD) of unknown cause characterized by a relapsing-remitting behavior. Growing evidence supports the idea that the epithelial barrier plays a central role in the pathogenesis of IBD as well as in its evolution over time, thus representing a potential target for novel therapeutic options. In the last decade, the introduction of 3D epithelial cultures from ex vivo-expanded intestinal adult stem cells (ASCs) has impacted our ability to study the function of the epithelium in several gastrointestinal disorders, including IBD. Here, we describe in detail a reproducible protocol to generate Matrigel-embedded epithelial organoids from ASCs of non-IBD and IBD donors using small colonic biopsies, including steps for its optimization. A slightly modified version of this protocol is also provided in case surgical samples are used. With this method, epithelial organoids can be expanded over several passages, thereby generating a large quantity of viable cells that can be used in multiple downstream analyses including genetic, transcriptional, proteomic and/or functional studies. In addition, 3D cultures generated using our protocol are suitable for the establishment of 2D cultures, which can model relevant cell-to-cell interactions that occur in IBD mucosa.


Subject(s)
Colitis, Ulcerative , Inflammatory Bowel Diseases , Adult , Humans , Organoids/pathology , Intestinal Mucosa/pathology , Proteomics , Inflammatory Bowel Diseases/pathology , Colon/pathology , Colitis, Ulcerative/pathology
3.
Front Immunol ; 12: 646906, 2021.
Article in English | MEDLINE | ID: mdl-33854511

ABSTRACT

Over the last decades, Adherent-Invasive Escherichia coli (AIEC) has been linked to the pathogenesis of Crohn's Disease. AIEC's characteristics, as well as its interaction with the gut immune system and its role in intestinal epithelial barrier dysfunction, have been extensively studied. Nevertheless, the currently available techniques to investigate the cross-talk between this pathogen and intestinal epithelial cells (IECs) are based on the infection of immortalized cell lines. Despite their many advantages, cell lines cannot reproduce the conditions in tissues, nor do they reflect interindividual variability or gut location-specific traits. In that sense, the use of human primary cultures, either healthy or diseased, offers a system that can overcome all of these limitations. Here, we developed a new infection model by using freshly isolated human IECs. For the first time, we generated and infected monolayer cultures derived from human colonic organoids to study the mechanisms and effects of AIEC adherence and invasion on primary human epithelial cells. To establish the optimal conditions for AIEC invasion studies in human primary organoid-derived epithelial monolayers, we designed an infection-kinetics study to assess the infection dynamics at different time points, as well as with two multiplicities of infection (MOI). Overall, this method provides a model for the study of host response to AIEC infections, as well as for the understanding of the molecular mechanisms involved in adhesion, invasion and intracellular replication. Therefore, it represents a promising tool for elucidating the cross-talk between AIEC and the intestinal epithelium in healthy and diseased tissues.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli Infections/metabolism , Intestinal Mucosa/metabolism , Organoids/metabolism , Algorithms , Bacterial Adhesion , Cell Culture Techniques/methods , Crohn Disease/metabolism , Crohn Disease/microbiology , Epithelial Cells/microbiology , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Models, Biological , Organoids/cytology , Organoids/microbiology , Reproducibility of Results
4.
PLoS One ; 16(2): e0246367, 2021.
Article in English | MEDLINE | ID: mdl-33556098

ABSTRACT

BACKGROUND: Personalized medicine requires finding relationships between variables that influence a patient's phenotype and predicting an outcome. Sparse generalized canonical correlation analysis identifies relationships between different groups of variables. This method requires establishing a model of the expected interaction between those variables. Describing these interactions is challenging when the relationship is unknown or when there is no pre-established hypothesis. Thus, our aim was to develop a method to find the relationships between microbiome and host transcriptome data and the relevant clinical variables in a complex disease, such as Crohn's disease. RESULTS: We present here a method to identify interactions based on canonical correlation analysis. We show that the model is the most important factor to identify relationships between blocks using a dataset of Crohn's disease patients with longitudinal sampling. First the analysis was tested in two previously published datasets: a glioma and a Crohn's disease and ulcerative colitis dataset where we describe how to select the optimum parameters. Using such parameters, we analyzed our Crohn's disease data set. We selected the model with the highest inner average variance explained to identify relationships between transcriptome, gut microbiome and clinically relevant variables. Adding the clinically relevant variables improved the average variance explained by the model compared to multiple co-inertia analysis. CONCLUSIONS: The methodology described herein provides a general framework for identifying interactions between sets of omic data and clinically relevant variables. Following this method, we found genes and microorganisms that were related to each other independently of the model, while others were specific to the model used. Thus, model selection proved crucial to finding the existing relationships in multi-omics datasets.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Transcriptome , Adolescent , Adult , Datasets as Topic , Female , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Male , Multivariate Analysis , Young Adult
5.
Mol Nutr Food Res ; 65(5): e2000188, 2021 03.
Article in English | MEDLINE | ID: mdl-32992407

ABSTRACT

Chronic inflammatory disorders are rising worldwide. The implication of the microbiota in persistent inflammation has been studied for years, but a direct causal relationship has not yet been stablished. Intestinal epithelial cells (IECs) form a protective barrier against detrimental luminal components. Indeed, a decrease in epithelial integrity may trigger a severe inflammatory reaction due to the infiltration of potentially harmful molecules and microorganisms. Bacterial imbalance, more commonly known as dysbiosis, occurs during inflammation and several strategies have been proposed to counteract this condition. Probiotics have been widely used to positively alter the inherited microbial composition and recover a eubiotic status. Nevertheless, probiotics are thought to impair the return of the indigenous microbiome, and to aggravate inflammation in compromised patients. In contrast, postbiotics-bacterial-free metabolites secreted by probiotic strains-have been proposed as a better and safer strategy. Recent scientific studies that have demonstrated the immunomodulatory properties and epithelial protection of postbiotics are summarized in this review, with an emphasis on the available methods that are currently in use to better understand the role of postbiotics in health and nutrition.


Subject(s)
Gastrointestinal Microbiome/physiology , Intestines/physiology , Probiotics/pharmacology , Animals , Dysbiosis/metabolism , Dysbiosis/microbiology , Epithelial Cells/metabolism , Fatty Acids, Volatile/metabolism , HT29 Cells , Humans , Indoles/metabolism , Indoles/pharmacokinetics , Intestines/microbiology , Polyamines/metabolism , Polysaccharides, Bacterial/metabolism , Vitamins/pharmacokinetics
6.
Nat Commun ; 11(1): 4322, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859898

ABSTRACT

Gut microbial and metabolite alterations have been linked to the pathogenesis of inflammatory bowel diseases. Here we perform a multi-omics microbiome and metabolite analysis of a longitudinal cohort of Crohn's disease patients undergoing autologous hematopoietic stem cell transplantation, and investigational therapy that induces drug free remission in a subset of patients. Via comparison of patients who responded and maintained remission, responded but experienced disease relapse and patients who did not respond to therapy, we identify shared functional signatures that correlate with disease activity despite the variability of gut microbiota profiles at taxonomic level. These signatures reflect the disease state when transferred to gnotobiotic mice. Taken together, the integration of microbiome and metabolite profiles from human cohort and mice improves the predictive modelling of disease outcome, and allows the identification of a network of bacteria-metabolite interactions involving sulfur metabolism as a key mechanism linked to disease activity in Crohn's disease.


Subject(s)
Crohn Disease/metabolism , Crohn Disease/microbiology , Gastrointestinal Microbiome/physiology , Sulfur/metabolism , Adolescent , Adult , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Crohn Disease/drug therapy , Disease Models, Animal , Feces/microbiology , Female , Hematopoietic Stem Cell Transplantation , Humans , Interleukin-10/genetics , Male , Metagenome , Mice , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Remission Induction , Young Adult
7.
Inflamm Bowel Dis ; 26(1): 43-55, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31211831

ABSTRACT

BACKGROUND: Butyrate-producing gut bacteria are reduced in patients with active inflammatory bowel disease (IBD), supporting the hypothesis that butyrate supplementation may be beneficial in this setting. Nonetheless, earlier studies suggest that the oxidation of butyrate in IBD patients is altered. We propose that inflammation may decrease epithelial butyrate consumption. METHODS: Non-IBD controls and IBD patients were recruited for the study. Stool samples were used for short-chain fatty acid and bacterial butyryl CoA:acetate CoA-transferase quantification. Colonic biopsies and ex vivo differentiated epithelial organoids (d-EpOCs) treated with butyrate and/or tumor necrosis factor alpha (TNFα) were used for analyzing the expression of transporters MCT1 and ABCG2, metabolic enzyme ACADS, and butyrate receptor GPR43, and for butyrate metabolism and consumption assays. RESULTS: We observed that lower stool content of butyrate-producing bacteria in active IBD patients did not correlate with decreased butyrate concentrations. Indeed, the intestinal epithelial expression of MCT1, ABCG2, ACADS, and GPR43 was altered in active IBD patients. Nonetheless, d-EpOCs derived from IBD patients showed SLC16A1 (gene encoding for MCT1 protein), ABCG2, ACADS, and GPR43 expression levels comparable to controls. Moreover, IBD- and non-IBD-derived d-EpOCs responded similarly to butyrate, as assessed by transcriptional regulation. TNFα significantly altered SLC16A1, ABCG2, and GPR43 transcription in d-EpOCs, mimicking the expression profile observed in biopsies from active IBD patients and resulting in reduced butyrate consumption. CONCLUSIONS: We provide evidence that the response to butyrate is not intrinsically altered in IBD patients. However, TNFα renders the epithelium less responsive to this metabolite, defeating the purpose of butyrate supplementation during active inflammation.


Subject(s)
Butyrates/metabolism , Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Case-Control Studies , Cell Culture Techniques , Colitis, Ulcerative/microbiology , Crohn Disease/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Inflammation , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/pathology , Intestines/pathology , Male , Middle Aged , Organoids/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Crohns Colitis ; 13(5): 634-647, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-30521002

ABSTRACT

BACKGROUND AND AIMS: Recent studies have shown the efficacy of autologous haematopoietic stem cell transplantation [HSCT] in severely refractory Crohn's disease [CD] patients. HSCT is thought to eliminate auto-reactive cells; however, no specific studies of immune reconstitution in CD patients are available. METHODS: We followed a group of CD patients [n = 18] receiving autologous HSCT, with 50% of them achieving endoscopic drug-free remission. To elucidate the mechanisms driving efficacy, we monitored changes after HSCT in blood and intestine immune-cell composition. CD patients [n = 22] receiving anti-tumour necrosis factor [TNF]-α were included for comparison. RESULTS: Severe immune ablation followed by HSCT induced dramatic changes in both peripheral blood T and B cells in all patients regardless of the efficacy of the treatment. Endoscopic remission at week 52 following HSCT was associated with significant intestinal transcriptional changes. A comparison of the remission signature with that of anti-TNFα identified both common and unique genes in the HSCT-induced response. Based on deconvolution analysis of intestinal biopsy transcriptome data, we show that response to HSCT, but not to anti-TNFα, is associated with an expansion of naïve B-cells, as seen in blood, and a decrease in the memory resting T-cell content. As expected, endoscopic remission, in response to both HSCT and anti-TNFα, led to a significant reduction in intestinal neutrophil and M1 macrophage content. CONCLUSIONS: Peripheral blood immune remodelling after HSCT does not predict efficacy. In contrast, a profound intestinal T-cell depletion that is maintained long after transplant is associated with mucosal healing following HSCT, but not anti-TNFα.


Subject(s)
Crohn Disease/therapy , Hematopoietic Stem Cell Transplantation , Lymphocyte Count , Adult , B-Lymphocytes , Crohn Disease/immunology , Female , Humans , Macrophages , Male , Neutrophils , T-Lymphocytes , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...