18 research outputs found

    Upregulated expression of ENaC in human CF nasal epithelium

    Get PDF
    AbstractCystic fibrosis (CF) is characterised by the absence of CFTR function resulting in a reduced Cl− secretion and an increase in Na+ absorption. This Na+ hyperabsorption is mediated by the human amiloride-sensitive epithelial sodium channel (ENaC), but the underlying mechanisms are still unknown. After demonstrating functional differences of the Na+ absorption in CF and non-CF epithelia in Ussing chamber experiments with human primary cultures, we compared ENaC sequences from CF and non-CF human nasal tissue (hnENaC), investigated the mRNA transcription levels via real-time PCR and studied the protein expression in Western blot analyses. We found no differences in the sequences of CF and non-CF hnENaC, but identified some polymorphisms. The real-time experiments revealed an enhanced mRNA amount of all three hnENaC subunits in CF tissue. By comparing the two groups on the protein level, we observed differences in the abundance of the Na+ channel. While the α- and β-hnENaC protein amount was increased in CF tissue the γ-hnENaC was decreased. We conclude that the Na+ hyperabsorption in CF is not caused by mutations in hnENaC, but by an increase in the transcription of the hnENaC subunits. This could be induced by a disturbed regulation of the channel in CF

    C57Bl/6 N mice on a western diet display reduced intestinal and hepatic cholesterol levels despite a plasma hypercholesterolemia

    No full text
    Abstract Background Small intestine and liver greatly contribute to whole body lipid, cholesterol and phospholipid metabolism but to which extent cholesterol and phospholipid handling in these tissues is affected by high fat Western-style obesogenic diets remains to be determined. Methods We therefore measured cholesterol and phospholipid concentration in intestine and liver and quantified fecal neutral sterol and bile acid excretion in C57Bl/6 N mice fed for 12 weeks either a cholesterol-free high carbohydrate control diet or a high fat Western diet containing 0.03% (w/w) cholesterol. To identify the underlying mechanisms of dietary adaptations in intestine and liver, changes in gene expression were assessed by microarray and qPCR profiling, respectively. Results Mice on Western diet showed increased plasma cholesterol levels, associated with the higher dietary cholesterol supply, yet, significantly reduced cholesterol levels were found in intestine and liver. Transcript profiling revealed evidence that expression of numerous genes involved in cholesterol synthesis and uptake via LDL, but also in phospholipid metabolism, underwent compensatory regulations in both tissues. Alterations in glycerophospholipid metabolism were confirmed at the metabolite level by phospolipid profiling via mass spectrometry. Conclusions Our findings suggest that intestine and liver react to a high dietary fat intake by an activation of de novo cholesterol synthesis and other cholesterol-saving mechanisms, as well as with major changes in phospholipid metabolism, to accommodate to the fat load.</p

    Hepatic Methionine Homeostasis Is Conserved in C57BL/6N Mice on High-Fat Diet Despite Major Changes in Hepatic One-Carbon Metabolism

    Get PDF
    <div><p>Obesity is an underlying risk factor in the development of cardiovascular disease, dyslipidemia and non-alcoholic fatty liver disease (NAFLD). Increased hepatic lipid accumulation is a hallmark in the progression of NAFLD and impairments in liver phosphatidylcholine (PC) metabolism may be central to the pathogenesis. Hepatic PC biosynthesis, which is linked to the one-carbon (C1) metabolism by phosphatidylethanolamine N-methyltransferase, is known to be important for hepatic lipid export by VLDL particles. Here, we assessed the influence of a high-fat (HF) diet and NAFLD status in mice on hepatic methyl-group expenditure and C1-metabolism by analyzing changes in gene expression, protein levels, metabolite concentrations, and nuclear epigenetic processes. In livers from HF diet induced obese mice a significant downregulation of cystathionine β-synthase (CBS) and an increased betaine-homocysteine methyltransferase (BHMT) expression were observed. Experiments <i>in vitro</i>, using hepatoma cells stimulated with peroxisome proliferator activated receptor alpha (PPARα) agonist WY14,643, revealed a significantly reduced Cbs mRNA expression. Moreover, metabolite measurements identified decreased hepatic cystathionine and L-α-amino-n-butyrate concentrations as part of the transsulfuration pathway and reduced hepatic betaine concentrations, but no metabolite changes in the methionine cycle in HF diet fed mice compared to controls. Furthermore, we detected diminished hepatic gene expression of <i>de novo</i> DNA methyltransferase 3b but no effects on hepatic global genomic DNA methylation or hepatic DNA methylation in the Cbs promoter region upon HF diet. Our data suggest that HF diet induces a PPARα-mediated downregulation of key enzymes in the hepatic transsulfuration pathway and upregulates BHMT expression in mice to accommodate to enhanced dietary fat processing while preserving the essential amino acid methionine.</p> </div

    Influence of HF diet on hepatic Dnmt gene expression and global DNA methylation.

    No full text
    <p>(<b>A–C</b>) Quantification of Dnmt gene expression after 12 weeks of feeding (n = 5–6). Control mice showed stronger expression for Dnmt1 (Ct = 25.13±0.06) and <i>de novo</i> Dnmt3a (Ct = 28.10±0.15) than for <i>de novo</i> Dnmt3b (Ct = 30.55±0.31). Upon HF feeding, Dnmt1 mRNA expression was unaltered (Ct = 25.03±0.11), but gene expression of Dnmt3a (Ct = 28.28±0.18) and Dnmt3b (Ct = 31.37±0.14) decreased, respectively. (<b>D</b>) Analysis of hepatic global DNA methylation of control and HF animals (n = 6). DNA methylation was calculated from the (<i>Hpa</i>II/<i>Msp</i>I) ratio, whereby a ratio of 1 indicates 0% methylation and a ratio approaching 0 corresponds to 100% DNA methylation at the investigated sites. Data are presented as mean ± SEM. Open and grey bars represent control and HF animals, respectively. Asterisk indicates statistical significance (p<0.05).</p

    Schematic presentation of analyzed changes in hepatic C1-metabolism after HF feeding in C57BL/6N mice.

    No full text
    <p>Observed changes of mRNAs (Bhmt, Cbs, Csad, Got, Gss, PPARα), proteins (BHMT and CBS) and measured metabolites (taurine, homocysteine, methionine, betaine, DMG) are depicted. Dotted lines represent inhibitory effects of insulin (reported by <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0057387#pone.0057387-Ratnam1" target="_blank">[48]</a>, <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0057387#pone.0057387-Ratnam2" target="_blank">[49]</a>) and PPARα (this study) on the regulation of transcription. Cross means disrupted inhibitory effect of insulin reported in hyperglycemic mice (40–43).</p

    Impact of HF diet on selected hepatic metabolite concentrations after 12 weeks of feeding.

    No full text
    <p>Data are presented as box and whisker plot. (<b>A</b>) Selected data for taurine, L-glutamine, L-α-amino-n-butyrate, L-citrulline, L-ornithine, hydroxyproline and L-methionine (n = 9–11). (<b>B</b>) Analysis of S-adenosyl-methionine, S-adenosyl-homocysteine, L-homocysteine, cystathionine (n = 5–6) and choline, betaine and dimethylglycine (n = 7–9). (<b>C</b>) Selected ratios between measured hepatic metabolite concentrations. Open and grey bars represent control and HF mice, respectively. Asterisk indicates statistical significance (p<0.05).</p
    corecore