Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Gut Microbes ; 16(1): 2298026, 2024.
Article En | MEDLINE | ID: mdl-38170633

Gut - brain communications disorders in irritable bowel syndrome (IBS) are associated with intestinal microbiota composition, increased gut permeability, and psychosocial disturbances. Symptoms of IBS are difficult to medicate, and hence much research is being made into alternative approaches. This study assesses the potential of a treatment with pasteurized Akkermansia muciniphila for alleviating IBS-like symptoms in two mouse models of IBS with different etiologies. Two clinically relevant animal models were used to mimic IBS-like symptoms in C57BL6/J mice: the neonatal maternal separation (NMS) paradigm and the Citrobacter rodentium infection model. In both models, gut permeability, colonic sensitivity, fecal microbiota composition and colonic IL-22 expression were evaluated. The cognitive performance and emotional state of the animals were also assessed by several tests in the C. rodentium infection model. The neuromodulation ability of pasteurized A. muciniphila was assessed on primary neuronal cells from mice dorsal root ganglia using a ratiometric calcium imaging approach. The administration of pasteurized A. muciniphila significantly reduced colonic hypersensitivity in both IBS mouse models, accompanied by a reinforcement of the intestinal barrier function. Beneficial effects of pasteurized A. muciniphila treatment have also been observed on anxiety-like behavior and memory defects in the C. rodentium infection model. Finally, a neuroinhibitory effect exerted by pasteurized A. muciniphila was observed on neuronal cells stimulated with two algogenic substances such as capsaicin and inflammatory soup. Our findings demonstrate novel anti-hyperalgesic and neuroinhibitory properties of pasteurized A. muciniphila, which therefore may have beneficial effects in relieving pain and anxiety in subjects with IBS.


Gastrointestinal Microbiome , Irritable Bowel Syndrome , Humans , Mice , Animals , Irritable Bowel Syndrome/therapy , Maternal Deprivation , Verrucomicrobia/physiology
2.
Sci Rep ; 12(1): 19776, 2022 11 17.
Article En | MEDLINE | ID: mdl-36396717

Probiotic supplementation can help to mitigate the pathogenesis of irritable bowel syndrome (IBS) by reinforcing the intestinal barrier, and reducing both inflammation and proteolytic activity. Here, a combination of in vitro tests was performed on 33 Bifidobacterium strains as probiotic candidates for IBS. In addition to the classical tests performed, the detection of the serine protease inhibitor (serpin) enzyme capable of decreasing the high proteolytic activity found in IBS patients was included. Three serpin-positive strains were selected: Bifidobacterium breve CNCM I-5644, Bifidobacterium longum subsp. infantis CNCM I-5645 and B. longum CNCM I-5646 for their immunomodulation properties and protection of intestinal epithelial integrity in vitro. Furthermore, we found that B. breve CNCM I-5644 strain prevented intestinal hyperpermeability by upregulating Cingulin and Tight Junction Protein 1 mRNA levels and reducing pro-inflammatory markers. The ability of CNCM I-5644 strain to restore intestinal hyperpermeability (FITC-dextran) was shown in the murine model of low-grade inflammation induced by dinitrobenzene sulfonic acid (DNBS). This effect of this strain was corroborated in a second model of IBS, the neonatal maternal separation model in mice. Altogether, these data suggest that serpin-positive B. breve CNCM I-5644 may partially prevent disorders associated with increased barrier permeability such as IBS.


Bifidobacterium breve , Irritable Bowel Syndrome , Serpins , Mice , Animals , Maternal Deprivation , Permeability , Inflammation , Bifidobacterium longum subspecies infantis
3.
World J Gastroenterol ; 22(31): 7111-23, 2016 Aug 21.
Article En | MEDLINE | ID: mdl-27610021

AIM: To investigate anti-hypersensitive effects of α2δ-1 ligands in non-inflammatory and inflammation-associated colonic hypersensitivity (CHS) mouse models. METHODS: To induce an inflammation-associated CHS, 1% dextran sulfate sodium (DSS) was administered to C57Bl/6J male mice, in drinking water, for 14 d. Regarding the non-inflammatory neonatal maternal separation (NMS) -induced CHS model, wild-type C57BI/6J pups were isolated from their mother from day 2 to day 14 (P2 to P14), three hours per day (from 9:00 a.m. to 12:00 p.m.). Colorectal distension was performed by inflating distension probe from 20 µL to 100 µL by 20 µL increment step every 10 s. After a first colorectal distension (CRD), drugs were administered subcutaneously, in a cumulative manner, (Gabapentin at 30 mg/kg and 100 mg/kg; Pregabalin at 10 mg/kg and 30 mg/kg; Carbamazepine at 10 mg/kg and 30 mg/kg) and a second CRD was performed one hour after each injection. RESULTS: The visceromotor response (VMR) to CRD was increased by our NMS paradigm protocol in comparison to non-handled (NH) mice, considering the highest distension volumes (80 µL: 0.783 ± 0.056 mV/s vs 0.531 ± 0.034 mV/s, P < 0.05 and 100 µL: 1.087 ± 0.056 mV/s vs 0.634 ± 0.038 mV/s, P < 0.05 for NMS and NH mice, respectively). In the inflammation-associated CHS, DSS-treated mice showed a dramatic and significant increase in VMR at 60 and 80 µL distension volumes when compared to control mice (60 µL: 0.920 ± 0.079 mV/s vs 0.426 ± 0.100 mV/s P < 0.05 and 80 µL: 1.193 ± 0.097 mV/s vs 0.681 ± 0.094 mV/s P < 0.05 for DSS- and Water-treated mice, respectively). Carbamazepine failed to significantly reduce CHS in both models. Gabapentin significantly reduced CHS in the DSS-induced model for both subcutaneous injections at 30 or 100 mg/kg. Pregabalin significantly reduced VMR to CRD in the non-inflammatory NMS-induced CHS model for the acute subcutaneous administration of the highest cumulative dose (30 mg/kg) and significantly reduced CHS in low-dose DSS-treated mice in a dose-dependent manner. Finally, the percent decrease of AUC induced by acute GBP or Pregabalin treatment were higher in the inflammatory DSS-induced CHS model in comparison to the non-inflammatory NMS-induced CHS model. CONCLUSION: This preclinical study demonstrates α2δ-1 ligands efficacy on inflammation-associated CHS, highlighting their potential clinical interest in patients with chronic abdominal pain and moderate intestinal inflammation.


Calcium Channels/physiology , Inflammatory Bowel Diseases/drug therapy , Amines/therapeutic use , Animals , Cyclohexanecarboxylic Acids/therapeutic use , Dextran Sulfate , Disease Models, Animal , Gabapentin , Ligands , Male , Mice , Mice, Inbred C57BL , Pregabalin/therapeutic use , gamma-Aminobutyric Acid/therapeutic use
4.
Immunity ; 41(3): 478-492, 2014 Sep 18.
Article En | MEDLINE | ID: mdl-25220212

Systems biological analysis of immunity to the trivalent inactivated influenza vaccine (TIV) in humans revealed a correlation between early expression of TLR5 and the magnitude of the antibody response. Vaccination of Trl5(-/-) mice resulted in reduced antibody titers and lower frequencies of plasma cells, demonstrating a role for TLR5 in immunity to TIV. This was due to a failure to sense host microbiota. Thus, antibody responses in germ-free or antibiotic-treated mice were impaired, but restored by oral reconstitution with a flagellated, but not aflagellated, strain of E. coli. TLR5-mediated sensing of flagellin promoted plasma cell differentiation directly and by stimulating lymph node macrophages to produce plasma cell growth factors. Finally, TLR5-mediated sensing of the microbiota also impacted antibody responses to the inactivated polio vaccine, but not to adjuvanted vaccines or the live-attenuated yellow fever vaccine. These results reveal an unappreciated role for gut microbiota in promoting immunity to vaccination.


Antibody Formation/immunology , Influenza Vaccines/immunology , Intestines/microbiology , Microbiota/immunology , Toll-Like Receptor 5/immunology , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , Cell Differentiation/immunology , Escherichia coli/immunology , Flagellin/immunology , Humans , Immunologic Memory/immunology , Influenza, Human/prevention & control , Intestines/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , Poliovirus Vaccine, Inactivated/immunology , Signal Transduction/immunology , Toll-Like Receptor 5/biosynthesis , Toll-Like Receptor 5/genetics , Yellow Fever Vaccine/immunology
5.
Gut ; 63(7): 1069-80, 2014 Jul.
Article En | MEDLINE | ID: mdl-23896971

BACKGROUND: Inflammatory bowel disease (IBD) is driven by a seemingly aberrant immune response to the gut microbiota with disease development dictated by genetics and environmental factors. A model exemplifying this notion is our recent demonstration that colonisation of adherent-invasive Escherichia coli (AIEC) during microbiota acquisition drove chronic colitis in mice lacking the flagellin receptor TLR5 (T5KO). T5KO colitis persisted beyond AIEC clearance and requires TLR4 and the NLRC4 inflammasome. We hypothesised that AIEC instigates chronic inflammation by increasing microbial lipopolysaccharide (LPS) and flagellin levels. GOAL: Examine if transient AIEC colonisation lastingly alters levels of LPS and flagellin and changes microbiota composition. METHODS: Germ-free mice (wild type (WT) and T5KO) were inoculated with AIEC strain LF82 and placed in standard housing allowing a complex microbiota that eliminated AIEC in both mice strains. Faeces were assayed for the inflammatory marker, lipocalin-2, bacterial loads, and microbiota composition by pyrosequencing. Faecal LPS and flagellin bioactivity were measured via a cell-based reporter assay. RESULTS: Transient AIEC colonisation, in WT mice, did not alter inflammatory markers, bacterial loads, microbiota composition, nor its pro-inflammatory potential. By contrast, transient AIEC colonisation of T5KO mice drove chronic inflammation which correlated with microbiota components having higher levels of bioactive LPS and flagellin. Such AIEC-induced elevation of LPS and flagellin persisted well beyond AIEC clearance, required AIEC be flagellated, and was associated with alteration in microbiota species composition including a loss of species diversity. CONCLUSIONS: AIEC, and perhaps other pathobionts, may instigate chronic inflammation in susceptible hosts by altering the gut microbiota composition so as to give it an inherently greater ability to activate innate immunity/pro-inflammatory gene expression.


Colitis/microbiology , Colon/microbiology , Escherichia coli Infections/complications , Intestinal Mucosa/microbiology , Microbiota , Toll-Like Receptor 5/deficiency , Acute-Phase Proteins/metabolism , Animals , Bacterial Adhesion/immunology , Bacterial Load , Biomarkers/metabolism , Chronic Disease , Colitis/immunology , DNA, Bacterial/analysis , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/immunology , Feces/chemistry , Feces/microbiology , Flagellin/metabolism , Germ-Free Life , Immunity, Innate , Intestinal Mucosa/immunology , Lipocalin-2 , Lipocalins/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Knockout , Oncogene Proteins/metabolism , Sequence Analysis, DNA
6.
Gut ; 63(2): 281-91, 2014 Feb.
Article En | MEDLINE | ID: mdl-23426893

OBJECTIVE: The inner mucus layer in mouse colon normally separates bacteria from the epithelium. Do humans have a similar inner mucus layer and are defects in this mucus layer a common denominator for spontaneous colitis in mice models and ulcerative colitis (UC)? METHODS AND RESULTS: The colon mucus layer from mice deficient in Muc2 mucin, Core 1 O-glycans, Tlr5, interleukin 10 (IL-10) and Slc9a3 (Nhe3) together with that from dextran sodium sulfate-treated mice was immunostained for Muc2, and bacterial localisation in the mucus was analysed. All murine colitis models revealed bacteria in contact with the epithelium. Additional analysis of the less inflamed IL-10(-/-) mice revealed a thicker mucus layer than wild-type, but the properties were different, as the inner mucus layer could be penetrated both by bacteria in vivo and by fluorescent beads the size of bacteria ex vivo. Clear separation between bacteria or fluorescent beads and the epithelium mediated by the inner mucus layer was also evident in normal human sigmoid colon biopsy samples. In contrast, mucus on colon biopsy specimens from patients with UC with acute inflammation was highly penetrable. Most patients with UC in remission had an impenetrable mucus layer similar to that of controls. CONCLUSIONS: Normal human sigmoid colon has an inner mucus layer that is impenetrable to bacteria. The colon mucus in animal models that spontaneously develop colitis and in patients with active UC allows bacteria to penetrate and reach the epithelium. Thus colon mucus properties can be modulated, and this suggests a novel model of UC pathophysiology.


Colitis, Ulcerative/microbiology , Colitis/microbiology , Colon/microbiology , Intestinal Mucosa/microbiology , Mucin-2/metabolism , Mucus/microbiology , Adolescent , Adult , Aged , Animals , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Female , Humans , In Situ Hybridization, Fluorescence , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Middle Aged , Young Adult
7.
Cell Host Microbe ; 12(2): 139-52, 2012 Aug 16.
Article En | MEDLINE | ID: mdl-22863420

Colitis results from breakdown of homeostasis between intestinal microbiota and the mucosal immune system, with both environmental and genetic influencing factors. Flagellin receptor TLR5-deficient mice (T5KO) display elevated intestinal proinflammatory gene expression and colitis with incomplete penetrance, providing a genetically sensitized system to study the contribution of microbiota to driving colitis. Both colitic and noncolitic T5KO exhibited transiently unstable microbiotas, with lasting differences in colitic T5KO, while their noncolitic siblings stabilized their microbiotas to resemble wild-type mice. Transient high levels of proteobacteria, especially enterobacteria species including E. coli, observed in close proximity to the gut epithelium were a striking feature of colitic microbiota. A Crohn's disease-associated E. coli strain induced chronic colitis in T5KO, which persisted well after the exogenously introduced bacterial species had been eliminated. Thus, an innate immune deficiency can result in unstable gut microbiota associated with low-grade inflammation, and harboring proteobacteria can drive and/or instigate chronic colitis.


Colitis/microbiology , Gastrointestinal Tract/microbiology , Proteobacteria/physiology , Toll-Like Receptor 5/immunology , Animals , Colitis/genetics , Colitis/immunology , Enterobacteriaceae/physiology , Female , Gastrointestinal Tract/immunology , Humans , Male , Metagenome , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics
8.
J Immunol ; 189(4): 1911-9, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22786765

Various states of inflammation, including sepsis, are associated with hypoferremia, which limits iron availability to pathogens and reduces iron-mediated oxidative stress. Lipocalin 2 (Lcn2; siderocalin, 24p3) plays a central role in iron transport. Accordingly, Lcn2-deficient (Lcn2KO) mice exhibit elevated intracellular labile iron. In this study, we report that LPS induced systemic Lcn2 by 150-fold in wild-type mice at 24 h. Relative to wild-type littermates, Lcn2KO mice were markedly more sensitive to endotoxemia, exhibiting elevated indices of organ damage (transaminasemia, lactate dehydrogenase) and increased mortality. Such exacerbated endotoxemia was associated with substantially increased caspase-3 cleavage and concomitantly elevated immune cell apoptosis. Furthermore, cells from Lcn2KO mice were hyperresponsive to LPS ex vivo, exhibiting elevated cytokine secretion. Additionally, Lcn2KO mice exhibited delayed LPS-induced hypoferremia despite normal hepatic hepcidin expression and displayed decreased levels of the tissue redox state indicators cysteine and glutathione in liver and plasma. Desferroxamine, an iron chelator, significantly protects Lcn2KO mice from LPS-induced toxicity, including mortality, suggesting that Lcn2 may act as an antioxidant in vivo by regulating iron homeostasis. Thus, Lcn2-mediated regulation of labile iron protects the host against sepsis. Its small size and simple structure may make Lcn2 a deployable treatment for sepsis.


Acute-Phase Proteins/metabolism , Homeostasis/physiology , Iron/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Sepsis/metabolism , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/immunology , Animals , Apoptosis/physiology , Endotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Lipocalin-2 , Lipocalins/immunology , Male , Mice , Mice, Knockout , Oncogene Proteins/deficiency , Oncogene Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/chemically induced , Sepsis/immunology
9.
Annu Rev Physiol ; 74: 177-98, 2012.
Article En | MEDLINE | ID: mdl-22035346

The well-being of the intestine and its host requires that this organ execute its complex function amid colonization by a large and diverse microbial community referred to as the gut microbiota. A myriad of interacting mechanisms of mucosal immunity permit the gut to corral the microbiota in such a way as to maximize the benefits and to minimize the danger of living in close proximity to this large microbial biomass. Toll-like receptors and Nod-like receptors, collectively referred to as pattern recognition receptors (PRRs), recognize a variety of microbial components and, hence, play a central role in governing the interface between host and microbiota. This review examines mechanisms by which PRR-microbiota interactions are regulated so as to allow activation of host defense when necessary while preventing excessive inflammation, which can have a myriad of negative consequences for the host. Analysis of published studies performed in human subjects and a variety of murine disease models reveals the central theme that PRRs play a key role in maintaining a healthful stable relationship between the intestine and its microbiota. In contrast, although select genetic ablations of PRR signaling may protect against some chronic diseases, the overriding theme of studies performed to date is that perturbations of PRR-microbiota interactions are more likely to promote disease states associated with inflammation.


Gastrointestinal Tract/microbiology , Metagenome/physiology , Toll-Like Receptors/physiology , Animals , Colitis/microbiology , Colitis/physiopathology , Escherichia coli/physiology , Gastrointestinal Neoplasms/physiopathology , Gastrointestinal Tract/immunology , Homeostasis/physiology , Humans , Metabolic Diseases/microbiology , Metabolic Diseases/physiopathology , Nod Signaling Adaptor Proteins/physiology , Receptors, Pattern Recognition/physiology , Signal Transduction/physiology
10.
Gut ; 61(3): 373-84, 2012 Mar.
Article En | MEDLINE | ID: mdl-21646247

BACKGROUND: The extent to which numerous strains of genetically engineered mice, including mice lacking Toll-like receptor 5 (T5KO), display colitis is environment dependent. Gut microbiota underlie much of the variation in phenotype. Accordingly, embryonic rederivation of T5KO mice ameliorated their spontaneous colitis despite only partially correcting elevated proinflammatory gene expression. It was postulated that endogenous anti-inflammatory pathways mediated the absence of overt inflammation in these mice when their gut microbiota were reset. Consequently, it was hypothesised that neutralisation of the anti-inflammatory cytokine interleukin 10 (IL-10) might induce uniform colitis in T5KO mice, and thus provide a practical means to study mechanisms underlying their inflammation. METHODS: Two distinct strains of non-colitic T5KO mice, as well as mice lacking MyD88, Toll-like receptor 4 (TLR4), IL-1 receptor (IL-1R) and various double knockouts (DKOs) were treated weekly for 4 weeks with 1 mg/mouse of IL-10 receptor neutralising antibody (IL-10R mAb) and colitis assayed 1 week later. The composition of the caecal microbiota was determined by 454 pyrosequencing of 16S rRNA genes. RESULTS: Anti-IL-10R mAb treatment led to severe uniform intestinal inflammation in both strains of T5KO mice. Such neutralisation of IL-10 signalling did not cause colitis in wild-type littermates nor mice lacking TLR4, MyD88 or IL-1R. The susceptibility of T5KO mice to this colitis model was not rescued by absence of TLR4 in that T4/T5 DKO mice displayed severe colitis in response to anti-IL-10R mAb treatment. IL-1ß signalling was crucial for this colitis model in that IL-1R/T5 DKOs were completely protected from colitis in response to IL-10R mAb treatment. Lastly, it was observed that blockade of IL-10R function was associated with changes in the composition of gut microbiota, which were observed in mice that were susceptible and resistant to IL-10R mAb-induced colitis. CONCLUSION: Regardless of whether they harbour a colitogenic microbiota, loss of TLR5 predisposes mice to colitis triggered by immune dysregulation via an IL-1ß-dependent pathway.


Colitis, Ulcerative/immunology , Interleukin-1beta/physiology , Toll-Like Receptor 5/deficiency , Animals , Antibodies, Monoclonal/immunology , Cecum/microbiology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling/methods , Male , Metagenome/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88 , Receptors, Interleukin-10/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 5/immunology
11.
J Inflamm (Lond) ; 8(1): 2, 2011 Feb 11.
Article En | MEDLINE | ID: mdl-21314942

BACKGROUND: Toll-like receptor-4 (TLR4) triggers inflammatory signaling in response to microbial lipoploysaccharide. It has been reported that loss of TLR4 protected against saturated fat-induced inflammation and insulin resistance. It is not known whether loss of TLR4 function offers protection against trans fat (TF) induced obesity, inflammation, and insulin resistance. We investigated whether mice with loss of function mutation in TLR4 were resistant to TF-induced pathologies such as obesity, inflammation, hyperglycemia, and hyperinsulinemia. METHODS: C57BL/6j and C57BL/10 mice were cross bred to generate TLR4 mutant and wild type (WT). TLR4 mutant (n = 12) and WT (n = 12) mice were fed either low fat (LF) (13.5% fat energy) or high TF diets (60% fat energy) for 12 weeks. In vitro experiments were conducted on mouse macrophage cells (RAW 264.7 and J774A.1) to investigate whether elaidic (trans 18:1) or oleic acid (cis 18:1) would upregulate inflammatory markers. RESULTS: TLR4 mutant mice were ~26.4% heavier than WT mice. In both genotypes, mice that received TF diet were significantly heavier than those mice that received LF diet (P < 0.01). TLR4 mutant mice compared to WT mice had significantly higher fasting blood glucose, serum insulin, insulin resistance, serum leptin, and serum cholesterol when they received TF diet (P < 0.05). No upregulation of iNOS or COX2 in response to either elaidic or oleic acid in macrophage cells was observed. CONCLUSIONS: Loss of function mutation in TLR4 not only did not protect mice from TF-induced obesity, hyperglycemia, hyperinsulinemia, and hypercholesterolemia but also exacerbated the above pathologies suggesting that functional TLR4 is necessary in attenuating TF-induced deleterious effects. It is likely that TF induces pathologies through pathways independent of TLR4.

12.
Eur J Immunol ; 40(12): 3528-34, 2010 Dec.
Article En | MEDLINE | ID: mdl-21072873

The fact that some TLR-based vaccine adjuvants maintain function in TLR-deficient hosts highlights that their mechanism of function remains incompletely understood. Thus, we examined the ability of flagellin to induce cytokines and elicit/promote murine antibody responses upon deletion of the flagellin receptors TLR5 and/or NLRC4 (also referred to as IPAF) using a prime/boost regimen. In TLR5-KO mice, flagellin failed to induce NF-κB-regulated cytokines such as keratinocyte-derived chemokine (CXCL1) but induced WT levels of the inflammasome cytokine IL-18 (IL-1F4). Conversely, in NLRC4-KO mice, flagellin induced keratinocyte-derived chemokine, but not IL-18, whereas TLR5/NLRC4-DKO lacked induction of all cytokines measured. Flagellin/ovalbumin treatment resulted in high-antibody titers to both flagellin and ovalbumin in WT, TLR5-KO and DKO mice but did not elicit antibodies to either in TLR5/NLRC4-DKO mice. Thus, flagellin's ability to elicit/promote humoral immunity requires a germ-line-encoded receptor capable of recognizing this molecule. Such promotion of adaptive immunity can be effectively driven by either TLR5-mediated activation of NF-κB or NLRC4-mediated activation of the inflammasome.


Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Flagellin/administration & dosage , Immunity, Humoral , Macrophages/metabolism , Toll-Like Receptor 5/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Cell Line , Chemokine CXCL1/metabolism , Flagellin/pharmacology , Host-Pathogen Interactions , Immunity, Humoral/genetics , Immunity, Innate , Immunization , Inflammasomes , Interleukin-18/genetics , Interleukin-18/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/immunology
13.
Science ; 328(5975): 228-31, 2010 Apr 09.
Article En | MEDLINE | ID: mdl-20203013

Metabolic syndrome is a group of obesity-related metabolic abnormalities that increase an individual's risk of developing type 2 diabetes and cardiovascular disease. Here, we show that mice genetically deficient in Toll-like receptor 5 (TLR5), a component of the innate immune system that is expressed in the gut mucosa and that helps defend against infection, exhibit hyperphagia and develop hallmark features of metabolic syndrome, including hyperlipidemia, hypertension, insulin resistance, and increased adiposity. These metabolic changes correlated with changes in the composition of the gut microbiota, and transfer of the gut microbiota from TLR5-deficient mice to wild-type germ-free mice conferred many features of metabolic syndrome to the recipients. Food restriction prevented obesity, but not insulin resistance, in the TLR5-deficient mice. These results support the emerging view that the gut microbiota contributes to metabolic disease and suggest that malfunction of the innate immune system may promote the development of metabolic syndrome.


Bacterial Physiological Phenomena , Immunity, Innate , Intestines/microbiology , Metabolic Syndrome/etiology , Toll-Like Receptor 5/metabolism , Animals , Blood Glucose/analysis , Body Fat Distribution , Body Weight , Caloric Restriction , Dietary Fats/administration & dosage , Female , Germ-Free Life , Hyperphagia/etiology , Insulin Resistance , Intestinal Mucosa/immunology , Male , Metabolic Syndrome/immunology , Metabolic Syndrome/microbiology , Mice , Mice, Knockout , Obesity/etiology , Obesity/immunology , Obesity/microbiology , Obesity/prevention & control , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics
14.
J Exp Med ; 206(10): 2179-89, 2009 Sep 28.
Article En | MEDLINE | ID: mdl-19737864

Abnormal expression of CEACAM6 is observed at the apical surface of the ileal epithelium in Crohn's disease (CD) patients, and CD ileal lesions are colonized by pathogenic adherent-invasive Escherichia coli (AIEC). We investigated the ability of AIEC reference strain LF82 to colonize the intestinal mucosa and to induce inflammation in CEABAC10 transgenic mice expressing human CEACAMs. AIEC LF82 virulent bacteria, but not nonpathogenic E. coli K-12, were able to persist in the gut of CEABAC10 transgenic mice and to induce severe colitis with reduced survival rate, marked weight loss, increased rectal bleeding, presence of erosive lesions, mucosal inflammation, and increased proinflammatory cytokine expression. The colitis depended on type 1 pili expression by AIEC bacteria and on intestinal CEACAM expression because no sign of colitis was observed in transgenic mice infected with type 1 pili-negative LF82-Delta fimH isogenic mutant or in wild-type mice infected with AIEC LF82 bacteria. These findings strongly support the hypothesis that in CD patients having an abnormal intestinal expression of CEACAM6, AIEC bacteria via type 1 pili expression can colonize the intestinal mucosa and induce gut inflammation. Thus, targeting AIEC adhesion to gut mucosa represents a new strategy for clinicians to prevent and/or to treat ileal CD.


Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Colitis/etiology , Crohn Disease/microbiology , Escherichia coli/pathogenicity , Gastrointestinal Tract/microbiology , Adhesins, Escherichia coli/physiology , Animals , Bacterial Adhesion , Fimbriae Proteins/physiology , GPI-Linked Proteins , Humans , Mice , Mice, Transgenic
15.
Inflamm Bowel Dis ; 14(8): 1051-60, 2008 Aug.
Article En | MEDLINE | ID: mdl-18338780

BACKGROUND: Ileal lesions in Crohn's disease patients are colonized by pathogenic adherent-invasive Escherichia coli (AIEC) that harbor various virulence factors involved in adhesion to and invasion of intestinal epithelial cultured cells. We investigated in a mouse model of colonic inflammation the behavior of virulent AIEC reference bacteria LF82 compared to that of nonflagellated LF82 mutants. METHODS: BALBc/J mice with intact or dextran sulfate sodium (DSS)-injured colon were orally challenged daily with 10(8) bacteria. The severity of colitis was assessed by determining disease activity index, colonic histological score, and myeoloperoxidase activity. Flagellin receptor and cytokine expression was measured by reverse-transcriptase polymerase chain reaction (RT-PCR) in colonic tissue. RESULTS: In contrast to nonpathogenic E. coli, virulent LF82 bacteria exacerbated colitis in DSS-treated mice, substantially reducing survival rate, greatly lowering stool consistency, inducing marked weight loss and increased rectal bleeding, and significantly increasing erosive lesions and mucosal inflammation. Nonflagellated LF82 mutants behaved like nonpathogenic E. coli K-12. Interestingly, oral infection with LF82 virulent bacteria, but not with a nonvirulent LF82 mutant, induced a 7.0-fold increase in the levels of TLR5 and a 3.1-fold increase in those of ipaf mRNA, which encode respectively membrane and cytosolic receptors involved in the recognition of flagellin. Hence, a 5.6-fold increase in IL-1beta and a 5.3-fold increase in mRNA of IL-6 were observed in mice challenged with AIEC LF82 bacteria. CONCLUSIONS: Crohn's disease-associated virulent AIEC LF82 bacteria, via expression of flagella, are able to potentiate an inflammatory mucosal immune response involving increased expression of TLR5 and IPAF flagellin receptors.


Colitis/microbiology , Crohn Disease/microbiology , Escherichia coli/pathogenicity , Flagellin/immunology , Gene Expression Regulation, Bacterial , Animals , Apoptosis Regulatory Proteins/biosynthesis , Calcium-Binding Proteins/biosynthesis , Colitis/immunology , Crohn Disease/immunology , Disease Models, Animal , Gene Expression , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Signal Transduction , Toll-Like Receptor 5/biosynthesis
16.
J Clin Invest ; 117(6): 1566-74, 2007 Jun.
Article En | MEDLINE | ID: mdl-17525800

The ileal mucosa of Crohn disease (CD) patients is abnormally colonized by adherent-invasive E. coli (AIEC) that are able to adhere to and invade intestinal epithelial cells. Here, we show that CD-associated AIEC strains adhere to the brush border of primary ileal enterocytes isolated from CD patients but not controls without inflammatory bowel disease. AIEC adhesion is dependent on type 1 pili expression on the bacterial surface and on carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) expression on the apical surface of ileal epithelial cells. We report also that CEACAM6 acts as a receptor for AIEC adhesion and is abnormally expressed by ileal epithelial cells in CD patients. In addition, our in vitro studies show that there is increased CEACAM6 expression in cultured intestinal epithelial cells after IFN-gamma or TNF-alpha stimulation and after infection with AIEC bacteria, indicating that AIEC can promote its own colonization in CD patients.


Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Crohn Disease/immunology , Crohn Disease/microbiology , Escherichia coli/pathogenicity , Adolescent , Adult , Antigens, CD/genetics , Bacterial Adhesion , Base Sequence , Case-Control Studies , Cell Adhesion Molecules/genetics , Cell Line , Female , GPI-Linked Proteins , Humans , Ileum/immunology , Ileum/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Microvilli/immunology , Microvilli/microbiology , Middle Aged , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
...