Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Neonatal Screen ; 10(2)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38920847

ABSTRACT

Vitamin B12 (B12) deficiency (B12D) can have detrimental effects on early growth and development. The Austrian newborn screening (NBS) program targets inborn errors of cobalamin metabolism and also detects B12D. Of 59 included neonates with B12D suspected by NBS, B12D was not further investigated in 16 (27%) retrospectively identified cases, not confirmed in 28 (48%), and confirmed in 15 (25%) cases. NBS and recall biomarkers were recorded. Age at sampling of the dried blood spots for NBS and the 1st-tier methionine/phenylalanine ratio were the strongest parameters to predict B12D (67.4% correct allocations). No differences between cases with confirmed, unconfirmed, or unknown B12D or differences to norms were observed for growth and psychomotor development (Vineland III scales, phone interviews with parents of children between months 10 and 14 of life). B12 intake was below recommendations in most mothers. NBS can detect reduced intracellular B12 activity. No advantage of NBS detection and treatment regarding infant cognitive development or growth could be proven. Since conspicuous NBS findings cannot be ignored, and to prevent exposing newborns to invasive diagnostics, assessment of maternal B12 status during pregnancy seems advisable.

2.
BMC Pediatr ; 20(1): 340, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32646411

ABSTRACT

BACKGROUND: Idiopathic or transient neonatal cholestasis (TNC) represents a group of cholestatic disorders with unidentified origin and remains a diagnosis of exclusion. Dysfunction of hepatobiliary transporters mediating excretion of biliary constituents from hepatocytes may play a central role in the pathogenesis of cholestasis. Despite variants of bile salt (BS) export pump (BSEP/ABCB11) have already been described in TNC, the pathogenic role of BSEP dysfunction in TNC remained so far elusive. CASE PRESENTATION: We report on a newly-identified heterozygous ABCB11 missense variant (c.1345G > A, p.Glu449Lys) which was associated with prolonged cholestasis in a term infant after a complicated neonatal period. Moreover, we show for the first time almost completely abolished BSEP expression on the hepatocellular membrane in TNC. CONCLUSION: This report demonstrates for the first time a close association between the prolonged cholestasis in infancy and impaired BSEP expression on the hepatocyte canalicular membrane in a heterozygous carrier of newly-identified ABCB11 variant.


Subject(s)
Cholestasis , Liver Diseases , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Cholestasis/genetics , Hepatocytes , Humans , Infant , Infant, Newborn , Mutation, Missense
3.
J Hepatol ; 64(3): 674-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26529078

ABSTRACT

BACKGROUND AND AIMS: Approximately 95% of bile acids (BAs) excreted into bile are reabsorbed in the gut and circulate back to the liver for further biliary secretion. Therefore, pharmacological inhibition of the ileal apical sodium-dependent BA transporter (ASBT/SLC10A2) may protect against BA-mediated cholestatic liver and bile duct injury. METHODS: Eight week old Mdr2(-/-) (Abcb4(-/-)) mice (model of cholestatic liver injury and sclerosing cholangitis) received either a diet supplemented with A4250 (0.01% w/w) - a highly potent and selective ASBT inhibitor - or a chow diet. Liver injury was assessed biochemically and histologically after 4weeks of A4250 treatment. Expression profiles of genes involved in BA homeostasis, inflammation and fibrosis were assessed via RT-PCR from liver and ileum homogenates. Intestinal inflammation was assessed by RNA expression profiling and immunohistochemistry. Bile flow and composition, as well as biliary and fecal BA profiles were analyzed after 1week of ASBT inhibitor feeding. RESULTS: A4250 improved sclerosing cholangitis in Mdr2(-/-) mice and significantly reduced serum alanine aminotransferase, alkaline phosphatase and BAs levels, hepatic expression of pro-inflammatory (Tnf-α, Vcam1, Mcp-1) and pro-fibrogenic (Col1a1, Col1a2) genes and bile duct proliferation (mRNA and immunohistochemistry for cytokeratin 19 (CK19)). Furthermore, A4250 significantly reduced bile flow and biliary BA output, which correlated with reduced Bsep transcription, while Ntcp and Cyp7a1 were induced. Importantly A4250 significantly reduced biliary BA secretion but preserved HCO3(-) and biliary phospholipid secretion resulting in an increased HCO3(-)/BA and PL/BA ratio. In addition, A4250 profoundly increased fecal BA excretion without causing diarrhea and altered BA pool composition, resulting in diminished concentrations of primary BAs tauro-ß-muricholic acid and taurocholic acid. CONCLUSIONS: Pharmacological ASBT inhibition attenuates cholestatic liver and bile duct injury by reducing biliary BA concentrations in mice.


Subject(s)
Bile Acids and Salts/metabolism , Bile Ducts/drug effects , Cholangitis, Sclerosing/drug therapy , Cholestasis/drug therapy , Intestinal Absorption , Liver/drug effects , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Animals , Bile Ducts/injuries , Bile Ducts/pathology , Cholestasis/metabolism , Gallbladder/drug effects , Liver/pathology , Mice
4.
Hepatology ; 59(3): 858-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24002947

ABSTRACT

UNLABELLED: Hepatic inflammation is a key feature of progressive liver disease. Alterations of fatty acid (FA) metabolism and signaling may play an important role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD) and its progression to nonalcoholic steatohepatitis (NASH). Moreover, FAs activate peroxisome proliferator-activated receptor α (PPARα) as a key transcriptional regulator of hepatic FA metabolism and inflammation. Since adipose triglyceride lipase (ATGL/PNPLA2) is the key enzyme for intracellular hydrolysis of stored triglycerides and determines FA signaling through PPARα, we explored the role of ATGL in hepatic inflammation in mouse models of NASH and endotoxemia. Mice lacking ATGL or hormone-sensitive lipase (HSL) were challenged with a methionine-choline-deficient (MCD) diet as a nutritional model of NASH or lipopolysaccharide (LPS) as a model of acute hepatic inflammation. We further tested whether a PPARα agonist (fenofibrate) treatment improves the hepatic phenotype in MCD- or LPS-challenged ATGL-knockout (KO) mice. MCD-fed ATGL-KO mice, although partially protected from peripheral lipolysis, showed exacerbated hepatic steatosis and inflammation. Moreover, ATGL-KO mice challenged by LPS showed enhanced hepatic inflammation, increased mortality, and torpor, findings which were attributed to impaired PPARα DNA binding activity due to reduced FABP1 protein levels, resulting in impaired nuclear FA import. Notably, liganding PPARα through fenofibrate attenuated hepatic inflammation in both MCD-fed and LPS-treated ATGL-KO mice. In contrast, mice lacking HSL had a phenotype similar to the WT mice on MCD and LPS challenge. CONCLUSION: These findings unravel a novel protective role of ATGL against hepatic inflammation which could have important implications for metabolic and inflammatory liver diseases.


Subject(s)
Endotoxemia/immunology , Endotoxemia/metabolism , Fatty Liver/immunology , Fatty Liver/metabolism , Lipase/metabolism , Animals , Choline Deficiency/metabolism , Choline Deficiency/pathology , Disease Models, Animal , Female , Lipase/genetics , Lipase/immunology , Lipopolysaccharides/toxicity , Liver/immunology , Liver/metabolism , Male , Methionine/deficiency , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease , PPAR alpha/immunology , PPAR alpha/metabolism , Signal Transduction/immunology
5.
Mol Aspects Med ; 37: 57-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24333169

ABSTRACT

Hepatobiliary bile salt (BS) transporters are critical determinants of BS homeostasis controlling intracellular concentrations of BSs and their enterohepatic circulation. Genetic or acquired dysfunction of specific transport systems causes intrahepatic and systemic retention of potentially cytotoxic BSs, which, in high concentrations, may disturb integrity of cell membranes and subcellular organelles resulting in cell death, inflammation and fibrosis. Transcriptional regulation of canalicular BS efflux through bile salt export pump (BSEP), basolateral elimination through organic solute transporters alpha and beta (OSTα/OSTß) as well as inhibition of hepatocellular BS uptake through basolateral Na(+)-taurocholate cotransporting polypeptide (NTCP) represent critical steps in protection from hepatocellular BS overload and can be targeted therapeutically. In this article, we review the potential clinical implications of the major BS transporters BSEP, OSTα/OSTß and NTCP in the pathogenesis of hereditary and acquired cholestatic syndromes, provide an overview on transcriptional control of these transporters by the key regulatory nuclear receptors and discuss the potential therapeutic role of novel transcriptional activators of BS transporters in cholestasis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholestasis/genetics , Membrane Transport Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/metabolism , Bile Acids and Salts/metabolism , Biological Transport , Cholestasis/pathology , Cholestasis/therapy , Gene Expression Regulation , Humans , Membrane Transport Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/genetics , Symporters/metabolism , Transcriptional Activation/genetics
6.
Clin Liver Dis ; 17(2): 161-89, 2013 May.
Article in English | MEDLINE | ID: mdl-23540496

ABSTRACT

Cholestatic liver diseases encompass a wide spectrum of disorders with different causes, resulting in impaired bile flow and accumulation of bile acids and other potentially hepatotoxic cholephils. The understanding of the molecular mechanisms of bile formation and cholestasis has recently improved significantly through new insights into nuclear receptor (patho)biology. Nuclear receptors are ligand-activated transcription factors, which act as central players in the regulation of genes responsible for elimination and detoxification of biliary constituents accumulating in cholestasis. They also control other pathophysiologic processes such as inflammation, fibrogenesis, and carcinogenesis involved in the pathogenesis and disease progression of cholestasis liver diseases.


Subject(s)
Bile Acids and Salts/biosynthesis , Cholestasis/drug therapy , Cholestasis/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Animals , Cholagogues and Choleretics/therapeutic use , Cholestasis/genetics , Constitutive Androstane Receptor , Homeostasis/drug effects , Humans , Ligands , Peroxisome Proliferator-Activated Receptors/metabolism , Pregnane X Receptor , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Signal Transduction/genetics , Ursodeoxycholic Acid/therapeutic use
7.
Dig Dis ; 30 Suppl 1: 39-47, 2012.
Article in English | MEDLINE | ID: mdl-23075867

ABSTRACT

Primary sclerosing cholangitis (PSC) is a chronic inflammatory bile duct disease of unknown etiology, frequently associated with inflammatory bowel disease and leading to end-stage liver disease requiring liver transplantation. Moreover, PSC is a premalignant condition associated with an increased risk for hepatobiliary and colorectal malignancy. Since effective medical therapy for PSC is still lacking, this disorder represents a potentially fatal disease with poor prognosis. This article is a summary of an overview given at the 5th Falk Gastro Conference in Munich 2012 and reviews the challenges associated with diagnosis, surveillance and therapy of PSC.


Subject(s)
Cholangitis, Sclerosing/diagnosis , Cholangitis, Sclerosing/therapy , Population Surveillance , Cholangitis, Sclerosing/drug therapy , Cholangitis, Sclerosing/epidemiology , Humans , Ursodeoxycholic Acid/therapeutic use
8.
Hepatology ; 54(4): 1303-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22006858

ABSTRACT

UNLABELLED: Chronic cholangiopathies have limited therapeutic options and represent an important indication for liver transplantation. The nuclear farnesoid X receptor (FXR) and the membrane G protein-coupled receptor, TGR5, regulate bile acid (BA) homeostasis and inflammation. Therefore, we hypothesized that activation of FXR and/or TGR5 could ameliorate liver injury in Mdr2(-/-) (Abcb4(-/-)) mice, a model of chronic cholangiopathy. Hepatic inflammation, fibrosis, as well as bile secretion and key genes of BA homeostasis were addressed in Mdr2(-/-) mice fed either a chow diet or a diet supplemented with the FXR agonist, INT-747, the TGR5 agonist, INT-777, or the dual FXR/TGR5 agonist, INT-767 (0.03% w/w). Only the dual FXR/TGR5 agonist, INT-767, significantly improved serum liver enzymes, hepatic inflammation, and biliary fibrosis in Mdr2(-/-) mice, whereas INT-747 and INT-777 had no hepatoprotective effects. In line with this, INT-767 significantly induced bile flow and biliary HCO 3- output, as well as gene expression of carbonic anhydrase 14, an important enzyme able to enhance HCO 3- transport, in an Fxr-dependent manner. In addition, INT-767 dramatically reduced bile acid synthesis via the induction of ileal Fgf15 and hepatic Shp gene expression, thus resulting in significantly reduced biliary bile acid output in Mdr2(-/-) mice. CONCLUSION: This study shows that FXR activation improves liver injury in a mouse model of chronic cholangiopathy by reduction of biliary BA output and promotion of HCO 3--rich bile secretion.


Subject(s)
Adenosine Triphosphatases/metabolism , Anion Transport Proteins/metabolism , Biliary Tract Diseases/drug therapy , Cholic Acids/pharmacology , Liver Diseases/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Analysis of Variance , Animals , Bile Acids and Salts/metabolism , Biliary Tract Diseases/prevention & control , Disease Models, Animal , Liver Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Random Allocation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Statistics, Nonparametric , ATP-Binding Cassette Sub-Family B Member 4
9.
J Clin Invest ; 121(9): 3724-34, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21804189

ABSTRACT

High plasma concentrations of lipoprotein(a) [Lp(a), which is encoded by the APOA gene] increase an individual's risk of developing diseases, such as coronary artery diseases, restenosis, and stroke. Unfortunately, increased Lp(a) levels are minimally influenced by dietary changes or drug treatment. Further, the development of Lp(a)-specific medications has been hampered by limited knowledge of Lp(a) metabolism. In this study, we identified patients suffering from biliary obstructions with very low plasma Lp(a) concentrations that rise substantially after surgical intervention. Consistent with this, common bile duct ligation in mice transgenic for human APOA (tg-APOA mice) lowered plasma concentrations and hepatic expression of APOA. To test whether farnesoid X receptor (FXR), which is activated by bile acids, was responsible for the low plasma Lp(a) levels in cholestatic patients and mice, we treated tg-APOA and tg-APOA/Fxr-/- mice with cholic acid. FXR activation markedly reduced plasma concentrations and hepatic expression of human APOA in tg-APOA mice but not in tg-APOA/Fxr-/- mice. Incubation of primary hepatocytes from tg-APOA mice with bile acids dose dependently downregulated APOA expression. Further analysis determined that the direct repeat 1 element between nucleotides -826 and -814 of the APOA promoter functioned as a negative FXR response element. This motif is also bound by hepatocyte nuclear factor 4α (HNF4α), which promotes APOA transcription, and FXR was shown to compete with HNF4α for binding to this motif. These findings may have important implications in the development of Lp(a)-lowering medications.


Subject(s)
Apolipoproteins A/genetics , Apolipoproteins A/metabolism , Gene Expression , Liver/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/administration & dosage , Bile Acids and Salts/blood , Cells, Cultured , Female , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Jaundice, Obstructive/blood , Liver/cytology , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/metabolism , Random Allocation , Receptors, Cytoplasmic and Nuclear/genetics
10.
Dig Dis ; 29(1): 98-102, 2011.
Article in English | MEDLINE | ID: mdl-21691114

ABSTRACT

Bile acids (BAs) are able to activate a range of dedicated nuclear receptors (NRs) which play a key role in the transcriptional control of critical steps of a wide range of hepatic functions ranging from BA homeostasis and bile formation, phase I/II metabolism of endo- and xenobiotics such as BAs and drugs, respectively, to hepatic lipids and glucose metabolism. Apart from these metabolic roles, BA-activated nuclear receptors also play a key role in the control of hepatic inflammation, fibrogenesis, replication of hepatitis B and C virus, liver regeneration and carcinogenesis. As such, several physiological and pathophysiological effects of BAs can now be explained through activation of regulatory NR networks. Moreover, BA-activated NRs are key for understanding the pathogenesis of several liver diseases and represent attractive drug targets. This article will provide a brief overview on the role of BA-activated NRs in cholestatic and fatty liver disease.


Subject(s)
Bile Acids and Salts/metabolism , Liver Diseases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cholestasis/metabolism , Homeostasis , Humans , Lipid Metabolism
11.
Dig Dis ; 28(1): 99-104, 2010.
Article in English | MEDLINE | ID: mdl-20460897

ABSTRACT

Improving our understanding of the pathogenesis of chronic immune-mediated cholangiopathies such as primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC), as well as the development of novel diagnostic, prognostic and therapeutic tools for these disorders critically depends on easily reproducible animal models. Recently, several spontaneous mouse models for PBC (not requiring previous manipulations for breakdown of immunotolerance) have been reported, including NOD.c3c4 and NOD.c3c4-derived mice, IL-2Ralpha(-/-) mice, dominant negative TGF-beta receptor II mice and Ae2(a,b)(-/-) mice. To date, no animal model exhibits all of the attributes of PSC. Rodent models induced by bacterial cell components or colitis may help to explain the strong association between PSC and inflammatory bowel disease. Other models include direct injury to biliary epithelia, peribiliary vascular endothelia or portal venous endothelia. Mice with targeted disruption of the Mdr2 (Abcb4) gene encoding a canalicular phospholipid flippase (Mdr2(-/-) mice) spontaneously develop sclerosing cholangitis with macroscopic and microscopic features of human PSC. Another example for a transporter involved in the pathogenesis of sclerosing cholangitis is the cystic fibrosis transmembrane conductance regulator (CFTR/ABCC7). Xenobiotics and drugs may also lead to bile duct injury and biliary fibrosis via direct toxic and indirect immune-mediated injury. Hydrophobic bile acids, such as lithocholic acid, cause bile duct injury and destructive cholangitis with periductal fibrosis resembling sclerosing cholangitis. These models have enhanced our understanding of the pathogenesis of PBC and PSC and will hopefully result in improved treatment of these disorders.


Subject(s)
Autoimmune Diseases/immunology , Cholangitis/immunology , Disease Models, Animal , Animals , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/physiopathology , Cholangitis, Sclerosing/therapy , Humans , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/physiopathology , Mice , Mice, Knockout
12.
Gut ; 59(4): 521-30, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20332524

ABSTRACT

BACKGROUND AND AIM: Chronic cholangiopathies have limited therapeutic options and represent an important indication for liver transplantation. Curcumin, the yellow pigment of the spice turmeric, has pleiotropic actions and attenuates hepatic damage in animal models of chemically-induced liver injury. Whether curcumin has beneficial effects in cholangiopathies is unknown. METHODS: Potential anticholestatic, anti-inflammatory and antifibrotic mechanisms of curcumin were explored in vivo in Mdr2(-/-) mice as a murine model of chronic cholangiopathy; as well as in vitro in a cholangiocyte cell line (HuCCT1) and portal myofibroblasts (MFBs) isolated from Mdr2(-/-) mice. RESULTS: Liver damage, cholestasis and fibrosis were reduced in Mdr2(-/-) mice after curcumin feeding. Moreover, curcumin inhibited cholangiocyte proliferation and expression of activation marker vascular cell adhesion molecule-1 in Mdr2(-/-) mice. Curcumin-similar to PPARgamma synthetic agonist troglitazone-directly inhibited TNF-alpha-induced inflammatory activation of cholangiocytes in vitro, whereas these beneficial effects of curcumin were largely blocked by a PPARgamma synthetic antagonist. In addition, curcumin blocked proliferation and activation of portal MFBs by inhibiting ERK1/2 phosphorylation, thus contributing to reduced fibrogenesis. CONCLUSIONS: These results show that curcumin may have multiple targets in liver including activation of PPARgamma in cholangiocytes and inhibition of ERK1/2 signalling in MFBs, thereby modulating several central cellular events in a mouse model of cholangiopathy. Targeting these pathways may be a promising therapeutic approach to cholangiopathies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cholangitis, Sclerosing/drug therapy , Curcumin/therapeutic use , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bile/metabolism , Bile Acids and Salts/biosynthesis , Bile Ducts/drug effects , Bile Ducts/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Curcumin/pharmacology , Drug Evaluation, Preclinical/methods , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Inflammation Mediators/metabolism , Liver Cirrhosis, Experimental/drug therapy , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PPAR gamma/metabolism , Signal Transduction/drug effects , ATP-Binding Cassette Sub-Family B Member 4
13.
Liver Int ; 28(7): 948-58, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18410282

ABSTRACT

BACKGROUND/AIMS: Multidrug resistance protein 2 (Abcb4) gene knockout mice (Mdr2(-/-)) lack phosphatidylcholine (PC) excretion into bile and spontaneously develop sclerosing cholangitis, biliary fibrosis and hepatocellular carcinomas. We therefore aimed to test whether formation and hepatic retention of abnormal PC metabolites contribute to the pathogenesis of liver injury in Mdr2(-/-) mice. METHODS: Mdr2(-/-) mice were either fed a diet supplemented with soybean lecithin 2.5% w/w [phosphatidylcholine-enriched diet (PCD), to increase hepatic PC content] or a choline-deficient diet (CDD, to reduce hepatic PC content) for 4 weeks; controls received chow with energy and nutrient content equivalent to PCD and CDD. Serum liver tests, liver histology, markers of fibrosis, cholangiocyte activation, cell proliferation and thin-layer chromatography for phospholipid (PL) composition were carried out. RESULTS: PCD decreased serum alkaline phosphatase and total bilirubin levels compared with controls, while liver histology as well as hepatic hydroxyproline content as markers of liver fibrosis did not differ among groups. Both PCD and CDD decreased hepatocellular proliferation compared with controls. Hepatic, serum and biliary PLs remained unchanged despite dietary manipulations and no potentially toxic PL metabolites were detected. CONCLUSIONS: Mdr2(-/-) mice maintain stable hepatic, serum and biliary PL metabolism in response to dietary PC manipulations. Our findings therefore suggest that liver injury in Mdr2(-/-) mice is not due to formation of toxic PL metabolites.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Cholestasis, Intrahepatic/metabolism , Choline Deficiency/metabolism , Disease Models, Animal , Lecithins/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Alkaline Phosphatase/blood , Animals , Bile/metabolism , Bilirubin/blood , Cell Proliferation/drug effects , Cholestasis, Intrahepatic/chemically induced , Cholestasis, Intrahepatic/pathology , Cholesterol/metabolism , Choline Deficiency/pathology , Diet , Drug Resistance, Multiple , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Lecithins/administration & dosage , Liver/drug effects , Liver/pathology , Liver Function Tests , Male , Mice , Mice, Knockout , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL
...