Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Environ Health Perspect ; 132(2): 27007, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38380914

ABSTRACT

BACKGROUND: The increasing prevalence of food sensitivities has been attributed to changes in gut microenvironment; however, ubiquitous environmental triggers such as inorganic nanoparticles (NPs) used as food additives have not been thoroughly investigated. OBJECTIVES: We explored the impact of the NP-structured food-grade silicon dioxide (fg-SiO2) on intestinal immune response involved in oral tolerance (OT) induction and evaluated the consequences of oral chronic exposure to this food-additive using a mouse model of OT to ovalbumin (OVA) and on gluten immunopathology in mice expressing the celiac disease risk gene, HLA-DQ8. METHODS: Viability, proliferation, and cytokine production of mesenteric lymph node (MLN) cells were evaluated after exposure to fg-SiO2. C57BL/6J mice and a mouse model of OT to OVA were orally exposed to fg-SiO2 or vehicle for 60 d. Fecal lipocalin-2 (Lcn-2), anti-OVA IgG, cytokine production, and immune cell populations were analyzed. Nonobese diabetic (NOD) mice expressing HLA-DQ8 (NOD/DQ8), exposed to fg-SiO2 or vehicle, were immunized with gluten and immunopathology was investigated. RESULTS: MLN cells exposed to fg-SiO2 presented less proliferative T cells and lower secretion of interleukin 10 (IL-10) and transforming growth factor beta (TGF-ß) by T regulatory and CD45+ CD11b+ CD103+ cells compared to control, two factors mediating OT. Mice given fg-SiO2 exhibited intestinal Lcn-2 level and interferon gamma (IFN-γ) secretion, showing inflammation and less production of IL-10 and TGF-ß. These effects were also observed in OVA-tolerized mice exposed to fg-SiO2, in addition to a breakdown of OT and a lower intestinal frequency of T cells. In NOD/DQ8 mice immunized with gluten, the villus-to-crypt ratio was decreased while the CD3+ intraepithelial lymphocyte counts and the Th1 inflammatory response were aggravated after fg-SiO2 treatment. DISCUSSION: Our results suggest that chronic oral exposure to fg-SiO2 blocked oral tolerance induction to OVA, and worsened gluten-induced immunopathology in NOD/DQ8 mice. The results should prompt investigation on the link between SiO2 exposure and food sensitivities in humans. https://doi.org/10.1289/EHP12758.


Subject(s)
Interleukin-10 , Silicon Dioxide , Humans , Animals , Mice , Interleukin-10/pharmacology , Silicon Dioxide/toxicity , Food Additives/pharmacology , Mice, Inbred C57BL , Immune Tolerance/genetics , Glutens/pharmacology , Ovalbumin/pharmacology , Administration, Oral , Mice, Inbred BALB C
2.
Gastroenterology ; 167(1): 34-50, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38286392

ABSTRACT

Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Severity of Illness Index , Celiac Disease/immunology , Celiac Disease/microbiology , Celiac Disease/genetics , Humans , Animals , Risk Factors , Genetic Predisposition to Disease , Glutens/immunology , Glutens/adverse effects , Diet/adverse effects , Disease Models, Animal
3.
Science ; 381(6663): 1153-1154, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37708269

ABSTRACT

Immunomodulation of lymphocytes by intestinal epithelial cells could lead to new therapies.


Subject(s)
Inflammatory Bowel Diseases , Humans , Epithelial Cells/immunology , Immunomodulation , Inflammatory Bowel Diseases/immunology
4.
Methods Cell Biol ; 179: 77-101, 2023.
Article in English | MEDLINE | ID: mdl-37625882

ABSTRACT

Celiac disease (CeD) is a common immune-mediated disease triggered by the ingestion of gluten in genetically predisposed individuals. CeD is unique in that the trigger (gluten), necessary genes (HLA-DQ2 and DQ8), and the autoantigen (tissue transglutaminase) have been identified, allowing additional environmental co-factors, like the intestinal microbiota, to be studied through relevant in vivo models. Murine models for CeD have come a long way in the past decade and there are now in vitro and in vivo tools available that mimic certain aspects of clinical disease. These models, many of which express the CeD risk genes, have recently been used to study the mechanisms through which the microbiota play a role in CeD pathogenesis through a gnotobiotic approach. Historically, the generation of gnotobiology technology in mid-20th century allowed for the study of immunity and physiology under a complete absence of microbes (axenic) or known colonized status (gnotobiotic). This enabled understanding of mechanisms by which certain bacteria contribute to health and disease. With this perspective, here, we will discuss the various murine models currently being used to study CeD. We will then describe how utilizing axenic and gnotobiotic CeD models has increased our understanding of how microbes influence relevant steps of CeD pathogenesis, and explain key methodology involved in axenic and gnotobiotic modeling.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Humans , Animals , Mice , Celiac Disease/genetics , Glutens
5.
Immunity ; 56(8): 1862-1875.e9, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37478853

ABSTRACT

Loss of oral tolerance (LOT) to gluten, driven by dendritic cell (DC) priming of gluten-specific T helper 1 (Th1) cell immune responses, is a hallmark of celiac disease (CeD) and can be triggered by enteric viral infections. Whether certain commensals can moderate virus-mediated LOT remains elusive. Here, using a mouse model of virus-mediated LOT, we discovered that the gut-colonizing protist Tritrichomonas (T.) arnold promotes oral tolerance and protects against reovirus- and murine norovirus-mediated LOT, independent of the microbiota. Protection was not attributable to antiviral host responses or T. arnold-mediated innate type 2 immunity. Mechanistically, T. arnold directly restrained the proinflammatory program in dietary antigen-presenting DCs, subsequently limiting Th1 and promoting regulatory T cell responses. Finally, analysis of fecal microbiomes showed that T. arnold-related Parabasalid strains are underrepresented in human CeD patients. Altogether, these findings will motivate further exploration of oral-tolerance-promoting protists in CeD and other immune-mediated food sensitivities.


Subject(s)
Antigens , Immunity, Innate , Animals , Mice , Humans , Diet , Glutens , Dendritic Cells , Immune Tolerance
6.
Gut Microbes ; 15(1): 2188874, 2023.
Article in English | MEDLINE | ID: mdl-36939195

ABSTRACT

Abdominal pain is common in patients with gastrointestinal disorders, but its pathophysiology is unclear, in part due to poor understanding of basic mechanisms underlying visceral sensitivity. Accumulating evidence suggests that gut microbiota is an important determinant of visceral sensitivity. Clinical and basic research studies also show that sex plays a role in pain perception, although the precise pathways are not elucidated. We investigated pain responses in germ-free and conventionally raised mice of both sexes, and assessed visceral sensitivity to colorectal distension, neuronal excitability of dorsal root ganglia (DRG) neurons and the production of substance P and calcitonin gene-related peptide (CGRP) in response to capsaicin or a mixture of G-protein coupled receptor (GPCR) agonists. Germ-free mice displayed greater in vivo responses to colonic distention than conventional mice, with no differences between males and females. Pretreatment with intracolonic capsaicin or GPCR agonists increased responses in conventional, but not in germ-free mice. In DRG neurons, gut microbiota and sex had no effect on neuronal activation by capsaicin or GPCR agonists. While stimulated production of substance P by DRG neurons was similar in germ-free and conventional mice, with no additional effect of sex, the CGRP production was higher in germ-free mice, mainly in females. Absence of gut microbiota increases visceral sensitivity to colorectal distention in both male and female mice. This is, at least in part, due to increased production of CGRP by DRG neurons, which is mainly evident in female mice. However, central mechanisms are also likely involved in this process.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Animals , Female , Male , Mice , Calcitonin Gene-Related Peptide/analysis , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Substance P/analysis , Substance P/metabolism
8.
Gastroenterology ; 163(5): 1351-1363.e15, 2022 11.
Article in English | MEDLINE | ID: mdl-35810781

ABSTRACT

BACKGROUND & AIMS: Genes and gluten are necessary but insufficient to cause celiac disease (CeD). Altered gut microbiota has been implicated as an additional risk factor. Variability in sampling site may confound interpretation and mechanistic insight, as CeD primarily affects the small intestine. Thus, we characterized CeD microbiota along the duodenum and in feces and verified functional impact in gnotobiotic mice. METHODS: We used 16S rRNA gene sequencing (Illumina) and predicted gene function (PICRUSt2) in duodenal biopsies (D1, D2 and D3), aspirates, and stool from patients with active CeD and controls. CeD alleles were determined in consented participants. A subset of duodenal samples stratified according to similar CeD risk genotypes (controls DQ2-/- or DQ2+/- and CeD DQ2+/-) were used for further analysis and to colonize germ-free mice for gluten metabolism studies. RESULTS: Microbiota composition and predicted function in CeD was largely determined by intestinal location. In the duodenum, but not stool, there was higher abundance of Escherichia coli (D1), Prevotella salivae (D2), and Neisseria (D3) in CeD vs controls. Predicted bacterial protease and peptidase genes were altered in CeD and impaired gluten degradation was detected only in mice colonized with CeD microbiota. CONCLUSIONS: Our results showed luminal and mucosal microbial niches along the gut in CeD. We identified novel microbial proteolytic pathways involved in gluten detoxification that are impaired in CeD but not in controls carrying DQ2, suggesting an association with active duodenal inflammation. Sampling site should be considered a confounding factor in microbiome studies in CeD.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Mice , Animals , Celiac Disease/complications , RNA, Ribosomal, 16S/genetics , Glutens/metabolism , Peptide Hydrolases
9.
Mucosal Immunol ; 15(2): 235-243, 2022 02.
Article in English | MEDLINE | ID: mdl-35031683

ABSTRACT

Celiac disease (CeD) is an immune-mediated disease, triggered by gluten ingestion, in genetically susceptible individuals. The gluten-free diet (GFD) is the only current treatment for CeD, but is difficult to follow, has high non-adherence rates, and does not always lead to symptomatic or mucosal remission. Microbially-mediated mechanisms have been proposed to contribute to disease pathogenesis, and clinical studies support an association, but mechanistic insight has been difficult to obtain. Recent advances using translational approaches have provided clues to the mechanisms through which bacteria could contribute to CeD pathogenesis. In this review we discuss these bacterially mediated mechanisms, which include the modulation of pathogenic or protective pathways. Targeting these pathways through microbial therapeutics could provide adjuvant therapies to the GFD.


Subject(s)
Celiac Disease , Bacteria , Celiac Disease/therapy , Diet, Gluten-Free , Genetic Predisposition to Disease , Glutens , Humans
11.
Am J Gastroenterol ; 116(5): 943-945, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33767100

ABSTRACT

ABSTRACT: Nonceliac gluten sensitivity, or the more preferred term, nonceliac wheat sensitivity (NCWS), is a heterogenous condition that is diagnosed purely on the basis of symptoms and without an understanding of disease mechanisms and triggers. Biomarkers to identify patients and implementation of dietary treatment in a personalized manner are needed. Mansueto et al. identified a population of NCWS patients with associated autoimmune markers and immune activation. The presence of these markers could be used, in combination with other serological tests, to help develop better diagnostic strategies for NCWS.


Subject(s)
Wheat Hypersensitivity , Autoimmunity , Biomarkers , Humans , Wheat Hypersensitivity/diagnosis
12.
Gastroenterology ; 160(5): 1532-1545, 2021 04.
Article in English | MEDLINE | ID: mdl-33310084

ABSTRACT

BACKGROUND & AIMS: Altered gut microbiota composition and function have been associated with inflammatory bowel diseases, including ulcerative colitis (UC), but the causality and mechanisms remain unknown. METHODS: We applied 16S ribosomal RNA gene sequencing, shotgun metagenomic sequencing, in vitro functional assays, and gnotobiotic colonizations to define the microbial composition and function in fecal samples obtained from a cohort of healthy individuals at risk for inflammatory bowel diseases (pre-UC) who later developed UC (post-UC) and matched healthy control individuals (HCs). RESULTS: Microbiota composition of post-UC samples was different from HC and pre-UC samples; however, functional analysis showed increased fecal proteolytic and elastase activity before UC onset. Metagenomics identified more than 22,000 gene families that were significantly different between HC, pre-UC, and post-UC samples. Of these, 237 related to proteases and peptidases, suggesting a bacterial component to the pre-UC proteolytic signature. Elastase activity inversely correlated with the relative abundance of Adlercreutzia and other potentially beneficial taxa and directly correlated with known proteolytic taxa, such as Bacteroides vulgatus. High elastase activity was confirmed in Bacteroides isolates from fecal samples. The bacterial contribution and functional significance of the proteolytic signature were investigated in germ-free adult mice and in dams colonized with HC, pre-UC, or post-UC microbiota. Mice colonized with or born from pre-UC-colonized dams developed higher fecal proteolytic activity and an inflammatory immune tone compared with HC-colonized mice. CONCLUSIONS: We have identified increased fecal proteolytic activity that precedes the clinical diagnosis of UC and associates with gut microbiota changes. This proteolytic signature may constitute a noninvasive biomarker of inflammation to monitor at-risk populations that can be targeted therapeutically with antiproteases.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/metabolism , Colitis, Ulcerative/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Peptide Hydrolases/metabolism , Adolescent , Adult , Animals , Bacteria/drug effects , Bacteria/genetics , Bacterial Proteins/genetics , Biomarkers/metabolism , Case-Control Studies , Child , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Disease Models, Animal , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Male , Metagenome , Metagenomics , Mice, Inbred C57BL , Peptide Hydrolases/genetics , Predictive Value of Tests , Prospective Studies , Protease Inhibitors/therapeutic use , Proteolysis , Reproducibility of Results , Ribotyping , Young Adult
13.
Sci Transl Med ; 12(566)2020 10 21.
Article in English | MEDLINE | ID: mdl-33087499

ABSTRACT

Metabolism of tryptophan by the gut microbiota into derivatives that activate the aryl hydrocarbon receptor (AhR) contributes to intestinal homeostasis. Many chronic inflammatory conditions, including celiac disease involving a loss of tolerance to dietary gluten, are influenced by cues from the gut microbiota. We investigated whether AhR ligand production by the gut microbiota could influence gluten immunopathology in nonobese diabetic (NOD) mice expressing DQ8, a celiac disease susceptibility gene. NOD/DQ8 mice, exposed or not exposed to gluten, were subjected to three interventions directed at enhancing AhR pathway activation. These included a high-tryptophan diet, gavage with Lactobacillus reuteri that produces AhR ligands or treatment with an AhR agonist. We investigated intestinal permeability, gut microbiota composition determined by 16S rRNA gene sequencing, AhR pathway activation in intestinal contents, and small intestinal pathology and inflammatory markers. In NOD/DQ8 mice, a high-tryptophan diet modulated gut microbiota composition and enhanced AhR ligand production. AhR pathway activation by an enriched tryptophan diet, treatment with the AhR ligand producer L. reuteri, or pharmacological stimulation using 6-formylindolo (3,2-b) carbazole (Ficz) decreased immunopathology in NOD/DQ8 mice exposed to gluten. We then determined AhR ligand production by the fecal microbiota and AhR activation in patients with active celiac disease compared to nonceliac control individuals. Patients with active celiac disease demonstrated reduced AhR ligand production and lower intestinal AhR pathway activation. These results highlight gut microbiota-dependent modulation of the AhR pathway in celiac disease and suggest a new therapeutic strategy for treating this disorder.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Animals , Humans , Inflammation , Ligands , Mice , RNA, Ribosomal, 16S , Receptors, Aryl Hydrocarbon/genetics
14.
Nat Commun ; 10(1): 3224, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324782

ABSTRACT

Proteolytic homeostasis is important at mucosal surfaces, but its actors and their precise role in physiology are poorly understood. Here we report that healthy human and mouse colon epithelia are a major source of active thrombin. We show that mucosal thrombin is directly regulated by the presence of commensal microbiota. Specific inhibition of luminal thrombin activity causes macroscopic and microscopic damage as well as transcriptomic alterations of genes involved in host-microbiota interactions. Further, luminal thrombin inhibition impairs the spatial segregation of microbiota biofilms, allowing bacteria to invade the mucus layer and to translocate across the epithelium. Thrombin cleaves the biofilm matrix of reconstituted mucosa-associated human microbiota. Our results indicate that thrombin constrains biofilms at the intestinal mucosa. Further work is needed to test whether thrombin plays similar roles in other mucosal surfaces, given that lung, bladder and skin epithelia also express thrombin.


Subject(s)
Bacteria/metabolism , Biofilms , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Thrombin/metabolism , Animals , Cell Line , Colon/microbiology , Colonic Neoplasms/microbiology , Epithelium/microbiology , Homeostasis , Humans , Lung , Mice , Mice, Inbred C57BL , Models, Animal , Skin , Thrombin/genetics , Urinary Bladder
15.
Nat Commun ; 10(1): 1198, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867416

ABSTRACT

Microbe-host interactions are generally homeostatic, but when dysfunctional, they can incite food sensitivities and chronic diseases. Celiac disease (CeD) is a food sensitivity characterized by a breakdown of oral tolerance to gluten proteins in genetically predisposed individuals, although the underlying mechanisms are incompletely understood. Here we show that duodenal biopsies from patients with active CeD have increased proteolytic activity against gluten substrates that correlates with increased Proteobacteria abundance, including Pseudomonas. Using Pseudomonas aeruginosa producing elastase as a model, we show gluten-independent, PAR-2 mediated upregulation of inflammatory pathways in C57BL/6 mice without villus blunting. In mice expressing CeD risk genes, P. aeruginosa elastase synergizes with gluten to induce more severe inflammation that is associated with moderate villus blunting. These results demonstrate that proteases expressed by opportunistic pathogens impact host immune responses that are relevant to the development of food sensitivities, independently of the trigger antigen.


Subject(s)
Bacterial Proteins/metabolism , Celiac Disease/immunology , Dietary Proteins/immunology , Host Microbial Interactions/immunology , Metalloendopeptidases/metabolism , Receptor, PAR-2/immunology , Adult , Aged , Animals , Antigens/immunology , Antigens/metabolism , Bacterial Proteins/genetics , Biopsy , Case-Control Studies , Celiac Disease/diagnostic imaging , Celiac Disease/microbiology , Celiac Disease/pathology , Cohort Studies , Colonoscopy , Dietary Proteins/metabolism , Disease Models, Animal , Duodenum/immunology , Duodenum/metabolism , Duodenum/microbiology , Duodenum/pathology , Female , Gastrointestinal Microbiome/immunology , Germ-Free Life , Glutens/immunology , Glutens/metabolism , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DQ Antigens/metabolism , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Middle Aged , Proteolysis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Receptor, PAR-2/metabolism , Up-Regulation , Young Adult
16.
Gastroenterology ; 156(8): 2266-2280, 2019 06.
Article in English | MEDLINE | ID: mdl-30802444

ABSTRACT

BACKGROUND & AIMS: Wheat-related disorders, a spectrum of conditions induced by the ingestion of gluten-containing cereals, have been increasing in prevalence. Patients with celiac disease have gluten-specific immune responses, but the contribution of non-gluten proteins to symptoms in patients with celiac disease or other wheat-related disorders is controversial. METHODS: C57BL/6 (control), Myd88-/-, Ticam1-/-, and Il15-/- mice were placed on diets that lacked wheat or gluten, with or without wheat amylase trypsin inhibitors (ATIs), for 1 week. Small intestine tissues were collected and intestinal intraepithelial lymphocytes (IELs) were measured; we also investigated gut permeability and intestinal transit. Control mice fed ATIs for 1 week were gavaged daily with Lactobacillus strains that had high or low ATI-degrading capacity. Nonobese diabetic/DQ8 mice were sensitized to gluten and fed an ATI diet, a gluten-containing diet or a diet with ATIs and gluten for 2 weeks. Mice were also treated with Lactobacillus strains that had high or low ATI-degrading capacity. Intestinal tissues were collected and IELs, gene expression, gut permeability and intestinal microbiota profiles were measured. RESULTS: In intestinal tissues from control mice, ATIs induced an innate immune response by activation of Toll-like receptor 4 signaling to MD2 and CD14, and caused barrier dysfunction in the absence of mucosal damage. Administration of ATIs to gluten-sensitized mice expressing HLA-DQ8 increased intestinal inflammation in response to gluten in the diet. We found ATIs to be degraded by Lactobacillus, which reduced the inflammatory effects of ATIs. CONCLUSIONS: ATIs mediate wheat-induced intestinal dysfunction in wild-type mice and exacerbate inflammation to gluten in susceptible mice. Microbiome-modulating strategies, such as administration of bacteria with ATI-degrading capacity, may be effective in patients with wheat-sensitive disorders.


Subject(s)
Celiac Disease/immunology , Diet, Gluten-Free/methods , Gliadin/adverse effects , Lactobacillus/immunology , Triticum/adverse effects , Amylases/antagonists & inhibitors , Animals , Celiac Disease/diet therapy , Celiac Disease/physiopathology , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Gliadin/immunology , Humans , Immunity, Innate/drug effects , Lactobacillus/metabolism , Mice , Mice, Inbred C57BL , Random Allocation , Reference Values , Sensitivity and Specificity , Triticum/immunology , Trypsin Inhibitors/immunology , Trypsin Inhibitors/pharmacology
17.
Front Pediatr ; 6: 350, 2018.
Article in English | MEDLINE | ID: mdl-30519552

ABSTRACT

Our understanding of celiac disease and how it develops has evolved significantly over the last half century. Although traditionally viewed as a pediatric illness characterized by malabsorption, it is now better seen as an immune illness with systemic manifestations affecting all ages. Population studies reveal this global disease is common and, in many countries, increasing in prevalence. These studies underscore the importance of specific HLA susceptibility genes and gluten consumption in disease development and suggest that other genetic and environmental factors could also play a role. The emerging data on viral and bacterial microbe-host interactions and their alterations in celiac disease provides a plausible mechanism linking environmental risk and disease development. Although the inflammatory lesion of celiac disease is complex, the strong HLA association highlights a central role for pathogenic T cells responding to select gluten peptides that have now been defined for the most common genetic form of celiac disease. What remains less understood is how loss of tolerance to gluten occurs. New insights into celiac disease are now providing opportunities to intervene in its development, course, diagnosis, and treatment.

18.
Sci Rep ; 8(1): 14184, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242285

ABSTRACT

The intestinal barrier encompasses structural, permeability and immune aspects of the gut mucosa that, when disrupted, may contribute to chronic inflammation. Although gnotobiotic studies have demonstrated the effects of microbiota on mucosal and systemic immunity, as well as intestinal barrier architecture and innate immune characteristics, its impact on barrier function remains unclear. We compared germ-free and conventional mice, as well as mice colonized with human fecal microbiota that were followed for 21 days post-colonization. Colonic barrier structure was investigated by immunohistochemistry, molecular and electron microscopy techniques. Permeability was assessed in colon tissue by Ussing chambers, and by serum LPS and MDP detection using TLR4- and NOD2-NFκB reporter assays. Microbiota profile was determined by Illumina 16S rRNA gene sequencing. Low dose dextran sodium sulfate was administered to assess microbiota-induced barrier changes on resistance to colonic injury. Permeability to paracellular probes and mucus layer structure resembled that of conventional mice by day 7 post-colonization, coinciding with reduced claudin-1 expression and transient IL-18 production by intestinal epithelial cells. These post-colonization adaptations were associated with decreased systemic bacterial antigen exposure and reduced susceptibility to intestinal injury. In conclusion, commensal colonization promotes physiological barrier structural and functional adaptations that contribute to intestinal homeostasis.


Subject(s)
Colon/microbiology , Colon/physiology , Gastrointestinal Microbiome/physiology , Homeostasis/physiology , Microbiota/physiology , Animals , Colon/drug effects , Dextran Sulfate/pharmacology , Feces , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life/drug effects , Germ-Free Life/physiology , Homeostasis/drug effects , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Intestines/drug effects , Intestines/microbiology , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Permeability/drug effects , RNA, Ribosomal, 16S/metabolism
19.
J Heart Lung Transplant ; 37(9): 1047-1059, 2018 09.
Article in English | MEDLINE | ID: mdl-30173823

ABSTRACT

BACKGROUND: Mycophenolate mofetil (MMF) is commonly prescribed after transplantation and has major advantages over other immunosuppressive drugs, but frequent gastrointestinal (GI) side-effects limit its use. The mechanism(s) underlying MMF-related GI toxicity have yet to be elucidated. METHODS: To investigate MMF-related GI toxicity, experimental mice were fed chow containing MMF (0.563%) and multiple indices of toxicity, including weight loss and colonic inflammation, were measured. Changes in intestinal microbial composition were detected using 16S rRNA Illumina sequencing, and downstream PICRUSt analysis was used to predict metagenomic pathways involved. Germ-free (GF) mice and mice treated with orally administered broad-spectrum antibiotics (ABX) were utilized to interrogate the importance of the microbiota in MMF-induced GI toxicity. RESULTS: Mice treated with MMF exhibited significant weight loss, related to loss of body fat and muscle, and marked colonic inflammation. MMF exposure was associated with changes in gut microbial composition, as demonstrated by a loss of overall diversity, expansion of Proteobacteria (specifically Escherichia/Shigella), and enrichment of genes involved in lipopolysaccharide (LPS) biosynthesis, which paralleled increased levels of LPS in the feces and serum. MMF-related GI toxicity was dependent on the intestinal microbiota, as MMF did not induce weight loss or colonic inflammation in GF mice. Furthermore, ABX prevented and reversed MMF-induced weight loss and colonic inflammation. CONCLUSIONS: An intact intestinal microbiota is required to initiate and sustain the GI toxicity of MMF. MMF treatment causes dynamic changes in the composition of the intestinal microbiota that may be a targetable driver of the GI side-effects of MMF.


Subject(s)
Disease Models, Animal , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Immunosuppressive Agents/toxicity , Microbiota/drug effects , Mycophenolic Acid/toxicity , Animals , Colon/drug effects , Colon/microbiology , Germ-Free Life , High-Throughput Nucleotide Sequencing , Humans , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Inbred Strains , Microbiota/immunology , Mycophenolic Acid/therapeutic use , Proteobacteria , RNA, Ribosomal, 16S , Sequence Analysis, RNA , Weight Loss/drug effects
20.
Nature ; 557(7706): 580-584, 2018 05.
Article in English | MEDLINE | ID: mdl-29769727

ABSTRACT

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Subject(s)
Asymptomatic Diseases , Bacterial Physiological Phenomena , Cell Proliferation , DNA-Binding Proteins/deficiency , Leukemia/microbiology , Leukemia/pathology , Proto-Oncogene Proteins/deficiency , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Physiological Phenomena/immunology , DNA-Binding Proteins/genetics , Dioxygenases , Female , Germ-Free Life , Inflammation/microbiology , Interleukin-6/immunology , Intestinal Mucosa/metabolism , Lactobacillus/chemistry , Lactobacillus/cytology , Lactobacillus/immunology , Male , Mice , Penetrance , Permeability , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 2/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...