Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell Rep ; 42(8): 112997, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37611587

ABSTRACT

Colorectal cancer (CRC) is driven by genomic alterations in concert with dietary influences, with the gut microbiome implicated as an effector in disease development and progression. While meta-analyses have provided mechanistic insight into patients with CRC, study heterogeneity has limited causal associations. Using multi-omics studies on genetically controlled cohorts of mice, we identify diet as the major driver of microbial and metabolomic differences, with reductions in α diversity and widespread changes in cecal metabolites seen in high-fat diet (HFD)-fed mice. In addition, non-classic amino acid conjugation of the bile acid cholic acid (AA-CA) increased with HFD. We show that AA-CAs impact intestinal stem cell growth and demonstrate that Ileibacterium valens and Ruminococcus gnavus are able to synthesize these AA-CAs. This multi-omics dataset implicates diet-induced shifts in the microbiome and the metabolome in disease progression and has potential utility in future diagnostic and therapeutic developments.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Microbiota , Animals , Mice , Bile Acids and Salts , Metabolome
2.
bioRxiv ; 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36993409

ABSTRACT

Disease tolerance is a defense strategy essential for survival of infections, limiting physiological damage without killing the pathogen. The disease course and pathology a pathogen may cause can change over the lifespan of a host due to the structural and functional physiological changes that accumulate with age. Since successful disease tolerance responses require the host to engage mechanisms that are compatible with the disease course and pathology caused by an infection, we predicted that this defense strategy would change with age. Animals infected with a lethal dose 50 (LD50) of a pathogen often display distinct health and sickness trajectories due to differences in disease tolerance, and thus can be used to delineate tolerance mechanisms. Using a polymicrobial sepsis model, we found that despite having the same LD50, old and young susceptible mice exhibited distinct disease courses. Young survivors employed a cardioprotective mechanism via FoxO1-mediated regulation of the ubiquitin-proteosome system that was necessary for survival and protection from cardiomegaly. This same mechanism was a driver of sepsis pathogenesis in aged hosts, causing catabolic remodeling of the heart and death. Our findings have implications for the tailoring of therapy to the age of an infected individual and suggest that disease tolerance alleles may exhibit antagonistic pleiotropy.

4.
Infect Immun ; 90(9): e0024222, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35924898

ABSTRACT

To combat infections, hosts employ a combination of antagonistic and cooperative defense strategies. The former refers to pathogen killing mediated by resistance mechanisms, while the latter refers to physiological defense mechanisms that promote host health during infection independent of pathogen killing, leading to an apparent cooperation between the host and the pathogen. Previous work has shown that Leptin, a pleiotropic hormone that plays a central role in regulating appetite and energy metabolism, is indispensable for resistance mechanisms, while a role for Leptin signaling in cooperative host-pathogen interactions remains unknown. Using a mouse model of Yersinia pseudotuberculosis (Yptb) infection, an emerging pathogen that causes fever, diarrhea, and mesenteric lymphadenitis in humans, we found that the physiological effects of chronic Leptin-signaling deficiency conferred protection from Yptb infection due to increased host-pathogen cooperation rather than greater resistance defenses. The protection against Yptb infection was independent of differences in food consumption, lipolysis, or fat mass. Instead, we found that the chronic absence of Leptin signaling protects from a shift to lipid utilization during infection that contributes to Yptb lethality. Furthermore, we found that the survival advantage conferred by Leptin deficiency was associated with increased liver and kidney damage. Our work reveals an additional level of complexity for the role of Leptin in infection defense and demonstrates that in some contexts, in addition to tolerating the pathogen, tolerating organ damage is more beneficial for survival than preventing the damage.


Subject(s)
Yersinia pseudotuberculosis Infections , Yersinia pseudotuberculosis , Host-Pathogen Interactions , Humans , Leptin/metabolism , Lipids , Yersinia pseudotuberculosis/metabolism
5.
Immunity ; 55(5): 824-826, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545032

ABSTRACT

The mechanisms of how infectious diseases contribute to allergy remain unanswered. In this issue of Immunity, Agaronyan et al. (2022) show that Pseudomonas aeruginosa drives immune deviation through induction of type 2 immune responses, resulting in niche remodeling that incites allergic responses to innocuous antigens.


Subject(s)
Hypersensitivity , Pseudomonas Infections , Humans , Pseudomonas , Pseudomonas aeruginosa , Virulence
6.
Clin Gastroenterol Hepatol ; 19(11): 2343-2352.e8, 2021 11.
Article in English | MEDLINE | ID: mdl-32827724

ABSTRACT

BACKGROUND & AIMS: Many patients with irritable bowel syndrome (IBS) perceive that their symptoms are triggered by wheat-containing foods. We assessed symptoms and gastrointestinal transit before and after a gluten-free diet (GFD) in unselected patients with IBS and investigated biomarkers associated with symptoms. METHODS: We performed a prospective study of 50 patients with IBS (ROME III, all subtypes), with and without serologic reactivity to gluten (antigliadin IgG and IgA), and 25 healthy subjects (controls) at a university hospital in Hamilton, Ontario, Canada, between 2012 and 2016. Gastrointestinal transit, gut symptoms, anxiety, depression, somatization, dietary habits, and microbiota composition were studied before and after 4 weeks of a GFD. HLA-DQ2/DQ8 status was determined. GFD compliance was assessed by a dietitian and by measuring gluten peptides in stool. RESULTS: There was no difference in symptoms among patients at baseline, but after the GFD, patients with antigliadin IgG and IgA reported less diarrhea than patients without these antibodies (P = .03). Compared with baseline, IBS symptoms improved in 18 of 24 patients (75%) with antigliadin IgG and IgA and in 8 of 21 patients (38%) without the antibodies. Although constipation, diarrhea, and abdominal pain were reduced in patients with antigliadin IgG and IgA, only pain decreased in patients without these antibodies. Gastrointestinal transit normalized in a higher proportion of patients with antigliadin IgG and IgA. Anxiety, depression, somatization, and well-being increased in both groups. The presence of antigliadin IgG was associated with overall reductions in symptoms (adjusted odds ratio compared with patients without this antibody, 128.9; 95% CI, 1.16-1427.8; P = .04). Symptoms were reduced even in patients with antigliadin IgG and IgA who reduced gluten intake but were not strictly compliant with the GFD. In controls, a GFD had no effect on gastrointestinal symptoms or gut function. CONCLUSIONS: Antigliadin IgG can be used as a biomarker to identify patients with IBS who might have reductions in symptoms, particularly diarrhea, on a GFD. Larger studies are needed to validate these findings. ClinicalTrials.gov: NCT03492333.


Subject(s)
Celiac Disease , Irritable Bowel Syndrome , Diarrhea , Diet, Gluten-Free , Humans , Immunoglobulin G , Prospective Studies
7.
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
8.
Annu Rev Immunol ; 38: 147-170, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340573

ABSTRACT

Metabolism is one of the strongest drivers of interkingdom interactions-including those between microorganisms and their multicellular hosts. Traditionally thought to fuel energy requirements and provide building blocks for biosynthetic pathways, metabolism is now appreciated for its role in providing metabolites, small-molecule intermediates generated from metabolic processes, to perform various regulatory functions to mediate symbiotic relationships between microbes and their hosts. Here, we review recent advances in our mechanistic understanding of how microbiota-derived metabolites orchestrate and support physiological responses in the host, including immunity, inflammation, defense against infections, and metabolism. Understanding how microbes metabolically communicate with their hosts will provide us an opportunity to better describe how a host interacts with all microbes-beneficial, pathogenic, and commensal-and an opportunity to discover new ways to treat microbial-driven diseases.


Subject(s)
Disease Susceptibility , Energy Metabolism , Homeostasis , Microbiota , Symbiosis , Animals , Disease Susceptibility/immunology , Host-Pathogen Interactions , Humans , Immune System/immunology , Immune System/metabolism , Microbiota/immunology
9.
Cell Metab ; 30(3): 409-411, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31484052

ABSTRACT

During infection, disease tolerance promotes host health without killing the pathogen. In a recent issue of Cell, Luan et al. (2019) identify GDF15 as a central regulator of disease tolerance of bacterial and viral challenges, while preventing cardiac damage, by mediating downstream cross-organ communication via the sympathetic and metabolic systems.


Subject(s)
Host-Pathogen Interactions , Immune Tolerance , Growth Differentiation Factor 15 , Humans , Inflammation
10.
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
11.
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
12.
Nat Chem Biol ; 14(11): 1021-1031, 2018 11.
Article in English | MEDLINE | ID: mdl-30327559

ABSTRACT

Fatty acid synthase (FASN) predominantly generates straight-chain fatty acids using acetyl-CoA as the initiating substrate. However, monomethyl branched-chain fatty acids (mmBCFAs) are also present in mammals but are thought to be primarily diet derived. Here we demonstrate that mmBCFAs are de novo synthesized via mitochondrial BCAA catabolism, exported to the cytosol by adipose-specific expression of carnitine acetyltransferase (CrAT), and elongated by FASN. Brown fat exhibits the highest BCAA catabolic and mmBCFA synthesis fluxes, whereas these lipids are largely absent from liver and brain. mmBCFA synthesis is also sustained in the absence of microbiota. We identify hypoxia as a potent suppressor of BCAA catabolism that decreases mmBCFA synthesis in obese adipose tissue, such that mmBCFAs are significantly decreased in obese animals. These results identify adipose tissue mmBCFA synthesis as a novel link between BCAA metabolism and lipogenesis, highlighting roles for CrAT and FASN promiscuity influencing acyl-chain diversity in the lipidome.


Subject(s)
Adipose Tissue/enzymology , Amino Acids, Branched-Chain/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/biosynthesis , Obesity/enzymology , 3T3 Cells , Adipocytes/cytology , Animals , CRISPR-Cas Systems , Carnitine O-Acetyltransferase/metabolism , Cytosol/metabolism , Female , Hypoxia , Lentivirus/genetics , Lipogenesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , RNA, Small Interfering/metabolism
13.
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
14.
Microbiome ; 6(1): 57, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29566748

ABSTRACT

BACKGROUND: Changes in hygiene and dietary habits, including increased consumption of foods high in fat, simple sugars, and salt that are known to impact the composition and function of the intestinal microbiota, may explain the increase in prevalence of chronic inflammatory diseases. High salt consumption has been shown to worsen autoimmune encephalomyelitis and colitis in mouse models through p38/MAPK signaling pathway. However, the effect of high salt diet (HSD) on gut microbiota and on intestinal immune homeostasis, and their roles in determining vulnerability to intestinal inflammatory stimuli are unknown. Here, we investigate the role of gut microbiota alterations induced by HSD on the severity of murine experimental colitis. RESULTS: Compared to control diet, HSD altered fecal microbiota composition and function, reducing Lactobacillus sp. relative abundance and butyrate production. Moreover, HSD affected the colonic, and to a lesser extent small intestine mucosal immunity by enhancing the expression of pro-inflammatory genes such as Rac1, Map2k1, Map2k6, Atf2, while suppressing many cytokine and chemokine genes, such as Ccl3, Ccl4, Cxcl2, Cxcr4, Ccr7. Conventionally raised mice fed with HSD developed more severe DSS- (dextran sodium sulfate) and DNBS- (dinitrobenzene sulfonic acid) induced colitis compared to mice on control diet, and this effect was absent in germ-free mice. Transfer experiments into germ-free mice indicated that the HSD-associated microbiota profile is critically dependent on continued exposure to dietary salt. CONCLUSIONS: Our results indicate that the exacerbation of colitis induced by HSD is associated with reduction in Lactobacillus sp. and protective short-chain fatty acid production, as well as changes in host immune status. We hypothesize that these changes alter gut immune homeostasis and lead to increased vulnerability to inflammatory insults.


Subject(s)
Butyrates/metabolism , Colitis/etiology , Colitis/metabolism , Diet , Gastrointestinal Microbiome , Lactobacillus , Salts , Animals , Colitis/pathology , Disease Models, Animal , Disease Progression , Fatty Acids/metabolism , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA, Ribosomal, 16S/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
Sci Rep ; 6: 30980, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27481162

ABSTRACT

Celiac disease is triggered by partially digested gluten proteins. Enzyme therapies that complete protein digestion in vivo could support a gluten-free diet, but the barrier to completeness is high. Current options require enzyme amounts on the same order as the protein meal itself. In this study, we evaluated proteolytic components of the carnivorous pitcher plant (Nepenthes spp.) for use in this context. Remarkably low doses enhance gliadin solubilization rates, and degrade gliadin slurries within the pH and temporal constraints of human gastric digestion. Potencies in excess of 1200:1 (substrate-to-enzyme) are achieved. Digestion generates small peptides through nepenthesin and neprosin, the latter a novel enzyme defining a previously-unknown class of prolyl endoprotease. The digests also exhibit reduced TG2 conversion rates in the immunogenic regions of gliadin, providing a twin mechanism for evading T-cell recognition. When sensitized and dosed with enzyme-treated gliadin, NOD/DQ8 mice did not show intestinal inflammation, when compared to mice challenged with only pepsin-treated gliadin. The low enzyme load needed for effective digestion suggests that gluten detoxification can be achieved in a meal setting, using metered dosing based on meal size. We demonstrate this by showing efficient antigen processing at total substrate-to-enzyme ratios exceeding 12,000:1.


Subject(s)
Celiac Disease/therapy , Diet, Gluten-Free , Enzyme Therapy , GTP-Binding Proteins/metabolism , Gliadin/metabolism , Glutens/metabolism , Transglutaminases/metabolism , Animals , Celiac Disease/enzymology , Celiac Disease/immunology , Drosophila/metabolism , Female , Humans , Hydrogen-Ion Concentration , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred NOD , Protein Glutamine gamma Glutamyltransferase 2 , Proteolysis
16.
Gastroenterology ; 151(4): 670-83, 2016 10.
Article in English | MEDLINE | ID: mdl-27373514

ABSTRACT

BACKGROUND & AIMS: Partially degraded gluten peptides from cereals trigger celiac disease (CD), an autoimmune enteropathy occurring in genetically susceptible persons. Susceptibility genes are necessary but not sufficient to induce CD, and additional environmental factors related to unfavorable alterations in the microbiota have been proposed. We investigated gluten metabolism by opportunistic pathogens and commensal duodenal bacteria and characterized the capacity of the produced peptides to activate gluten-specific T-cells from CD patients. METHODS: We colonized germ-free C57BL/6 mice with bacteria isolated from the small intestine of CD patients or healthy controls, selected for their in vitro gluten-degrading capacity. After gluten gavage, gliadin amount and proteolytic activities were measured in intestinal contents. Peptides produced by bacteria used in mouse colonizations from the immunogenic 33-mer gluten peptide were characterized by liquid chromatography tandem mass spectrometry and their immunogenic potential was evaluated using peripheral blood mononuclear cells from celiac patients after receiving a 3-day gluten challenge. RESULTS: Bacterial colonizations produced distinct gluten-degradation patterns in the mouse small intestine. Pseudomonas aeruginosa, an opportunistic pathogen from CD patients, exhibited elastase activity and produced peptides that better translocated the mouse intestinal barrier. P aeruginosa-modified gluten peptides activated gluten-specific T-cells from CD patients. In contrast, Lactobacillus spp. from the duodenum of non-CD controls degraded gluten peptides produced by human and P aeruginosa proteases, reducing their immunogenicity. CONCLUSIONS: Small intestinal bacteria exhibit distinct gluten metabolic patterns in vivo, increasing or reducing gluten peptide immunogenicity. This microbe-gluten-host interaction may modulate autoimmune risk in genetically susceptible persons and may underlie the reported association of dysbiosis and CD.


Subject(s)
Celiac Disease/immunology , Celiac Disease/microbiology , Duodenum/microbiology , Glutens/immunology , Glutens/metabolism , Immunogenetic Phenomena , Animals , Bacterial Translocation , Case-Control Studies , Celiac Disease/genetics , Humans , Lactobacillus/physiology , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/physiology , T-Lymphocytes/immunology
17.
Therap Adv Gastroenterol ; 9(4): 580-93, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27366225

ABSTRACT

The gut microbiota contributes to the maintenance of health and, when disrupted, may drive gastrointestinal and extragastrointestinal disease. This can occur through direct pathways such as interaction with the epithelial barrier and mucosal immune system or indirectly via production of metabolites. There is no current curative therapy for chronic inflammatory conditions such as inflammatory bowel disease, which are complex multifactorial disorders involving genetic predisposition, and environmental triggers. Therapies are directed to suppress inflammation rather than the driver, and these approaches are not devoid of adverse effects. Therefore, there is great interest in modulation of the gut microbiota to provide protection from disease. Interventions that modulate the microbiota include diet, probiotics and more recently the emergence of experimental therapies such as fecal microbiota transplant or phage therapy. Emerging data indicate that certain bacteria can induce protective immune responses and enhance intestinal barrier function, which could be potential therapeutic targets. However, mechanistic links and specific therapeutic recommendations are still lacking. Here we provide a pathophysiological overview of potential therapeutic applications of the gut microbiota.

18.
Am J Physiol Gastrointest Liver Physiol ; 311(1): G40-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27151946

ABSTRACT

Celiac disease (CD) is an immune-mediated enteropathy triggered by gluten in genetically susceptible individuals. Innate immunity contributes to the pathogenesis of CD, but the mechanisms remain poorly understood. Although previous in vitro work suggests that gliadin peptide p31-43 acts as an innate immune trigger, the underlying pathways are unclear and have not been explored in vivo. Here we show that intraluminal delivery of p31-43 induces morphological changes in the small intestinal mucosa of normal mice consistent with those seen in CD, including increased cell death and expression of inflammatory mediators. The effects of p31-43 were dependent on MyD88 and type I IFNs, but not Toll-like receptor 4 (TLR4), and were enhanced by coadministration of the TLR3 agonist polyinosinic:polycytidylic acid. Together, these results indicate that gliadin peptide p31-43 activates the innate immune pathways in vivo, such as IFN-dependent inflammation, relevant to CD. Our findings also suggest a common mechanism for the potential interaction between dietary gluten and viral infections in the pathogenesis of CD.


Subject(s)
Celiac Disease/immunology , Gliadin/toxicity , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Peptide Fragments/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Celiac Disease/metabolism , Celiac Disease/pathology , Gene Expression Regulation , Genotype , Gliadin/administration & dosage , Inflammation Mediators/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Peptide Fragments/administration & dosage , Phenotype , Poly I-C/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction/drug effects , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism
19.
J Cell Physiol ; 231(11): 2529-40, 2016 11.
Article in English | MEDLINE | ID: mdl-27100271

ABSTRACT

Polymorphisms in the PTPN11 gene encoding for the tyrosine phosphatase SHP-2 were described in patients with ulcerative colitis. We have recently demonstrated that mice with an intestinal epithelial cell-specific deletion of SHP-2 (SHP-2(IEC-KO) ) develop severe colitis 1 month after birth. However, the mechanisms by which SHP-2 deletion induces colonic inflammation remain to be elucidated. We generated SHP-2(IEC-KO) mice lacking Myd88 exclusively in the intestinal epithelium. The colonic phenotype was histologically analyzed and cell differentiation was determined by electron microscopy and lysozyme or Alcian blue staining. Microbiota composition was analyzed by 16S sequencing. Results show that innate defense genes including those specific to Paneth cells were strongly up-regulated in SHP-2-deficient colons. Expansion of intermediate cells (common progenitors of the Goblet and Paneth cell lineages) was found in the colon of SHP-2(IEC-KO) mice whereas Goblet cell number was clearly diminished. These alterations in Goblet/intermediate cell ratio were noticed 2 weeks after birth, before the onset of inflammation and were associated with significant alterations in microbiota composition. Indeed, an increase in Enterobacteriaceae and a decrease in Firmicutes were observed in the colon of these mice, indicating that dysbiosis also occurred prior to inflammation. Importantly, loss of epithelial Myd88 expression inhibited colitis development in SHP-2(IEC-KO) mice, rescued Goblet/intermediate cell ratio, and prevented NFκB hyperactivation and inflammation. These data indicate that SHP-2 is functionally important for the maintenance of appropriate barrier function and host-microbiota homeostasis in the large intestine. J. Cell. Physiol. 231: 2529-2540, 2016. © 2016 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation , Colon/pathology , Homeostasis , Inflammation/pathology , Inflammation/prevention & control , Microbiota , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Animals, Newborn , Antimicrobial Cationic Peptides/metabolism , Biomarkers/metabolism , Goblet Cells/metabolism , Goblet Cells/pathology , Inflammation/genetics , Mice, Inbred C57BL , Muramidase/metabolism , Myeloid Differentiation Factor 88/metabolism , Paneth Cells/metabolism , Paneth Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Up-Regulation/genetics
20.
Am J Pathol ; 185(11): 2969-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26456581

ABSTRACT

Celiac disease (CD) is an immune-mediated enteropathy triggered by gluten in genetically susceptible individuals. The recent increase in CD incidence suggests that additional environmental factors, such as intestinal microbiota alterations, are involved in its pathogenesis. However, there is no direct evidence of modulation of gluten-induced immunopathology by the microbiota. We investigated whether specific microbiota compositions influence immune responses to gluten in mice expressing the human DQ8 gene, which confers moderate CD genetic susceptibility. Germ-free mice, clean specific-pathogen-free (SPF) mice colonized with a microbiota devoid of opportunistic pathogens and Proteobacteria, and conventional SPF mice that harbor a complex microbiota that includes opportunistic pathogens were used. Clean SPF mice had attenuated responses to gluten compared to germ-free and conventional SPF mice. Germ-free mice developed increased intraepithelial lymphocytes, markers of intraepithelial lymphocyte cytotoxicity, gliadin-specific antibodies, and a proinflammatory gliadin-specific T-cell response. Antibiotic treatment, leading to Proteobacteria expansion, further enhanced gluten-induced immunopathology in conventional SPF mice. Protection against gluten-induced immunopathology in clean SPF mice was reversed after supplementation with a member of the Proteobacteria phylum, an enteroadherent Escherichia coli isolated from a CD patient. The intestinal microbiota can both positively and negatively modulate gluten-induced immunopathology in mice. In subjects with moderate genetic susceptibility, intestinal microbiota changes may be a factor that increases CD risk.


Subject(s)
Antibodies/blood , Celiac Disease/microbiology , Gastrointestinal Microbiome , Glutens/adverse effects , Animals , Anti-Bacterial Agents/administration & dosage , Celiac Disease/immunology , Celiac Disease/pathology , Cell Proliferation , Cytokines/analysis , Feces/microbiology , Female , Gliadin/adverse effects , Humans , Male , Mice , Mice, Inbred NOD , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology , Vancomycin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...