Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










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
8.
J Nutr ; 147(5): 770-780, 2017 05.
Article in English | MEDLINE | ID: mdl-28356436

ABSTRACT

Background: The consumption of a Western-style diet (WSD) and high fructose intake are risk factors for metabolic diseases. The underlying mechanisms are largely unclear.Objective: To unravel the mechanisms by which a WSD and fructose promote metabolic disease, we investigated their effects on the gut microbiome and barrier function.Methods: Adult female C57BL/6J mice were fed a sugar- and fat-rich WSD or control diet (CD) for 12 wk and given access to tap water or fructose-supplemented water. The microbiota was analyzed with the use of 16S rRNA gene sequencing. Barrier function was studied with the use of permeability tests, and endotoxin, mucus thickness, and gene expressions were measured.Results: The WSD increased body weight gain but not endotoxin translocation compared with the CD. In contrast, high fructose intake increased endotoxin translocation 2.6- and 3.8-fold in the groups fed the CD + fructose and WSD + fructose, respectively, compared with the CD group. The WSD + fructose treatment also induced a loss of mucus thickness in the colon (-46%) and reduced defensin expression in the ileum and colon. The lactulose:mannitol ratio in the WSD + fructose mice was 1.8-fold higher than in the CD mice. Microbiota analysis revealed that fructose, but not the WSD, increased the Firmicutes:Bacteroidetes ratio by 88% for CD + fructose and 63% for WSD + fructose compared with the CD group. Bifidobacterium abundance was greater in the WSD mice than in the CD mice (63-fold) and in the WSD + fructose mice than in the CD + fructose mice (330-fold).Conclusions: The consumption of a WSD or high fructose intake differentially affects gut permeability and the microbiome. Whether these differences are related to the distinct clinical outcomes, whereby the WSD primarily promotes weight gain and high fructose intake causes barrier dysfunction, needs to be investigated in future studies.


Subject(s)
Bacteria/drug effects , Diet, Western , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Fructose/pharmacology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Animals , Bacteria/growth & development , Bacteroidetes/drug effects , Bacteroidetes/growth & development , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Colon/drug effects , Colon/metabolism , Defensins/metabolism , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/metabolism , Dietary Fats/administration & dosage , Dietary Supplements , Drinking Water/administration & dosage , Endotoxins/metabolism , Feeding Behavior , Female , Firmicutes/drug effects , Firmicutes/growth & development , Fructose/administration & dosage , Fructose/metabolism , Ileum/drug effects , Ileum/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , Mucus/metabolism , Permeability , RNA, Ribosomal, 16S , Weight Gain
9.
Am J Clin Nutr ; 105(1): 127-135, 2017 01.
Article in English | MEDLINE | ID: mdl-28049662

ABSTRACT

BACKGROUND: Obesity and associated metabolic disorders are related to impairments of the intestinal barrier. OBJECTIVE: We examined lactulose:mannitol (Lac:Man) permeability in obese individuals with and without liver steatosis undergoing a weight-reduction program to test whether an effective weight-loss program improves gut barrier function and whether obese patients with or without liver steatosis differ in this function. DESIGN: Twenty-seven adult, nondiabetic individuals [mean ± SD body mass index (BMI; in kg/m2): 43.7 ± 5.2; 78% with moderate or severe liver steatosis] were included in the follow-up intervention study (n = 13 by month 12). All patients reduced their weight to a mean ± SD BMI of 36.4 ± 5.1 within 12 mo. We assessed barrier functions by the oral Lac:Man and the fecal zonulin tests. Insulin resistance was assessed by the homeostatic model assessment index (HOMA), and liver steatosis by sonography and the fatty liver index (FLI). RESULTS: The Lac:Man ratio and circulating interleukin (IL) 6 concentration decreased during intervention from 0.080 (95% CI: 0.073, 0.093) to 0.027 (95% CI: 0.024, 0.034; P < 0.001) and from 4.2 ± 1.4 to 2.8 ± 1.6 pg/mL (P < 0.01), respectively. At study start, the Lac:Man ratio was higher in patients with moderate or severe steatosis than in those without any steatosis (P < 0.001). The Lac:Man ratio tended to correlate with HOMA (ρ = 0.55, P = 0.052), which correlated with FLI (ρ = 0.75, P < 0.01). A multiple-regression analysis led to a final model explaining FLI best through BMI, waist circumference, and the Lac:Man ratio. CONCLUSIONS: Intestinal permeability is increased in obese patients with steatosis compared with obese patients without. The increased permeability fell to within the previously reported normal range after weight reduction. The data suggest that a leaky gut barrier is linked with liver steatosis and could be a new target for future steatosis therapies. This trial was registered at clinicaltrials.gov as NCT01344525.


Subject(s)
Body Mass Index , Fatty Liver/physiopathology , Insulin Resistance , Intestinal Absorption , Intestines/physiopathology , Obesity/therapy , Weight Loss/physiology , Adult , Body Weight , Cholera Toxin/metabolism , Fatty Liver/etiology , Female , Follow-Up Studies , Haptoglobins , Humans , Interleukin-6/blood , Lactulose/metabolism , Male , Mannitol/metabolism , Middle Aged , Models, Biological , Obesity/blood , Obesity/physiopathology , Permeability , Protein Precursors , Waist Circumference , Young Adult
10.
Tissue Barriers ; 4(3): e1208468, 2016.
Article in English | MEDLINE | ID: mdl-27583194

ABSTRACT

The intestinal barrier is gaining increasing attention because it is related to intestinal homeostasis and disease. Different parameters have been used in the past to assess intestinal barrier functions in experimental studies; however most of them are poorly defined in healthy mice. Here, we compared a number of barrier markers in healthy mice, established normal values and correlations. In 48 mice (24 C57BL/6J, 24 BALB/cJ background), we measured mucus thickness, and expression of mucin-2, α-defensin-1 and -4, zonula occludens-1, occludin, junctional adhesion molecule-A, claudin-1, 2 and -5. We also analyzed claudin-3 and fatty acid binding protein-2 in urine and plasma, respectively. A higher expression of mucin-2 protein was found in the colon compared to the ileum. In contrast, the α-defensins-1 and -4 were expressed almost exclusively in the ileum. The protein expression of the tight junction molecules claudin-1, occludin and zonula occludens-1 did not differ between colon and ileum, although some differences occurred at the mRNA level. No age- or gender-related differences were found. Differences between C57BL/6J and BALB/cJ mice were found for α-defensin-1 and -4 mRNA expression, and for urine and plasma marker concentrations. The α-defensin-1 mRNA correlated with claudin-5 mRNA, whereas α-defensin-4 mRNA correlated with claudin-3 concentrations in urine. In conclusion, we identified a number of murine intestinal barrier markers requiring tissue analyses or measurable in urine or plasma. We provide normal values for these markers in mice of different genetic background. Such data might be helpful for future animal studies in which the intestinal barrier is of interest.


Subject(s)
Intestinal Mucosa/metabolism , Mucins/metabolism , Tight Junction Proteins/metabolism , alpha-Defensins/metabolism , Animals , Capillary Permeability , Colon/growth & development , Colon/metabolism , Fatty Acid-Binding Proteins/blood , Fatty Acid-Binding Proteins/urine , Female , Ileum/growth & development , Ileum/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucins/genetics , Species Specificity , Tight Junction Proteins/genetics , alpha-Defensins/genetics
11.
Int J Med Microbiol ; 306(5): 328-333, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27432516

ABSTRACT

Data from literature suggests that laboratory mice are often overfed and malnourished. This might have several reasons, including: (i) we usually offer an ad libitum diet, which is not the natural way of feeding for a wild mouse; (ii) many commercial diets we use contain rather high amounts of carbohydrates, particularly of sugars, and low amounts of fat; and (iii) laboratory mice live in a warm and constricted environment in which energy expenditure is lower than in the wild. Such selective or global overfeeding in laboratory mice, which resembles the widespread overfeeding in humans, although it does not always result in overweight, likely affects a number of outcome variables analyzed in laboratory mice, such as microbiota composition and function, metabolic alterations, longevity, intestinal permeability and inflammation. Therefore, a careful selection of experimental diets and their way of administration, as well as detailed documentation, is mandatory in order to understand and compare scientific data obtained from different mouse experiments.


Subject(s)
Diet/methods , Disease Models, Animal , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Animals , Mice , Research Design/standards
12.
Int J Med Microbiol ; 306(5): 343-355, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27053239

ABSTRACT

The intestinal microbiota is involved in many physiological processes and it is increasingly recognized that differences in community composition can influence the outcome of a variety of murine models used in biomedical research. In an effort to describe and account for the variation in intestinal microbiota composition across the animal facilities of participating members of the DFG Priority Program 1656 "Intestinal Microbiota", we performed a survey of C57BL/6J mice from 21 different mouse rooms/facilities located at 13 different institutions across Germany. Fresh feces was sampled from five mice per room/facility using standardized procedures, followed by extraction and 16S rRNA gene profiling (V1-V2 region, Illumina MiSeq) at both the DNA and RNA (reverse transcribed to cDNA) level. In order to determine the variables contributing to bacterial community differences, we collected detailed questionnaires of animal husbandry practices and incorporated this information into our analyses. We identified considerable variation in a number of descriptive aspects including the proportions of major phyla, alpha- and beta diversity, all of which displayed significant associations to specific aspects of husbandry. Salient findings include a reduction in alpha diversity with the use of irradiated chow, an increase in inter-individual variability (beta diversity) with respect to barrier access and open cages and an increase in bacterial community divergence with time since importing from a vendor. We further observe a high degree of facility-level individuality, which is likely due to each facility harboring its own unique combination of multiple varying attributes of animal husbandry. While it is important to account and control for such differences between facilities, the documentation of such diversity may also serve as a valuable future resource for investigating the origins of microbial-driven host phenotypes.


Subject(s)
Animal Husbandry/methods , Feces/microbiology , Gastrointestinal Microbiome , Animals , Cluster Analysis , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Germany , Male , Mice, Inbred C57BL , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Surveys and Questionnaires
13.
Dig Dis Sci ; 61(3): 737-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26520109

ABSTRACT

BACKGROUND: Intestinal permeability is thought to be of major relevance for digestive and nutrition-related diseases, and therefore has been studied in numerous mouse models of disease. However, it is unclear which tools are the preferable ones, and how normal values should be defined. AIMS: To compare different in vivo permeability tests in healthy mice of commonly used genetic backgrounds. METHODS: We assessed the intestinal barrier in male and female C57BL/6J and BALB/cJ mice of different ages, using four orally administered permeability markers, FITC-dextran 4000 (FITC-D4000) and ovalbumin (OVA) measured in plasma, and polyethylene glycol (PEG) and lactulose/mannitol (Lac/Man) measured in urine, and by assessing lipopolysaccharide (LPS) in portal vein plasma. RESULTS: After gavage, FITC-D4000, OVA, Lac/Man, and PEG400, but not PEG4000, were detectable in plasma or urine. Female mice tended to have a higher permeability according to the FITC-D4000, OVA, and PEG400 tests, but the Lac/Man ratio was higher in males. No significant differences between the two mouse strains of young and old mice were observed except for mannitol recovery, which was higher in BALB/cJ mice compared to C57BL/6J mice (p < 0.05). Virtually no LPS was detected in healthy mice. For all markers, normal values have been defined based on 5th-95th percentile ranges of our data. CONCLUSION: Selected oral permeability tests, such as FITC-D4000, OVA, PEG400, and Lac/Man, as well as LPS measurements in portal vein plasma, could be suitable for the evaluation of the intestinal barrier in mice, if used in a standardized way.


Subject(s)
Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Intestinal Mucosa/metabolism , Lactulose/metabolism , Lipopolysaccharides/metabolism , Mannitol/metabolism , Ovalbumin/metabolism , Permeability , Polyethylene Glycols/metabolism , Animals , Dextrans/blood , Female , Fluorescein-5-isothiocyanate/metabolism , Lactulose/urine , Lipopolysaccharides/blood , Male , Mannitol/urine , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/blood , Portal Vein
14.
Eur J Nutr ; 52(2): 527-35, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22543623

ABSTRACT

PURPOSE: As a diet rich in fructose and an impaired intestinal barrier function have been proposed to be risk factors for the development of non-alcoholic fatty liver disease (NAFLD), the aim of the present pilot study was to determine whether a dietary intervention focusing on a reduction of fructose intake (-50 % in comparison with baseline) has a beneficial effect on liver status. METHODS: A total of 15 patients with NAFLD were enrolled in the study of which 10 finished the study. Fructose and total nutrient intake were assessed using a diet history. At baseline and after 6 months liver status and markers of intestinal barrier function as well as plasminogen activator inhibitor (PAI-) 1 concentration were determined in plasma. RESULTS: Hepatic lipid content and transaminases in plasma as well as body mass index and some parameters of glucose metabolism (e.g., fasting plasma insulin) were significantly lower at the end of the intervention when compared to baseline. Whereas the dietary intervention had no effect on the prevalence of bacterial overgrowth, orocecal transit time and the intestinal permeability or blood ethanol levels endotoxin and PAI-1 concentration in plasma were significantly lower at the end of 6 months intervention period than at baseline. CONCLUSIONS: Taken together, our results indicate that a dietary intervention focusing only on one dietary parameter like fructose may help to decrease intrahepatic fat content of NAFLD patients.


Subject(s)
Fatty Liver/diet therapy , Feeding Behavior , Liver/metabolism , Weight Loss , Adult , Blood Glucose/analysis , Body Mass Index , Energy Intake , Ethanol/blood , Female , Fructose/administration & dosage , Humans , Insulin/blood , Intra-Abdominal Fat/metabolism , Lipids/blood , Magnetic Resonance Imaging , Male , Middle Aged , Motor Activity , Non-alcoholic Fatty Liver Disease , Pilot Projects , Plasminogen Activator Inhibitor 1/blood , Transaminases/blood
15.
Dig Dis Sci ; 57(7): 1932-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22427130

ABSTRACT

BACKGROUND: A role of an altered dietary pattern (e.g., a diet rich in sugar) but also alterations at the level of the intestinal barrier have repeatedly been discussed to be involved in the development and progression of nonalcoholic fatty liver disease (NAFLD). AIMS: To determine if the nutritional intake, intestinal flora, and permeability and the development of NAFLD are related in humans. METHODS: Ten controls and 20 patients with NAFLD ranging from simple steatosis to steatohepatitis were included in the study. Bacterial overgrowth, orocecal transit time, and intestinal permeability were assessed. Alcohol, endotoxin, and plasminogen activator inhibitor (PAI-) 1 concentration were determined in plasma. Nutritional intake was assessed using a dietary history. RESULTS: Despite no differences in the prevalence of bacterial overgrowth and in the orocecal transit time, intestinal permeability, alcohol, and endotoxin levels in plasma were significantly higher in patients with NAFLD than in controls. Similar results were also found for PAI-1 plasma concentrations. Patients with NAFLD had a significantly higher intake of protein, total carbohydrates, and mono- as well as disaccharides than controls. PAI-1, endotoxin, and ALT plasma levels were positively related to total protein and carbohydrate intake. CONCLUSIONS: Taken together, our results indicate that intestinal permeability, endogenous alcohol synthesis, and nutritional intake are markedly altered in patients with NAFLD.


Subject(s)
Ethanol/blood , Fatty Liver/blood , Fatty Liver/physiopathology , Intestines/physiology , Nutritional Status/physiology , Adult , Case-Control Studies , Dietary Carbohydrates/pharmacology , Dietary Proteins/pharmacology , Disease Progression , Endotoxins/blood , Female , Humans , Intestines/microbiology , Male , Non-alcoholic Fatty Liver Disease , Permeability/drug effects , Plasminogen Activator Inhibitor 1/blood
16.
Br J Nutr ; 107(12): 1727-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22018861

ABSTRACT

Studies in animals and human subjects indicate that gut-derived bacterial endotoxins may play a critical role in the development of non-alcoholic fatty liver disease (NAFLD). In the present study, we investigated if the liver is also sensitised by other microbial components during the onset of fructose-induced steatosis in a mouse model. C57BL/6 mice were either fed with 30 % fructose solution or tap water (control) with or without antibiotics for 8 weeks. Expression of toll-like receptors (TLR)1-9, TNF-α, inducible NO synthase (iNOS), myeloid differentiation factor 88 (MyD88) and number of F4/80 positive cells in the liver were assessed. Occludin protein, DNA of microbiota in the small and large intestine and retinol binding protein 4 (RBP4) in plasma were analysed using Western blot, DNA fingerprinting and ELISA, respectively. F4/80 positive cells were determined by immunohistochemistry. The accumulation of TAG found in the livers of fructose-fed mice was associated with a significant induction of TLR 1-4 and 6-8. Plasma RBP4 concentration and hepatic mRNA expression levels of TNF-α, iNOS, MyD88 and number of F4/80 positive cells of fructose-fed animals were significantly higher than those of controls; however, these effects of fructose were attenuated in antibiotic-treated mice. Whereas protein concentration of occludin was lower in the duodenum of fructose-treated mice, no systematic alterations of microbiota were found in this part of the intestine. Taken together, these data support the hypothesis that (1) an increased intestinal translocation of microbial components and (2) an increased number of F4/80 positive cells and induction of several TLR and dependent pathways (e.g. MyD88 and iNOS) may be involved in the onset of fructose-induced NAFLD.


Subject(s)
Bacterial Translocation , Fatty Liver/metabolism , Fructose/adverse effects , Intestines/microbiology , Liver/metabolism , Toll-Like Receptors/metabolism , Triglycerides/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Differentiation/metabolism , Disease Models, Animal , Duodenum/metabolism , Duodenum/microbiology , Fatty Liver/chemically induced , Fatty Liver/microbiology , Membrane Proteins/metabolism , Metagenome , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Non-alcoholic Fatty Liver Disease , Occludin , RNA, Messenger/metabolism , Retinol-Binding Proteins, Plasma/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
J Lipid Res ; 51(12): 3414-24, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20847296

ABSTRACT

Fructose intake is being discussed as a key dietary factor in the development of nonalcoholic fatty liver disease (NAFLD). Bile acids have been shown to modulate energy metabolism. We tested the effects of bile acids on fructose-induced hepatic steatosis. In C57BL/6J mice treated with a combination of chenodeoxycholic acid and cholic acid (100 mg/kg body weight each) while drinking water or a 30% fructose solution for eight weeks and appropriate controls, markers of hepatic steatosis, portal endotoxin levels, and markers of hepatic lipogenesis were determined. In mice concomitantly treated with bile acids, the onset of fructose-induced hepatic steatosis was markedly attenuated compared to mice only fed fructose. The protective effects of the bile acid treatment were associated with a downregulation of tumor necrosis factor (TNF)α, sterol regulatory element-binding protein (SREBP)1, FAS mRNA expression, and lipid peroxidation in the liver, whereas hepatic farnesoid X receptor (FXR) or short heterodimer partner (SHP) protein concentration did not differ between groups fed fructose. Rather, bile acid treatment normalized occludin protein concentration in the duodenum, portal endotoxin levels, and markers of Kupffer cell activation to the level of water controls. Taken together, these data suggest that bile acids prevent fructose-induced hepatic steatosis in mice through mechanisms involving protection against the fructose-induced translocation of intestinal bacterial endotoxin.


Subject(s)
Bile Acids and Salts/metabolism , Dietary Sucrose/metabolism , Fatty Liver/chemically induced , Fatty Liver/metabolism , Fructose/metabolism , Animals , Duodenum/metabolism , Endotoxins/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Lipid Peroxidation/drug effects , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Occludin , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Am J Physiol Gastrointest Liver Physiol ; 298(3): G335-44, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19713474

ABSTRACT

Elevated dietary fructose intake, altered intestinal motility, and barrier function may be involved in the development of nonalcoholic fatty liver disease (NAFLD). Because intestinal motility and permeability are also regulated through the bioavailability of serotonin (5-HT), we assessed markers of hepatic injury in serotonin reuptake transporter knockout (SERT(-/-)) and wild-type mice chronically exposed to different monosaccharide solutions (30% glucose or fructose solution) or water for 8 wk. The significant increase in hepatic triglyceride, TNF-alpha, and 4-hydroxynonenal adduct as well as portal endotoxin levels found in fructose-fed mice was associated with a significant decrease of SERT and the tight-junction occludin in the duodenum. Similar effects were not found in mice fed glucose. In contrast, in SERT(-/-) mice fed glucose, portal endotoxin levels, concentration of occludin, and indices of hepatic damage were similar to those found in wild-type and SERT(-/-) mice fed fructose. In fructose-fed mice treated with a 5-HT3 receptor antagonist, hepatic steatosis was significantly attenuated. Our data suggest that a loss of intestinal SERT is a critical factor in fructose-induced impairment of intestinal barrier function and subsequently the development of steatosis.


Subject(s)
Fatty Liver/chemically induced , Fatty Liver/metabolism , Fructose/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Aldehydes/metabolism , Animals , Body Weight/drug effects , Caco-2 Cells , Duodenum/drug effects , Duodenum/metabolism , Endotoxins/blood , Endotoxins/metabolism , Fatty Liver/pathology , Fructose/administration & dosage , Gastrointestinal Motility/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Glucose/administration & dosage , Glucose/pharmacology , Humans , Indoles/pharmacology , Intestinal Absorption/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neutrophils/pathology , Occludin , Organ Size/drug effects , Permeability/drug effects , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Triglycerides/metabolism , Tropisetron , Tumor Necrosis Factor-alpha/genetics
19.
J Nutr ; 139(3): 482-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19126670

ABSTRACT

Acute and chronic consumption of alcohol can cause increased intestinal permeability and bacterial overgrowth, thereby increasing portal endotoxin levels. This barrier impairment subsequently leads to an activation of hepatic Kupffer cells and increased release of reactive oxygen species as well as of tumor necrosis factor-alpha (TNFalpha). Recent studies have suggested that cinnamon extract may have antiinflammatory effects. In the present study, the protective effects of an alcoholic extract of cinnamon bark was assessed in a mouse model of acute alcohol-induced steatosis and in RAW 264.7 macrophages, used here as a model of Kupffer cells. Acute alcohol ingestion caused a >20-fold increase in hepatic lipid accumulation. Pretreatment with cinnamon extract significantly reduced the hepatic lipid accumulation. This protective effect of cinnamon extract was associated with an inhibition of the induction of the myeloid differentiation primary response gene (MyD) 88, inducible nitric oxide (NO) synthase (iNOS), and plasminogen activator inhibitor 1 mRNA expression found in livers of alcohol-treated animals. In vitro prechallenge with cinnamon extract suppressed lipopolysaccharide (LPS)-induced MyD88, iNOS, and TNFalpha expression as well as NO formation almost completely. Furthermore, LPS treatment of RAW 264.7 macrophages further resulted in degradation of inhibitor kappaB; this effect was almost completely blocked by cinnamon extract. Taken together, our data show that an alcohol extract of cinnamon bark may protect the liver from acute alcohol-induced steatosis through mechanisms involving the inhibition of MyD88 expression.


Subject(s)
Cinnamomum zeylanicum/chemistry , Ethanol/toxicity , Fatty Liver/chemically induced , Fatty Liver/prevention & control , Plant Extracts/pharmacology , Animals , Cell Line , Gene Expression Regulation/drug effects , Macrophages , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Plant Bark/chemistry , Plant Extracts/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serpin E2 , Serpins/genetics , Serpins/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Alcohol Clin Exp Res ; 33(3): 557-62, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19120061

ABSTRACT

BACKGROUND: Chronic alcohol abuse increases both intestinal bacterial overgrowth and intestinal permeability to macromolecules. Intestinal permeability of endotoxin, a component of the outer cell membrane of Gram-negative bacteria, plays a crucial role in the development of alcohol-induced liver disease (ALD). As impaired bile flow leads to endotoxemia and the bile component phosphatidylcholine (PC) is therapeutically active in ALD, we tested the hypothesis that conjugated primary bile salts (CPBS) and PC inhibit ethanol-enhanced transepithelial permeability of endotoxin and the subsequent transepithelial activation of human leukocytes. METHODS: For this purpose, we used a model in which intestinal epithelial cells (Caco-2) were basolaterally cocultivated with mononuclear leukocytes. Cells were challenged apically with endotoxin from Escherichia coli K12 and were incubated with or without the addition of CPBS (1.5 mM), PC (0.38 mM), pooled human bile (2%) in combination with ethanol (0 to 66 mM). RESULTS: Ethanol decreased integrity of intestinal epithelial cell monolayer and enhanced transepithelial permeability of endotoxin. Both the transepithelial permeability of endotoxin and the transepithelial stimulation of leukocytes were nearly completely abolished after the apical supplementation of PC with CPBS, but not by CPBS alone. Ethanol up to 66 mM was not able to reverse this effect. CONCLUSIONS: A considerable part of the therapeutic and preventive effect of PC supplementation in ALD might result from a reduction of ethanol-enhanced permeability of endotoxin through the intestinal barrier.


Subject(s)
Cell Membrane Permeability/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Intestinal Mucosa/drug effects , Lipopolysaccharides/metabolism , Phosphatidylcholines/pharmacology , Bile Acids and Salts/pharmacology , Caco-2 Cells , Coculture Techniques , Cytokines/metabolism , Electric Impedance , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Leukocytes, Mononuclear , Lymphocyte Activation/drug effects , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...