Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cells ; 10(12)2021 12 09.
Article in English | MEDLINE | ID: mdl-34943976

ABSTRACT

Niemann-Pick C1-like 1 (NPC1L1) mediates intestinal uptake of dietary and biliary cholesterol and is the target of ezetimibe, a cholesterol absorption inhibitor used to treat hypercholesterolemia. Genetic deletion of NPC1L1 or ezetimibe treatment protects mice from high-fat diet (HFD)-induced obesity; however, the molecular mechanisms responsible for this therapeutic benefit remain unknown. A major metabolic fate of cholesterol is its conversion to bile acids. We found that NPC1L1 knockout (L1-KO) mice fed an HFD had increased energy expenditure, bile acid pool size, and fecal bile acid excretion rates. The elevated bile acid pool in the HFD-fed L1-KO mice was enriched with tauro-ß-muricholic acid. These changes in the L1-KO mice were associated with reduced ileal mRNA expression of fibroblast growth factor 15 (FGF15) and increased hepatic mRNA expression of cholesterol 7α-hydroxylase (Cyp7A1) and mitochondrial sterol 27-hydroxylase (Cyp27A1). In addition, mRNA expression of the membrane bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) and type 2 iodothyronine deiodinase (Dio2) were elevated in brown adipose tissue of L1-KO mice, which is known to promote energy expenditure. Thus, altered bile acid homeostasis and signaling may play a role in protecting L1-KO mice against HFD-induced obesity.


Subject(s)
Bile Acids and Salts/genetics , Fibroblast Growth Factors/genetics , Ileum/metabolism , Membrane Transport Proteins/genetics , Obesity/genetics , Animals , Bile Acids and Salts/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/genetics , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Humans , Ileum/drug effects , Ileum/pathology , Iodide Peroxidase/genetics , Liver/metabolism , Liver/pathology , Membrane Transport Proteins/deficiency , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Receptors, G-Protein-Coupled/genetics , Iodothyronine Deiodinase Type II
2.
Cell Mol Gastroenterol Hepatol ; 5(4): 499-522, 2018.
Article in English | MEDLINE | ID: mdl-29930976

ABSTRACT

BACKGROUND & AIMS: Ileal bile acid absorption is mediated by uptake via the apical sodium-dependent bile acid transporter (ASBT), and export via the basolateral heteromeric organic solute transporter α-ß (OSTα-OSTß). In this study, we investigated the cytotoxic effects of enterocyte bile acid stasis in Ostα-/- mice, including the temporal relationship between intestinal injury and initiation of the enterohepatic circulation of bile acids. METHODS: Ileal tissue morphometry, histology, markers of cell proliferation, gene, and protein expression were analyzed in male and female wild-type and Ostα-/- mice at postnatal days 5, 10, 15, 20, and 30. Ostα-/-Asbt-/- mice were generated and analyzed. Bile acid activation of intestinal Nrf2-activated pathways was investigated in Drosophila. RESULTS: As early as day 5, Ostα-/- mice showed significantly increased ileal weight per length, decreased villus height, and increased epithelial cell proliferation. This correlated with premature expression of the Asbt and induction of bile acid-activated farnesoid X receptor target genes in neonatal Ostα-/- mice. Expression of reduced nicotinamide adenine dinucleotide phosphate oxidase-1 and Nrf2-anti-oxidant responsive genes were increased significantly in neonatal Ostα-/- mice at these postnatal time points. Bile acids also activated Nrf2 in Drosophila enterocytes and enterocyte-specific knockdown of Nrf2 increased sensitivity of flies to bile acid-induced toxicity. Inactivation of the Asbt prevented the changes in ileal morphology and induction of anti-oxidant response genes in Ostα-/- mice. CONCLUSIONS: Early in postnatal development, loss of Ostα leads to bile acid accumulation, oxidative stress, and a restitution response in ileum. In addition to its essential role in maintaining bile acid homeostasis, Ostα-Ostß functions to protect the ileal epithelium against bile acid-induced injury. NCBI Gene Expression Omnibus: GSE99579.

3.
Atherosclerosis ; 229(2): 374-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23880190

ABSTRACT

OBJECTIVE: Interruption of the enterohepatic circulation of bile acids induces hepatic bile acid synthesis, increases hepatic cholesterol demand, and increases clearance of apoB-containing lipoproteins in plasma. Based on these effects, bile acid sequestrants have been used for many years to treat hypercholesterolemia and the associated atherosclerosis. The objective of this study was to determine the effect of blocking ileal apical versus basolateral membrane bile acid transport on the development of hypercholesterolemia and atherosclerosis in mouse models. METHODS AND RESULTS: ApoE(-/-) and Ldlr(-/-) mice deficient in the apical sodium-dependent bile acid transporter (Asbt) or apoE(-/-) mice deficient in the basolateral bile acid transporter (Ostα) were fed an atherogenic diet for 16 weeks. Bile acid metabolism, cholesterol metabolism, gene expression, and development of atherosclerosis were examined. Mice deficient in Asbt exhibited the classic response to interruption of the enterohepatic circulation of bile acids, including significant reductions in hepatic and plasma cholesterol levels, and reduced aortic cholesteryl ester content. Ileal Fibroblast Growth Factor-15 (FGF15) expression was significantly reduced in Asbt(-/-)apoE(-/-) mice and was inversely correlated with expression of hepatic cholesterol 7-hydroxylase (Cyp7a1). Ileal FGF15 expression was directly correlated with plasma cholesterol levels and aortic cholesterol content. In contrast, plasma and hepatic cholesterol levels and atherosclerosis development were not reduced in apoE(-/-) mice deficient in Ostα. CONCLUSIONS: Decreases in ileal FGF15, with subsequent increases in hepatic Cyp7a1 expression and bile acid synthesis appear to be necessary for the plasma cholesterol-lowering and atheroprotective effects associated with blocking intestinal bile acid absorption.


Subject(s)
Atherosclerosis/metabolism , Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Fibroblast Growth Factors/metabolism , Membrane Transport Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cell Polarity/physiology , Cholesterol/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Disease Models, Animal , Female , Fibroblast Growth Factors/genetics , Ileum/metabolism , Liver/metabolism , Male , Membrane Transport Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Symporters/genetics , Triglycerides/blood
4.
J Hepatol ; 57(2): 359-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22542490

ABSTRACT

BACKGROUND & AIMS: Blocking intestinal bile acid (BA) absorption by inhibiting or inactivating the apical sodium-dependent BA transporter (Asbt) classically induces hepatic BA synthesis. In contrast, blocking intestinal BA absorption by inactivating the basolateral BA transporter, organic solute transporter alpha-beta (Ostα-Ostß) is associated with an altered homeostatic response and decreased hepatic BA synthesis. The aim of this study was to determine the mechanisms underlying this phenotype, including the role of the farnesoid X receptor (FXR) and fibroblast growth factor 15 (FGF15). METHODS: BA and cholesterol metabolism, intestinal phenotype, expression of genes important for BA metabolism, and intestinal FGF15 expression were examined in wild type, Ostα(-/-), Fxr(-/-), and Ostα(-/-)Fxr(-/-) mice. RESULTS: Inactivation of Ostα was associated with decreases in hepatic cholesterol 7α-hydroxylase (Cyp7a1) expression, BA pool size, and intestinal cholesterol absorption. Ostα(-/-) mice exhibited significant small intestinal changes, including altered ileal villus morphology, and increases in intestinal length and mass. Total ileal FGF15 expression was elevated almost 20-fold in Ostα(-/-) mice as a result of increased villus epithelial cell number and ileocyte FGF15 protein expression. Ostα(-/-)Fxr(-/-) mice exhibited decreased ileal FGF15 expression, restoration of intestinal cholesterol absorption, and increases in hepatic Cyp7a1 expression, fecal BA excretion, and BA pool size. FXR deficiency did not reverse the intestinal morphological changes or compensatory decrease for ileal Asbt expression in Ostα(-/-) mice. CONCLUSIONS: These results indicate that signaling via FXR is required for the paradoxical repression of hepatic BA synthesis but not the complex intestinal adaptive changes in Ostα(-/-) mice.


Subject(s)
Bile Acids and Salts/metabolism , Fibroblast Growth Factors/genetics , Membrane Transport Proteins/physiology , Animals , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/physiology
5.
Dig Dis ; 29(1): 18-22, 2011.
Article in English | MEDLINE | ID: mdl-21691100

ABSTRACT

BACKGROUND/AIMS: Mutations in the apical sodium-dependent bile acid transporter (SLC10A2) block intestinal bile acid absorption, resulting in a compensatory increase in hepatic bile acid synthesis. Inactivation of the basolateral membrane bile acid transporter (OSTα-OSTß) also impairs intestinal bile acid absorption, but hepatic bile acid synthesis was paradoxically repressed. We hypothesized that the altered bile acid homeostasis resulted from ileal trapping of bile acids that act via the farnesoid X receptor (FXR) to induce overexpression of FGF15. To test this hypothesis, we investigated whether blocking FXR signaling would reverse the bile acid synthesis phenotype in Ostα null mice. METHODS: The corresponding null mice were crossbred to generate OstαFxr double-null mice. All experiments compared wild-type, Ostα, Fxr and OstαFxr null littermates. Analysis of the in vivo phenotype included measurements of bile acid fecal excretion, pool size and composition. Hepatic and intestinal gene and protein expression were also examined. RESULTS: OstαFxr null mice exhibited increased bile acid fecal excretion and pool size, and decreased bile acid pool hydrophobicity, as compared with Ostα null mice. Inactivation of FXR reversed the increase in ileal total FGF15 expression, which was associated with a significant increase in hepatic Cyp7a1 expression. CONCLUSIONS: Inactivation of FXR largely unmasked the bile acid malabsorption phenotype and corrected the bile acid homeostasis defect in Ostα null mice, suggesting that inappropriate activation of the FXR-FGF15-FGFR4 pathway partially underlies this phenotype. Intestinal morphological changes and reduced apical sodium-dependent bile acid transporter expression were maintained in Ostα(-/-)Fxr(-/-) mice, indicating that FXR is not required for these adaptive responses.


Subject(s)
Bile Acids and Salts/metabolism , Homeostasis , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/metabolism , Animals , Feces/chemistry , Gene Expression Regulation , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Mice, Knockout , Models, Biological , Phenotype , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism
6.
Biochim Biophys Acta ; 1811(9): 549-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21683156

ABSTRACT

Niemann-Pick C1-Like 1 (NPC1L1) is highly expressed in the small intestine across mammalian species and is the target of ezetimibe, a potent cholesterol absorption inhibitor. In humans, NPC1L1 is also expressed in the liver. We found that transgenic overexpression of NPC1L1 in the wild-type mouse liver inhibits biliary cholesterol secretion and raises blood cholesterol, which can be reversed by ezetimibe treatment. Unfortunately, the high expression of endogenous NPC1L1 in the intestine hampered a definitive establishment of the role of hepatic NPC1L1 in cholesterol metabolism and ezetimibe action in the liver because intestinal NPC1L1 dramatically influences cholesterol homeostasis and is a target of ezetimibe. To circumvent this obstacle, we crossed liver-specific NPC1L1 transgenic mice to NPC1L1 knockout (L1-KO) mice and created a mouse line expressing no endogenous NPC1L1, but human NPC1L1 in liver only (L1(LivOnly) mice). Compared to L1-KO mice, L1(LivOnly) mice on a 0.2% cholesterol diet showed significantly increased hepatic and plasma cholesterol, and despite a 90% reduction in biliary cholesterol excretion, their fecal cholesterol excretion remained completely unaltered. Remarkably, 4days of ezetimibe treatment significantly restored biliary cholesterol secretion in L1(LivOnly) mice. These findings demonstrated a direct role of hepatic NPC1L1 in regulating biliary cholesterol excretion and hepatic/blood cholesterol levels, and unequivocally established hepatic NPC1L1 as a target of ezetimibe.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Bile/chemistry , Cholesterol/metabolism , Liver/drug effects , Liver/metabolism , Membrane Transport Proteins/metabolism , Animals , Dietary Fats , Ezetimibe , Feces/chemistry , Humans , Lipid Metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
7.
Gut ; 59(7): 888-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20581237

ABSTRACT

BACKGROUND AND AIMS: The transcription factor GATA4 is expressed throughout most of the small intestine except distal ileum, and restricts expression of the apical sodium-dependent bile acid transporter (ASBT), the rate-limiting intestinal bile acid transporter, to distal ileum. The hypothesis was tested that reduction of GATA4 activity in mouse small intestine results in an induction of bile acid transport in proximal small intestine sufficient to restore bile acid absorption and homeostasis after ileocaecal resection (ICR). METHODS: Bile acid homeostasis was characterised in non-surgical, sham or ICR mice using two recombinant Gata4 models in which Asbt expression is induced to different levels. RESULTS: Reduction of intestinal GATA4 activity resulted in an induction of ASBT expression, bile acid absorption and expression of bile acid-responsive genes in proximal small intestine, and a reduction of luminal bile acids in distal small intestine. While faecal bile acid excretion and bile acid pool size remained unchanged, the bile acid pool became more hydrophilic due to a relative increase in tauro-beta-muricholate absorption. Furthermore, proximal induction of Asbt in both Gata4 mutant models corrected ICR-associated bile acid malabsorption, reversing the decrease in bile acid pool size and increase in faecal bile acid excretion and hepatic cholesterol 7alpha-hydroxylase expression. CONCLUSIONS: Reduction of intestinal GATA4 activity induces bile acid absorption in proximal small intestine without inducing major changes in bile acid homeostasis. This induction is sufficient to correct bile acid malabsorption caused by ICR in mice.


Subject(s)
Bile Acids and Salts/metabolism , GATA4 Transcription Factor/physiology , Intestinal Absorption/physiology , Intestine, Small/metabolism , Animals , Disease Models, Animal , GATA4 Transcription Factor/genetics , Gene Deletion , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Homeostasis/physiology , Intestinal Absorption/genetics , Intestine, Small/surgery , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Taurocholic Acid/analogs & derivatives , Taurocholic Acid/metabolism
8.
Drug Metab Dispos ; 37(12): 2375-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741038

ABSTRACT

The macrolide antibiotics azithromycin and clarithromycin are large molecular weight compounds that exhibit moderate to excellent oral bioavailability in preclinical species and humans. Previous concomitant dosing studies in rats using rifamycin SV, a general organic anion-transporting polypeptide (OATP) inhibitor, suggested that the high oral absorption of azithromycin and clarithromycin may be caused by facilitative uptake by intestinal Oatps. In this study, we used OATP/Oatp-expressing cells to investigate the interaction of macrolides with rat Oatp1a5, human OATP1A2, and human/rat OATP2B1/Oatp2b1. These experiments showed that azithromycin and clarithromycin were potent inhibitors of rat Oatp1a5-mediated taurocholate uptake with apparent inhibitor constant (K(i)) values of 3.3 and 2.4 microM, respectively. The macrolides functioned as noncompetitive inhibitors but were not transport substrates for rat Oatp1a5, as assessed by direct uptake measurements of radiolabeled azithromycin and clarithromycin. cis-Inhibition and direct uptake studies further showed that azithromycin and clarithromycin were only very weak inhibitors and not substrates for human OATP1A2 and human/rat OATP2B1/Oatp2b1. In summary, these results indicate that the macrolides azithromycin and clarithromycin potently inhibit rat Oatp1a5 but do not significantly interact with OATP1A2 and OATP2B1/Oatp2b1. These intestinally expressed OATP/Oatp(s) are not responsible for the postulated facilitative uptake of azithromycin and clarithromycin, and alternative facilitative pathways must exist for their intestinal absorption.


Subject(s)
Anti-Bacterial Agents/pharmacology , Intestines/drug effects , Macrolides/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Animals , Anti-Bacterial Agents/metabolism , Azithromycin/pharmacology , Biological Transport , COS Cells , Chlorocebus aethiops , Clarithromycin/pharmacology , Dogs , Dose-Response Relationship, Drug , Erythromycin/pharmacology , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Intestinal Mucosa/metabolism , Kinetics , Macrolides/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Rats , Taurocholic Acid/metabolism , Transfection
9.
Proc Natl Acad Sci U S A ; 105(10): 3891-6, 2008 Mar 11.
Article in English | MEDLINE | ID: mdl-18292224

ABSTRACT

The apical sodium-dependent bile acid transporter (Asbt) is responsible for transport across the intestinal brush border membrane; however, the carrier(s) responsible for basolateral bile acid export into the portal circulation remains to be determined. Although the heteromeric organic solute transporter Ostalpha-Ostbeta exhibits many properties predicted for a candidate intestinal basolateral bile acid transporter, the in vivo functions of Ostalpha-Ostbeta have not been investigated. To determine the role of Ostalpha-Ostbeta in intestinal bile acid absorption, the Ostalpha gene was disrupted by homologous recombination in mice. Ostalpha(-/-) mice were physically indistinguishable from wild-type mice. In everted gut sac experiments, transileal transport of taurocholate was reduced by >80% in Ostalpha(-/-) vs. wild-type mice; the residual taurocholate transport was further reduced to near-background levels in gut sacs prepared from Ostalpha(-/-)Mrp3(-/-) mice. The bile acid pool size was significantly reduced (>65%) in Ostalpha(-/-) mice, but fecal bile acid excretion was not elevated. The decreased pool size in Ostalpha(-/-) mice resulted from reduced hepatic Cyp7a1 expression that was inversely correlated with ileal expression of fibroblast growth factor 15 (FGF15). These data indicate that Ostalpha-Ostbeta is essential for intestinal bile acid transport in mice. Unlike a block in intestinal apical bile acid uptake, genetic ablation of basolateral bile acid export disrupts the classical homeostatic control of hepatic bile acid biosynthesis.


Subject(s)
Bile Acids and Salts/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , Animals , Biological Transport/drug effects , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholic Acid/administration & dosage , Cholic Acid/pharmacology , Feces/chemistry , Gene Expression Regulation/drug effects , Gene Targeting , Homeostasis/drug effects , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestines/drug effects , Lipids/isolation & purification , Liver/drug effects , Liver/metabolism , Male , Membrane Transport Proteins/deficiency , Mice , Mice, Knockout , Models, Biological , Phenotype , Serous Membrane/drug effects , Serous Membrane/metabolism
10.
Am J Physiol Gastrointest Liver Physiol ; 290(5): G912-22, 2006 May.
Article in English | MEDLINE | ID: mdl-16357058

ABSTRACT

The mechanisms responsible for bile acid regulation of mouse intestinal organic solute transporter alpha-beta (Ostalpha-Ostbeta) expression were investigated. Expression of Ostalpha-Ostbeta mRNA was increased in cecum and proximal colon of cholic acid-fed mice and in chenodeoxycholate-treated mouse CT26 colon adenocarcinoma cells. Sequence analysis revealed potential cis-acting elements for farnesoid X receptor (FXR) and liver receptor homolog-1 (LRH-1) in the mouse Ostalpha and Ostbeta promoters and reporter constructs containing Ostalpha and Ostbeta 5'-flanking sequences were positively regulated by bile acids. Expression of a dominant-negative FXR, reduction of FXR with interfering small RNA (siRNA), or mutation of the potential FXR elements decreased Ostalpha and Ostbeta promoter activity and abolished the induction by chenodeoxycolic acid. Negative regulation of the Ostalpha and Ostbeta promoters by bile acids was mediated through LRH-1 elements. Ostalpha and Ostbeta promoter activities were increased by coexpression of LRH-1 and decreased by coexpression of SHP. Mutation of the potential LRH-1 elements and siRNA-mediated reduction of LRH-1 expression decreased basal promoter activity. As predicted from the promoter analyses, ileal Ostalpha and Ostbeta mRNA expressions were increased in wild-type mice administered the FXR agonist GW4064 and decreased in FXR-null mice. Immunoblotting analysis revealed that Ostalpha and Ostbeta intestinal protein expressions correlated with mRNA expression. The mouse Ostalpha and Ostbeta promoters are unusual in that they contain functional FXR and LRH elements, which mediate, respectively, positive and negative feedback regulation by bile acids. Although the positive regulatory pathway appears to be dominant, this arrangement provides a mechanism to finely titrate Ostalpha-Ostbeta expression to the bile acid flux.


Subject(s)
Adenocarcinoma/metabolism , Bile Acids and Salts/pharmacology , Colonic Neoplasms/metabolism , Gene Expression Regulation , Membrane Transport Proteins/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
11.
J Biol Chem ; 280(8): 6960-8, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15563450

ABSTRACT

Bile acids are transported across the ileal enterocyte brush border membrane by the well characterized apical sodium-dependent bile acid transporter (Asbt) Slc10a2; however, the carrier(s) responsible for transporting bile acids across the ileocyte basolateral membrane into the portal circulation have not been fully identified. Transcriptional profiling of wild type and Slc10a2 null mice was employed to identify a new candidate basolateral bile acid carrier, the heteromeric organic solute transporter (Ost)alpha-Ostbeta. By Northern blot analysis, Ostalpha and Ostbeta mRNA was detected only in mouse kidney and intestine, mirroring the horizontal gradient of expression of Asbt in the gastrointestinal tract. Analysis of Ostalpha and Ostbeta protein expression by immunohistochemistry localized both subunits to the basolateral surface of the mouse ileal enterocyte. The transport properties of Ostalpha-Ostbeta were analyzed in stably transfected Madin-Darby canine kidney cells. Co-expression of mouse Ostalpha-Ostbeta, but not the individual subunits, stimulated Na(+)-independent bile acid uptake and the apical-to-basolateral transport of taurocholate. In contrast, basolateral-to-apical transport was not affected by Ostalpha-Ostbeta expression. Co-expression of Ostalpha and Ostbeta was required to convert the Ostalpha subunit to a mature glycosylated endoglycosidase H-resistant form, suggesting that co-expression facilitates the trafficking of Ostalpha through the Golgi apparatus. Immunolocalization studies showed that co-expression was necessary for plasma membrane expression of both Ostalpha and Ostbeta. These results demonstrate that the mouse Ostalpha-Ostbeta heteromeric transporter is a basolateral bile acid carrier and may be responsible for bile acid efflux in ileum and other ASBT-expressing tissues.


Subject(s)
Carrier Proteins , Ileum/chemistry , Membrane Glycoproteins , Membrane Transport Proteins/physiology , Animals , Bile Acids and Salts/metabolism , Cell Line , Gene Expression Profiling , Humans , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent/genetics , Protein Transport , RNA, Messenger/analysis , Symporters/genetics , Taurocholic Acid/metabolism , Tissue Distribution
12.
J Biol Chem ; 278(36): 33920-7, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12819193

ABSTRACT

The ileal apical sodium bile acid cotransporter participates in the enterohepatic circulation of bile acids. In patients with primary bile acid malabsorption, mutations in the ileal bile acid transporter gene (Slc10a2) lead to congenital diarrhea, steatorrhea, and reduced plasma cholesterol levels. To elucidate the quantitative role of Slc10a2 in intestinal bile acid absorption, the Slc10a2 gene was disrupted by homologous recombination in mice. Animals heterozygous (Slc10a2+/-) and homozygous (Slc10a2-/-) for this mutation were physically indistinguishable from wild type mice. In the Slc10a2-/- mice, fecal bile acid excretion was elevated 10- to 20-fold and was not further increased by feeding a bile acid binding resin. Despite increased bile acid synthesis, the bile acid pool size was decreased by 80% and selectively enriched in cholic acid in the Slc10a2-/- mice. On a low fat diet, the Slc10a2-/- mice did not have steatorrhea. Fecal neutral sterol excretion was increased only 3-fold, and intestinal cholesterol absorption was reduced only 20%, indicating that the smaller cholic acid-enriched bile acid pool was sufficient to facilitate intestinal lipid absorption. Liver cholesteryl ester content was reduced by 50% in Slc10a2-/- mice, and unexpectedly plasma high density lipoprotein cholesterol levels were slightly elevated. These data indicate that Slc10a2 is essential for efficient intestinal absorption of bile acids and that alternative absorptive mechanisms are unable to compensate for loss of Slc10a2 function.


Subject(s)
Bile Acids and Salts/metabolism , Carrier Proteins/genetics , Carrier Proteins/physiology , Gene Deletion , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent , Symporters , Alleles , Animals , Biological Transport , Blotting, Northern , Cholesterol/blood , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol Esters/metabolism , DNA, Complementary/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutation , RNA/metabolism , Recombination, Genetic , Sex Factors , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL