Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Gastroenterology ; 152(5): 1126-1138.e6, 2017 04.
Article in English | MEDLINE | ID: mdl-28065787

ABSTRACT

BACKGROUND & AIMS: The role of the intestine in the maintenance of cholesterol homeostasis increasingly is recognized. Fecal excretion of cholesterol is the last step in the atheroprotective reverse cholesterol transport pathway, to which biliary and transintestinal cholesterol excretion (TICE) contribute. The mechanisms controlling the flux of cholesterol through the TICE pathway, however, are poorly understood. We aimed to identify mechanisms that regulate and stimulate TICE. METHODS: We performed studies with C57Bl/6J mice, as well as with mice with intestine-specific knockout of the farnesoid X receptor (FXR), mice that express an FXR transgene specifically in the intestine, and ABCG8-knockout mice. Mice were fed a control diet or a diet supplemented with the FXR agonist PX20606, with or without the cholesterol absorption inhibitor ezetimibe. Some mice with intestine-specific knockout of FXR were given daily injections of fibroblast growth factor (FGF)19. To determine fractional cholesterol absorption, mice were given intravenous injections of cholesterol D5 and oral cholesterol D7. Mice were given 13C-acetate in drinking water for measurement of cholesterol synthesis. Bile cannulations were performed and biliary cholesterol secretion rates were assessed. In a separate set of experiments, bile ducts of male Wistar rats were exteriorized, allowing replacement of endogenous bile by a model bile. RESULTS: In mice, we found TICE to be regulated by intestinal FXR via induction of its target gene Fgf15 (FGF19 in rats and human beings). Stimulation of this pathway caused mice to excrete up to 60% of their total cholesterol content each day. PX20606 and FGF19 each increased the ratio of muricholate:cholate in bile, inducing a more hydrophilic bile salt pool. The altered bile salt pool stimulated robust secretion of cholesterol into the intestinal lumen via the sterol-exporting heterodimer adenosine triphosphate binding cassette subfamily G member 5/8 (ABCG5/G8). Of note, the increase in TICE induced by PX20606 was independent of changes in cholesterol absorption. CONCLUSIONS: Hydrophilicity of the bile salt pool, controlled by FXR and FGF15/19, is an important determinant of cholesterol removal via TICE. Strategies that alter bile salt pool composition might be developed for the prevention of cardiovascular disease. Transcript profiling: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=irsrayeohfcntqx&acc=GSE74101.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Fibroblast Growth Factors/metabolism , Intestinal Elimination/genetics , Intestinal Mucosa/metabolism , Lipoproteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Animals , Anticholesteremic Agents/pharmacology , Benzoates/pharmacology , Bile Ducts , Ezetimibe/pharmacology , Intestinal Elimination/drug effects , Intestinal Mucosa/drug effects , Intestines/drug effects , Isoxazoles/pharmacology , Lipoproteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/agonists
2.
Arterioscler Thromb Vasc Biol ; 37(4): 643-646, 2017 04.
Article in English | MEDLINE | ID: mdl-28232326

ABSTRACT

OBJECTIVE: Reverse cholesterol transport comprises efflux of cholesterol from macrophages and its subsequent removal from the body with the feces and thereby protects against formation of atherosclerotic plaques. Because of lack of suitable animal models that allow for evaluation of the respective contributions of biliary cholesterol secretion and transintestinal cholesterol excretion (TICE) to macrophage reverse cholesterol transport under physiological conditions, the relative importance of both pathways in this process has remained controversial. APPROACH AND RESULTS: To separate cholesterol traffic via the biliary route from TICE, bile flow was mutually diverted between rats, continuously, for 3 days. Groups of 2 weight-matched rats were designated as a pair, and both rats were equipped with cannulas in the bile duct and duodenum. Bile from rat 1 was diverted to the duodenum of rat 2, whereas bile from rat 2 was rerouted to the duodenum of rat 1. Next, rat 1 was injected with [3H]cholesterol-loaded macrophages. [3H]Cholesterol secreted via the biliary route was consequently diverted to rat 2 and could thus be quantified from the feces of that rat. On the other hand, [3H]cholesterol tracer in the feces of rat 1 reflected macrophage-derived cholesterol excreted via TICE. Using this setup, we found that 63% of the label secreted with the fecal neutral sterols had travelled via the biliary route, whereas 37% was excreted via TICE. CONCLUSIONS: TICE and biliary cholesterol secretion contribute to macrophage reverse cholesterol transport in rats. The majority of macrophage-derived cholesterol is however excreted via the hepatobiliary route.


Subject(s)
Bile/metabolism , Cholesterol/metabolism , Duodenum/metabolism , Intestinal Secretions/metabolism , Macrophages/metabolism , Animals , Biological Transport , Feces/chemistry , Hepatobiliary Elimination , Intestinal Elimination , Male , Models, Animal , Rats, Wistar , Time Factors
3.
Gastroenterology ; 150(3): 650-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26602218

ABSTRACT

BACKGROUND & AIMS: Reducing postprandial triglyceridemia may be a promising strategy to lower the risk of cardiovascular disorders associated with obesity and type 2 diabetes. In enterocytes, scavenger receptor class B, type 1 (SR-B1, encoded by SCARB1) mediates lipid-micelle sensing to promote assembly and secretion of chylomicrons. The nuclear receptor subfamily 1, group H, members 2 and 3 (also known as liver X receptors [LXRs]) regulate genes involved in cholesterol and fatty acid metabolism. We aimed to determine whether intestinal LXRs regulate triglyceride absorption. METHODS: C57BL/6J mice were either fed a cholesterol-enriched diet or given synthetic LXR agonists (GW3965 or T0901317). We measured the production of chylomicrons and localized SR-B1 by immunohistochemistry. Mechanisms of postprandial triglyceridemia and SR-B1 regulation were studied in Caco-2/TC7 cells incubated with LXR agonists. RESULTS: In mice and in the Caco-2/TC7 cell line, LXR agonists caused localization of intestinal SR-B1 from apical membranes to intracellular organelles and reduced chylomicron secretion. In Caco-2/TC7 cells, LXR agonists reduced SR-B1-dependent lipidic-micelle-induced Erk phosphorylation. LXR agonists also reduced intracellular trafficking of the apical apolipoprotein B pool toward secretory compartments. LXR reduced levels of SR-B1 in Caco-2/TC7 cells via a post-transcriptional mechanism that involves microRNAs. CONCLUSION: In Caco-2/TC7 cells and mice, intestinal activation of LXR reduces the production of chylomicrons by a mechanism dependent on the apical localization of SR-B1.


Subject(s)
Intestinal Absorption , Intestinal Mucosa/metabolism , Jejunum/metabolism , Orphan Nuclear Receptors/metabolism , Scavenger Receptors, Class B/metabolism , Triglycerides/metabolism , Animals , Apolipoprotein B-100/metabolism , Apolipoproteins B/metabolism , Benzoates/pharmacology , Benzylamines/pharmacology , Caco-2 Cells , Cholesterol, Dietary/metabolism , Chylomicrons/metabolism , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Down-Regulation , Humans , Hydrocarbons, Fluorinated/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Jejunum/drug effects , Liver X Receptors , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Orphan Nuclear Receptors/agonists , Protein Transport , RNA Interference , Ribonuclease III/deficiency , Ribonuclease III/genetics , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Signal Transduction , Sulfonamides/pharmacology , Transcription, Genetic , Transfection
4.
Curr Opin Lipidol ; 27(3): 295-301, 2016 06.
Article in English | MEDLINE | ID: mdl-27031274

ABSTRACT

PURPOSE OF REVIEW: The view on bile salts has evolved over the years from being regarded as simple detergents that aid intestinal absorption of fat-soluble nutrients to being important hormone-like integrators of metabolism. This review provides an update on the rapidly developing field of interactions between bile salts and lipid metabolism, with a particular emphasis on the underlying mechanisms. RECENT FINDINGS: The nuclear receptor farnesoid X receptor (FXR) plays major roles in bile salt-mediated signaling pathways. The recent identification of novel FXR targets and factors involved in FXR signaling highlights the interactions of bile acids with lipid metabolism. Exciting data have been reported on the use of intestine-specific FXR agonists as well as antagonists. In addition, encouraging results for treatment of hepatic steatosis obtained with obeticholic acid in the FLINT trial underline the therapeutic potential of bile salt signaling and metabolism for the treatment of lipid disorders. SUMMARY: Modulation of FXR activity appears to be a potent target, not only for improving bile salt homeostasis, but also to improve lipid metabolism. Depending on the metabolic context both, FXR agonists as well as antagonists, could prove to be of therapeutic benefit.


Subject(s)
Bile Acids and Salts/metabolism , Lipid Metabolism , Animals , Bariatric Surgery , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/microbiology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
5.
J Lipid Res ; 57(8): 1455-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27313057

ABSTRACT

Statins are competitive inhibitors of HMG-CoA reductase, the rate-limiting enzyme of cholesterol synthesis. Statins reduce plasma cholesterol levels, but whether this is actually caused by inhibition of de novo cholesterol synthesis has not been clearly established. Using three different statins, we investigated the effects on cholesterol metabolism in mice in detail. Surprisingly, direct measurement of whole body cholesterol synthesis revealed that cholesterol synthesis was robustly increased in statin-treated mice. Measurement of organ-specific cholesterol synthesis demonstrated that the liver is predominantly responsible for the increase in cholesterol synthesis. Excess synthesized cholesterol did not accumulate in the plasma, as plasma cholesterol decreased. However, statin treatment led to an increase in cholesterol removal via the feces. Interestingly, enhanced cholesterol excretion in response to rosuvastatin and lovastatin treatment was mainly mediated via biliary cholesterol secretion, whereas atorvastatin mainly stimulated cholesterol removal via the transintestinal cholesterol excretion pathway. Moreover, we show that plasma cholesterol precursor levels do not reflect cholesterol synthesis rates during statin treatment in mice. In conclusion, cholesterol synthesis is paradoxically increased upon statin treatment in mice. However, statins potently stimulate the excretion of cholesterol from the body, which sheds new light on possible mechanisms underlying the cholesterol-lowering effects of statins.


Subject(s)
Cholesterol/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/metabolism , Lovastatin/pharmacology , Animals , Cholesterol/blood , Drug Evaluation, Preclinical , Gene Expression/drug effects , Glutarates/metabolism , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Intestinal Elimination/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Liver/drug effects , Male , Mice, Inbred C57BL
6.
J Lipid Res ; 55(12): 2554-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25348863

ABSTRACT

Plant sterols and stanols are structurally similar to cholesterol and when added to the diet they are able to reduce serum total- and LDL-cholesterol concentrations. They also lower serum triglyceride concentrations in humans, particularly under conditions of hypertriglyceridemia. The aim of this study was to unravel the mechanism by which plant sterols and stanols reduce serum triglyceride concentrations in high-fat diet (HFD) fed mice. Male C57BL/6J mice were fed HFD for 4 weeks. Subsequently, they received HFD, HFD supplemented with 3.1% plant sterol ester (PSE) or HFD supplemented with 3.1% plant stanol ester (PSA) for another three weeks. Both PSE and PSA feeding resulted in decreased plasma triglyceride concentrations compared with HFD, while plasma cholesterol levels were unchanged. Interestingly, hepatic cholesterol levels were decreased in the PSE/PSA groups compared with HFD and no differences were found in hepatic triglyceride levels between groups. To investigate the mechanism underlying the hypotriglyceridemic effects from PSE/PSA feeding, we measured chylomicron and VLDL secretion. PSE and PSA feeding resulted in reduced VLDL secretion, while no differences were found between groups in chylomicron secretion. In conclusion, our data indicate that plasma triglyceride-lowering resulting from PSE and PSA feeding is associated with decreased hepatic VLDL secretion.


Subject(s)
Dietary Supplements , Esters/therapeutic use , Hypertriglyceridemia/diet therapy , Hypolipidemic Agents/therapeutic use , Lipoproteins, VLDL/metabolism , Liver/metabolism , Phytosterols/therapeutic use , Sitosterols/therapeutic use , Animals , Cholesterol/blood , Cholesterol/metabolism , Chylomicrons/blood , Chylomicrons/metabolism , Diet, High-Fat/adverse effects , Esters/metabolism , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Lipoproteins, VLDL/blood , Male , Mice, Inbred C57BL , Phytosterols/metabolism , Postprandial Period , Reproducibility of Results , Sitosterols/metabolism , Triglycerides/blood , Triglycerides/metabolism
7.
Acta Diabetol ; 54(2): 191-199, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27796655

ABSTRACT

AIMS: The dyslipidemia associated with type 2 diabetes is a major risk factor for the development of atherosclerosis. Trans-intestinal cholesterol excretion (TICE) has recently been shown to contribute, together with the classical hepatobiliary route, to fecal cholesterol excretion and cholesterol homeostasis. The aim of this study was to develop an in vitro cell model to investigate enterocyte-related processes of TICE. METHODS: Differentiated Caco-2/TC7 cells were grown on transwells and incubated basolaterally (blood side) with human plasma and apically (luminal side) with lipid micelles. Radioactive and fluorescent cholesterol tracers were used to investigate cholesterol uptake at the basolateral membrane, intracellular distribution and apical excretion. RESULTS: Our results show that cholesterol is taken up at the basolateral membrane, accumulates intracellularly as lipid droplets and undergoes a cholesterol acceptor-facilitated and progressive excretion through the apical membrane of enterocytes. The overall process is abolished at 4 °C, suggesting a biologically active phenomenon. Moreover, this trans-enterocytic retrograde cholesterol transport displays some TICE features like modulation by PCSK9 and an ABCB1 inhibitor. Finally, we highlight the involvement of microtubules in the transport of plasma cholesterol from basolateral to apical pole of enterocytes. CONCLUSIONS: The human Caco-2/TC7 cell line appears a good in vitro model to investigate the enterocytic molecular mechanisms of TICE, which may help to identify intestinal molecular targets to enhance reverse cholesterol transport and fight against dyslipidemia.


Subject(s)
Atherosclerosis/complications , Cholesterol/metabolism , Diabetes Mellitus, Type 2/complications , Dyslipidemias/metabolism , Enterocytes/metabolism , Exocytosis , Caco-2 Cells , Dyslipidemias/etiology , Humans
8.
Cell Metab ; 24(6): 783-794, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27818259

ABSTRACT

Except for conversion to bile salts, there is no major cholesterol degradation pathway in mammals. Efficient excretion from the body is therefore a crucial element in cholesterol homeostasis. Yet, the existence and importance of cholesterol degradation pathways in humans is a matter of debate. We quantified cholesterol fluxes in 15 male volunteers using a cholesterol balance approach. Ten participants repeated the protocol after 4 weeks of treatment with ezetimibe, an inhibitor of intestinal and biliary cholesterol absorption. Under basal conditions, about 65% of daily fecal neutral sterol excretion was bile derived, with the remainder being contributed by direct transintestinal cholesterol excretion (TICE). Surprisingly, ezetimibe induced a 4-fold increase in cholesterol elimination via TICE. Mouse studies revealed that most of ezetimibe-induced TICE flux is mediated by the cholesterol transporter Abcg5/Abcg8. In conclusion, TICE is active in humans and may serve as a novel target to stimulate cholesterol elimination in patients at risk for cardiovascular disease.


Subject(s)
Cholesterol/metabolism , Ezetimibe/pharmacology , Feces/chemistry , Intestinal Mucosa/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Adult , Animals , Bile/chemistry , Bile Acids and Salts/metabolism , Biological Transport/drug effects , Cholesterol/blood , Female , Humans , Intestines/drug effects , Kinetics , Lipoproteins/deficiency , Lipoproteins/metabolism , Male , Mice, Inbred C57BL , Middle Aged
9.
Lipids ; 50(6): 529-41, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25931382

ABSTRACT

The kinetics of plant stanol uptake and routing in 8-week-old C57BL/6J mice were determined after a plant stanol ester gavage. In addition, acute changes in intestinal and hepatic gene expression were investigated. Mice were fed a plant sterol/stanol poor diet from weaning. At the age of 8 weeks, they received an oral gavage consisting of 0.25 mg cholesterol + 50 mg plant stanol esters dissolved in olive oil. Animals were euthanized at different time points. In a second comparable set-up, mesenteric lymph-cannulated versus sham-operated mice received the same oral gavage, which was now deuterium labeled. Intestinal and hepatic sitostanol concentrations increased within 15 min post-gavage. This rapid hepatic appearance was absent in lymph-cannulated mice, suggesting a very fast lymph-mediated uptake. Hepatic mRNA expression of SREBP2 and its target genes rapidly decreased, whereas expression of LXR target genes increased. The intestinal SREBP2 pathway was increased, whereas the expression of LXR target genes hardly changed. The fivefold and sixfold increased expression of intestinal LDLr and PCSK9 is suggestive of TICE activation. We conclude that in C57BL/6J mice plant stanol kinetics are fast, and affect intestinal and hepatic gene expression within 15 min postprandial after lymph-mediated uptake.


Subject(s)
Gene Expression , Intestinal Mucosa/metabolism , Lipid Metabolism , Lipoproteins/metabolism , Liver/metabolism , Sitosterols/pharmacokinetics , Animals , Animals, Newborn , Cholesterol/blood , Cholesterol/genetics , Cholesterol/metabolism , Female , Liver X Receptors , Male , Mice, Inbred C57BL , Orphan Nuclear Receptors/metabolism , Proprotein Convertase 9 , Proprotein Convertases/metabolism , RNA, Messenger/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Sitosterols/blood , Sterol Regulatory Element Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL