Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-31470114

ABSTRACT

BACKGROUND & AIMS: The bile acid (BA)-activated farnesoid X receptor (FXR) controls hepatic BA synthesis and cell proliferation via the intestinal hormone fibroblast growth factor 19. Because cystic fibrosis (CF) is associated with intestinal dysbiosis, anomalous BA handling, and biliary cirrhosis, we investigated FXR signaling in CF. METHODS: Intestinal and hepatic expression of FXR target genes and inflammation markers was assessed in Cftr null mice and controls. Localization of the apical sodium-dependent BA transporter was assessed, and BAs in gastrointestinal tissues were analyzed. The CF microbiota was characterized and FXR signaling was investigated in intestinal tissue and organoids. RESULTS: Ileal murine fibroblast growth factor 19 ortholog (Fgf15) expression was strongly reduced in CF mice, compared with controls. Luminal BA levels and localization of apical sodium-dependent BA transporter was not affected, and BAs induced Fgf15 up to normal levels in CF ileum, ex vivo, and CF organoids. CF mice showed a dysbiosis that was associated with a marked up-regulation of genes involved in host-microbe interactions, including those involved in mucin glycosylation, antimicrobial defense, and Toll-like receptor signaling. Antibiotic treatment reversed the up-regulation of inflammatory markers and restored intestinal FXR signaling in CF mice. Conversely, FXR-dependent gene induction in ileal tissue and organoids was repressed by bacterial lipopolysaccharide and proinflammatory cytokines, respectively. Loss of intestinal FXR activity was associated with a markedly blunted hepatic trophic response to oral BA supplementation, and with impaired repression of Cyp7a1, the gene encoding the rate-limiting enzyme in BA synthesis. CONCLUSIONS: In CF mice, the gut microbiota represses intestinal FXR activity, and, consequently, FXR-dependent hepatic cell proliferation and feedback control of BA synthesis.


Subject(s)
Cystic Fibrosis/immunology , Dysbiosis/immunology , Fibroblast Growth Factors/metabolism , Ileum/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/immunology , Cell Proliferation , Cholesterol 7-alpha-Hydroxylase/metabolism , Cystic Fibrosis/complications , Cystic Fibrosis/pathology , Disease Models, Animal , Down-Regulation , Dysbiosis/microbiology , Dysbiosis/pathology , Feedback, Physiological , Female , Gastrointestinal Microbiome/immunology , Host Microbial Interactions/immunology , Humans , Ileum/immunology , Ileum/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Liver/cytology , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred CFTR , Up-Regulation
2.
J Cyst Fibros ; 18(2): 286-293, 2019 03.
Article in English | MEDLINE | ID: mdl-30279125

ABSTRACT

OBJECTIVE: Disruption of the enterohepatic circulation of bile acids (BAs) is part of the gastrointestinal phenotype of cystic fibrosis (CF). Ivacaftor (VX-770), a cystic fibrosis transmembrane conductance regulator (CFTR) potentiator, improves pulmonary function in CF patients with class III gating mutations. We studied the effect of ivacaftor on the enterohepatic circulation by assessing markers of BA homeostasis and their changes in CF patients. METHODS: In CF patients with an S1251N mutation (N = 16; age 9-35 years S125N study/NTR4873) or a G551D mutation (N = 101; age 10-24 years; GOAL study/ NCT01521338) we analyzed plasma fibroblast growth factor 19 (FGF19) and 7α-hydroxy-4-cholesten-3-one (C4) levels, surrogate markers for intestinal BA absorption and hepatic synthesis, respectively, before and after treatment with ivacaftor. RESULTS: At baseline, median FGF19 was lower (52% and 53%, P < .001) and median C4 higher (350% and 364%, P < .001), respectively, for the S1251 N and G551D mutation patient groups compared to healthy controls. Treatment with ivacaftor significantly increased FGF19 and reduced C4 levels towards normalization in both cohorts but this did not correlate with CFTR function in other organs, as measured by sweat chloride levels or pulmonary function. CONCLUSIONS: We demonstrate that patients with CFTR gating mutations display interruption of the enterohepatic circulation of BAs reflected by lower FGF19 and elevated C4 levels. Treatment with ivacaftor partially restored this disruption of BA homeostasis. The improvement did not correlate with established outcome measures of CF, suggesting involvement of modulating factors of CFTR correction in different organs.


Subject(s)
Bile Acids and Salts , Cholestenones/blood , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Enterohepatic Circulation/drug effects , Fibroblast Growth Factors/blood , Adolescent , Adult , Aminophenols/pharmacokinetics , Aminophenols/therapeutic use , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/metabolism , Biological Availability , Child , Chloride Channel Agonists/pharmacokinetics , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Female , Homeostasis/drug effects , Humans , Male , Mutation , Netherlands , Quinolones/pharmacokinetics , Quinolones/therapeutic use
3.
Sci Rep ; 7(1): 846, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28404991

ABSTRACT

Peroxisome proliferator-activated receptor δ (PPARδ) is a ligand-activated transcription factor that has an important role in lipid metabolism. Activation of PPARδ stimulates fatty acid oxidation in adipose tissue and skeletal muscle and improves dyslipidemia in mice and humans. PPARδ is highly expressed in the intestinal tract but its physiological function in this organ is not known. Using mice with an intestinal epithelial cell-specific deletion of PPARδ, we show that intestinal PPARδ protects against diet-induced obesity, insulin resistance and dyslipidemia. Furthermore, absence of intestinal PPARδ abolished the ability of PPARδ agonist GW501516 to increase plasma levels of HDL-cholesterol. Together, our findings show that intestinal PPARδ is important in maintaining metabolic homeostasis and suggest that intestinal-specific activation of PPARδ could be a therapeutic approach for treatment of the metabolic syndrome and dyslipidemia, while avoiding systemic toxicity.


Subject(s)
Dyslipidemias/metabolism , Insulin Resistance , Intestinal Mucosa/metabolism , Obesity/metabolism , PPAR delta/genetics , Animals , Cholesterol, HDL/blood , Dyslipidemias/genetics , Mice , Mice, Inbred C57BL , Obesity/genetics , PPAR delta/agonists , PPAR delta/metabolism
4.
Biochim Biophys Acta ; 1861(9 Pt A): 1132-1141, 2016 09.
Article in English | MEDLINE | ID: mdl-27344248

ABSTRACT

Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is a key enzyme in triacylglycerol (TG) biosynthesis. Here we show that genetic deficiency and pharmacological inhibition of DGAT1 in mice alters cholesterol metabolism. Cholesterol absorption, as assessed by acute cholesterol uptake, was significantly decreased in the small intestine and liver upon DGAT1 deficiency/inhibition. Ablation of DGAT1 in the intestine (I-DGAT1(-/-)) alone is sufficient to cause these effects. Consequences of I-DGAT1 deficiency phenocopy findings in whole-body DGAT1(-/-) and DGAT1 inhibitor-treated mice. We show that deficiency/inhibition of DGAT1 affects cholesterol metabolism via reduced chylomicron size and increased trans-intestinal cholesterol excretion. These effects are independent of cholesterol uptake at the apical surface of enterocytes but mediated through altered dietary fatty acid metabolism. Our findings provide insight into a novel role of DGAT1 and identify a pathway by which intestinal DGAT1 deficiency affects whole-body cholesterol homeostasis in mice. Targeting intestinal DGAT1 may represent a novel approach for treating hypercholesterolemia.


Subject(s)
Cholesterol/metabolism , Diacylglycerol O-Acyltransferase/genetics , Hypercholesterolemia/drug therapy , Lipid Metabolism/genetics , Triglycerides/metabolism , Animals , Diacylglycerol O-Acyltransferase/deficiency , Diacylglycerol O-Acyltransferase/metabolism , Dietary Fats , Fatty Acids/metabolism , Hypercholesterolemia/metabolism , Intestinal Absorption/genetics , Lipogenesis/genetics , Liver/metabolism , Mice
5.
J Hepatol ; 63(3): 697-704, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26022694

ABSTRACT

BACKGROUND & AIMS: Regulation of bile acid homeostasis in mammals is a complex process regulated via extensive cross-talk between liver, intestine and intestinal microbiota. Here we studied the effects of gut microbiota on bile acid homeostasis in mice. METHODS: Bile acid homeostasis was assessed in four mouse models. Germfree mice, conventionally-raised mice, Asbt-KO mice and intestinal-specific Gata4-iKO mice were treated with antibiotics (bacitracin, neomycin and vancomycin; 100 mg/kg) for five days and subsequently compared with untreated mice. RESULTS: Attenuation of the bacterial flora by antibiotics strongly reduced fecal excretion and synthesis of bile acids, but increased the expression of the bile acid synthesis enzyme CYP7A1. Similar effects were seen in germfree mice. Intestinal bile acid absorption was increased and accompanied by increases in plasma bile acid levels, biliary bile acid secretion and enterohepatic cycling of bile acids. In the absence of microbiota, the expression of the intestinal bile salt transporter Asbt was strongly increased in the ileum and was also expressed in more proximal parts of the small intestine. Most of the effects of antibiotic treatment on bile acid homeostasis could be prevented by genetic inactivation of either Asbt or the transcription factor Gata4. CONCLUSIONS: Attenuation of gut microbiota alters Gata4-controlled expression of Asbt, increasing absorption and decreasing synthesis of bile acids. Our data support the concept that under physiological conditions microbiota stimulate Gata4, which suppresses Asbt expression, limiting the expression of this transporter to the terminal ileum. Our studies expand current knowledge on the bacterial control of bile acid homeostasis.


Subject(s)
Bile Acids and Salts/metabolism , GATA4 Transcription Factor/physiology , Gastrointestinal Microbiome/physiology , Intestinal Absorption , Organic Anion Transporters, Sodium-Dependent/physiology , Symporters/physiology , Animals , Anti-Bacterial Agents/pharmacology , Cholesterol 7-alpha-Hydroxylase/genetics , Homeostasis , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
6.
J Cyst Fibros ; 14(4): 440-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25633479

ABSTRACT

BACKGROUND: Cftr(-/-tm1Unc) mice develop progressive hepato-biliary pathology. We hypothesize that this liver pathology is related to alterations in biliary bile hydrophobicity and bile salt metabolism in Cftr(-/-tm1Unc) mice. METHODS: We determined bile production, biliary and fecal bile salt- and lipid compositions and fecal bacterial composition of C57BL/6J Cftr(-/-tm1Unc) and control mice. RESULTS: We found no differences between the total biliary bile salt or lipid concentrations of Cftr(-/-) and controls. Compared to controls, Cftr(-/-) mice had a ~30% higher bile production and a low bile hydrophobicity, related to a ~7 fold higher concentration of the choleretic and hydrophilic bile salt ursocholate. These findings coexisted with a significantly smaller quantity of fecal Bacteroides bacteria. CONCLUSIONS: Liver pathology in Cftr(-/-tm1Unc) is not related to increased bile hydrophobicity. Cftr(-/-) mice do however display a biliary phenotype characterized by increased bile production and decreased biliary hydrophobicity. Our findings suggest Cftr dependent, alterations in intestinal bacterial biotransformation of bile salts.


Subject(s)
Bile Acids and Salts/metabolism , Bile Ducts/pathology , Bile/physiology , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Liver/pathology , Animals , Cystic Fibrosis Transmembrane Conductance Regulator , Disease Models, Animal , Feces/chemistry , Feces/microbiology , Female , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Congenic , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...