Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nutrients ; 15(2)2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36678247

ABSTRACT

Vitamin A and D deficiencies are associated with immune modulatory effects and intestinal barrier impairment. However, the underlying mechanisms remain unclear. C57BL/6J mice were fed either a diet lacking in vitamin A (VAd), vitamin D (VDd) or a control diet (CD) for 12 weeks. Gut barrier function, antimicrobial peptide (AMP) defense and regulatory pathways were assessed. VAd mice compared to CD mice showed a reduced villus length in the ileum (p < 0.01) and decreased crypt depth in the colon (p < 0.05). In both VAd- and VDd-fed mice, ileal α-defensin 5 (p < 0.05/p < 0.0001 for VAd/VDd) and lysozyme protein levels (p < 0.001/p < 0.0001) were decreased. Moreover, mRNA expression of lysozyme (p < 0.05/p < 0.05) and total cryptdins (p < 0.001/p < 0.01) were reduced compared to controls. Furthermore, matrix metalloproteinase-7 (Mmp7) mRNA (p < 0.0001/p < 0.001) as well as components of the Wnt signaling pathway were decreased. VAd- and VDd-fed mice, compared to control mice, exhibited increased expression of pro-inflammatory markers and ß-defensins in the colon. Organoid cell culture confirmed that vitamins A and D regulate AMP expression, likely through the Jak/STAT5 signaling pathway. In conclusion, our data show that vitamin A and D regulate intestinal antimicrobial peptide defense through Wnt and STAT5 signaling pathways.


Subject(s)
Muramidase , Vitamin A , Animals , Mice , STAT5 Transcription Factor , Mice, Inbred C57BL , Vitamins , Diet , Signal Transduction , RNA, Messenger/genetics , Antimicrobial Peptides
2.
Dig Dis Sci ; 67(9): 4422-4433, 2022 09.
Article in English | MEDLINE | ID: mdl-35394589

ABSTRACT

BACKGROUND: Cereals are known to trigger for wheat allergy, celiac disease and non-celiac wheat sensitivity (NCWS). Inflammatory processes and intestinal barrier impairment are suspected to be involved in NCWS, although the molecular triggers are unclear. AIMS: We were interested if different bread types influence inflammatory processes and intestinal barrier function in a mouse model of inflammatory bowel disease. METHODS: Epithelial caspase-8 gene knockout (Casp8ΔIEC) and control (Casp8fl) mice were randomized to eight groups, respectively. The groups received different diets for 28 days (gluten-free diet, gluten-rich diet 5 g%, or different types of bread at 50 g%). Breads varied regarding grain, milling and fermentation. All diets were isocaloric. RESULTS: Regardless of the diet, Casp8ΔIEC mice showed pronounced inflammation in colon compared to ileum, whereas Casp8fl mice were hardly inflamed. Casp8fl mice could tolerate all bread types. Especially yeast fermented rye and wheat bread from superfine flour but not pure gluten challenge increased colitis and mortality in Casp8ΔIEC mice. Hepatic expression of lipopolysaccharide-binding protein and colonic expression of tumor necrosis factor-α genes were inversely related to survival. The bread diets, but not the gluten-rich diet, also decreased colonic tight junction expression to variable degrees, without clear association to survival and inflammation. CONCLUSIONS: Bread components, especially those from yeast-fermented breads from wheat and rye, increase colitis and mortality in Casp8ΔIEC mice highly susceptible to intestinal inflammation, whereas control mice can tolerate all types of bread without inflammation. Yet unidentified bread components other than gluten seem to play the major role.


Subject(s)
Bread , Colitis , Animals , Mice , Colitis/chemically induced , Diet, Gluten-Free , Glutens , Inflammation , Saccharomyces cerevisiae , Secale/chemistry
3.
Front Immunol ; 12: 678360, 2021.
Article in English | MEDLINE | ID: mdl-34177920

ABSTRACT

Defects in the mucosal barrier have been associated with metabolic diseases such as obesity and non-alcoholic fatty liver disease (NAFLD). Mice fed a Western-style diet (WSD) develop obesity and are characterized by a diet-induced intestinal barrier dysfunction, bacterial endotoxin translocation and subsequent liver steatosis. To examine whether inulin or sodium butyrate could improve gut barrier dysfunction, C57BL/6 mice were fed a control diet or a WSD ± fructose supplemented with either 10% inulin or 5% sodium butyrate for 12 weeks respectively. Inulin and sodium butyrate attenuated hepatosteatitis in the WSD-induced obesity mouse model by reducing weight gain, liver weight, plasma and hepatic triglyceride level. Furthermore, supplementation with inulin or sodium butyrate induced expression of Paneth cell α-defensins and matrix metalloproteinase-7 (MMP7), which was impaired by the WSD and particularly the fructose-added WSD. Effects on antimicrobial peptide function in the ileum were accompanied by induction of ß-defensin-1 and tight junction genes in the colon resulting in improved intestinal permeability and endotoxemia. Organoid culture of small intestinal crypts revealed that the short chain fatty acids (SCFA) butyrate, propionate and acetate, fermentation products of inulin, induce Paneth cell α-defensin expression in vitro, and that histone deacetylation and STAT3 might play a role in butyrate-mediated induction of α-defensins. In summary, inulin and sodium butyrate attenuate diet-induced barrier dysfunction and induce expression of Paneth cell antimicrobials. The administration of prebiotic fiber or sodium butyrate could be an interesting therapeutic approach to improve diet-induced obesity.


Subject(s)
Butyric Acid/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Inulin/administration & dosage , Obesity/metabolism , Pore Forming Cytotoxic Proteins/biosynthesis , Prebiotics/administration & dosage , Animal Feed , Animals , Biomarkers , Dietary Supplements , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Liver/drug effects , Liver/metabolism , Mice , Obesity/drug therapy , Obesity/etiology , Permeability , Tight Junctions/metabolism
4.
Int J Med Microbiol ; 311(4): 151499, 2021 May.
Article in English | MEDLINE | ID: mdl-33864957

ABSTRACT

The gut barrier has been recognized as being of relevance in the pathogenesis of multiple different diseases ranging from inflammatory bowel disease, irritable bowel syndrome, inflammatory joint disease, fatty liver disease, and cardiometabolic disorders. The regulation of the gut barrier is, however, poorly understood. Especially, the role of food components such as sugars and complex carbohydrates has been discussed controversially in this respect. More recently, the intestinal microbiota has been proposed as an important regulator of the gut barrier. Whether the microbiota affects the barrier by its own, or whether food components such as carbohydrates mediate their effects through alterations of the microbiota composition or its metabolites, is still not clear. In this review, we will summarize the current knowledge on this topic derived from both animal and human studies and discuss data for possible clinical impact.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Animals , Carbohydrates , Diet , Humans
5.
Inflamm Bowel Dis ; 26(1): 66-79, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31276162

ABSTRACT

Crohn's disease (CD) patients can be grouped into patients suffering from ileitis, ileocolitis, jejunoileitis, and colitis. The pathophysiological mechanism underlying this regional inflammation is still unknown. Although most murine models of inflammatory bowel disease (IBD) develop inflammation in the colon, there is an unmet need for novel models that recapitulate the spontaneous and fluctuating nature of inflammation as seen in CD. Recently, mice with an intestinal epithelial cell-specific deletion for Caspase-8 (Casp8ΔIEC mice), which are characterized by cell death-driven ileitis and disrupted Paneth cell homeostasis, have been identified as a novel model of CD-like ileitis. Here we uncovered that genetic susceptibility alone is sufficient to drive ileitis in Casp8ΔIEC mice. In sharp contrast, environmental factors, such as a disease-relevant microbial flora, determine colonic inflammation. Accordingly, depending on the microbial environment, isogenic Casp8ΔIEC mice either exclusively developed ileitis or suffered from pathologies in several parts of the gastrointestinal tract. Colitis in these mice was characterized by massive epithelial cell death, leading to spread of commensal gut microbes to the extra-intestinal space and hence an aberrant activation of the systemic immunity. We further uncovered that Casp8ΔIEC mice show qualitative and quantitative changes in the intestinal microbiome associated with an altered mucosal and systemic immune response. In summary, we identified that inflammation in this murine model of CD-like inflammation is characterized by an immune reaction, presumably directed against a disease-relevant microbiota in a genetically susceptible host, with impaired mucosal barrier function and bacterial clearance at the epithelial interface.


Subject(s)
Crohn Disease/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Ileitis/microbiology , Intestinal Mucosa/microbiology , Animals , Caspase 8 , Crohn Disease/genetics , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Ileitis/genetics , Inflammation , Intestinal Mucosa/immunology , Mice
6.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G493-G507, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31411503

ABSTRACT

Genetically modified mice have been successfully used as models for inflammatory bowel diseases; however, dietary effects were poorly examined. Here, we studied the impact of particular nutrients and supplements on gut functions related to the knockout of the epithelial caspase-8 gene. Caspase-8 knockout (Casp8∆IEC) and control (Casp8fl) mice were fed for 4 wk a control diet (CD) enriched with 10% inulin (CD-Inu) or 5% sodium butyrate (CD-But) while having free access to plain water or water supplemented with 30% fructose (+F). Body weight changes, intestinal inflammation, and selected markers for barrier function and of liver steatosis were assessed. Casp8∆IEC mice developed ileocolitis accompanied by changes in intestinal barrier morphology and reduced expression of barrier-related genes such as mucin-2 (Muc2) and defensins in the ileum and Muc2 in the colon. Casp8∆IEC mice fed a CD also showed impaired body weight gain compared with Casp8fl mice, which was even more pronounced in mice receiving water supplemented with fructose. Furthermore, we observed a marked liver steatosis and inflammation in some but not all Casp8∆IEC mice under a CD, which was on average similar to that observed in control mice under a fructose-rich diet. Hepatic lipid accumulation, as well as markers of ileal barrier function, but not intestinal pathohistology or body weight loss, were attenuated by diets enriched with inulin or butyrate, especially in the absence of fructose supplementation. Our data show that ileocolitis, barrier dysfunction, and malassimilation in Caspase-8 knockout mice can be partially attenuated by oral inulin or butyrate supplementation.NEW & NOTEWORTHY Genetic mouse models for ileocolitis are important to understand inflammatory bowel disease in humans. We examined dietetic factors that might aggravate or attenuate ileocolitis and related pathologies in such a model. Deletion of the caspase-8 gene results not only in ileocolitis but also in gut barrier dysfunction, liver steatosis, and malassimilation, which can be partially attenuated by oral inulin or sodium butyrate. Our data indicate that diet modifications can contribute to disease variability and therapy.


Subject(s)
Butyric Acid/pharmacology , Caspase 8/genetics , Caspase 8/physiology , Crohn Disease/genetics , Crohn Disease/pathology , Intestinal Mucosa/pathology , Inulin/pharmacology , Animals , Body Weight/genetics , Crohn Disease/drug therapy , Diet, Western , Dietary Supplements , Female , Gastrointestinal Microbiome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics , Mucin-2/metabolism , Nutrients
7.
Eur J Nutr ; 58(5): 1933-1945, 2019 Aug.
Article in English | MEDLINE | ID: mdl-29926176

ABSTRACT

PURPOSE: Experimental liver steatosis induced by overfeeding is associated with enhanced gut permeability and endotoxin translocation to the liver. We examined the role of the gut microbiota for steatosis formation by performing the feeding experiments in mice raised under conventional and germ-free (GF) housing. METHODS: Adult wild-type and GF mice were fed a Western-style diet (WSD) or a control diet (CD), the latter combined with liquid fructose supplementation (F) or not, for 8 weeks. Markers of liver steatosis and gut permeability were measured after intervention. RESULTS: Mice fed a WSD increased body weight compared to those fed a CD (p < 0.01) under conventional, but not under GF conditions. Increased liver weight, liver-to-body-weight ratio and hepatic triglycerides observed in both the WSD and the CD + F groups, when compared with the CD group, were not apparent under GF conditions, whereas elevated plasma triglycerides were visible (p < 0.05). Wild-type mice fed a WSD or a CD + F, respectively, had thinner adherent mucus layer compared to those fed a CD (p < 0.01), whereas GF mice had always a thin mucus layer independently of the diet. GF mice fed a CD showed increased plasma levels of FITC-dextran 4000 (1.9-fold, p < 0.05) and intestinal fatty acid-binding protein-2 (2.4-fold, p < 0.05) compared with wild-type mice. CONCLUSIONS: GF housing results in an impaired weight gain and a lack of steatosis following a WSD. Also the fructose-induced steatosis, which is unrelated to body weight changes, is absent in GF mice. Thus, diet-induced experimental liver steatosis depends in multiple ways on intestinal bacteria.


Subject(s)
Diet, Western , Fructose/administration & dosage , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Disease Models, Animal , Female , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...