Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Hepatology ; 56(1): 270-80, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22271167

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is characterized by triglyceride (TG) accumulation and endoplasmic reticulum (ER) stress. Because fatty acids (FAs) may trigger ER stress, we hypothesized that the absence of adipose triglyceride lipase (ATGL/PNPLA2)-the main enzyme for intracellular lipolysis, releasing FAs, and closest homolog to adiponutrin (PNPLA3) recently implicated in the pathogenesis of NAFLD-protects against hepatic ER stress. Wild-type (WT) and ATGL knockout (KO) mice were challenged with tunicamycin (TM) to induce ER stress. Serum biochemistry, hepatic TG and FA profiles, liver histology, and gene expression for markers of hepatic lipid metabolism, ER stress, and inflammation were explored. Moreover, cell-culture experiments were performed in Hepa1.6 cells after the knockdown of ATGL before FA and TM treatment. TM increased hepatic TG accumulation in ATGL KO, but not in WT, mice. Lipogenesis and ß-oxidation were repressed at the gene-expression level (sterol regulatory element-binding transcription factor 1c, fatty acid synthase, acetyl coenzyme A carboxylase 2, and carnitine palmitoyltransferase 1 alpha) in both WT and ATGL KO mice. Genes for very-low-density lipoprotein (VLDL) synthesis (microsomal triglyceride transfer protein and apolipoprotein B) were down-regulated by TM in WT and even more in ATGL KO mice, which displayed strongly reduced serum VLDL cholesterol levels. Notably, ER stress markers glucose-regulated protein, C/EBP homolog protein, spliced X-box-binding protein, endoplasmic-reticulum-localized DnaJ homolog 4, and inflammatory markers Tnfα and iNos were induced exclusively in TM-treated WT, but not ATGL KO, mice. Total hepatic FA profiling revealed a higher palmitic acid/oleic acid (PA/OA) ratio in WT mice, compared to ATGL KO mice, at baseline. Phosphoinositide-3-kinase inhibitor-known to be involved in FA-derived ER stress and blocked by OA-was increased in TM-treated WT mice only. In line with this, in vitro OA protected hepatocytes from TM-induced ER stress. CONCLUSIONS: Lack of ATGL may protect from hepatic ER stress through alterations in FA composition. ATGL could constitute a new therapeutic strategy to target ER stress in NAFLD.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Fatty Liver/metabolism , Lipase/metabolism , Lipogenesis/physiology , Animals , Blotting, Western , Cells, Cultured , Cholesterol , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Fatty Liver/chemically induced , Fatty Liver/pathology , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Immunohistochemistry , Lipase/genetics , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipogenesis/genetics , Lipoproteins , Lipoproteins, VLDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease , RNA, Messenger/metabolism , Random Allocation , Real-Time Polymerase Chain Reaction , Triglycerides/metabolism , Tunicamycin/pharmacology
2.
Gastroenterology ; 142(1): 140-151.e12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22001865

ABSTRACT

BACKGROUND & AIMS: The liver controls central processes of lipid and bile acid homeostasis. We aimed to investigate whether alterations in lipid metabolism contribute to the pathogenesis of chronic cholestatic liver disease in mice. METHODS: We used microarray and metabolic profiling analyses to identify alterations in systemic and hepatic lipid metabolism in mice with disruption of the gene ATP-binding cassette sub-family B member 4 (Abcb4(-/-) mice), a model of inflammation-induced cholestatic liver injury, fibrosis, and cancer. RESULTS: Alterations in Abcb4(-/-) mice, compared with wild-type mice, included deregulation of genes that control lipid synthesis, storage, and oxidation; decreased serum levels of cholesterol and phospholipids; and reduced hepatic long-chain fatty acyl-CoAs (LCA-CoA). Feeding Abcb4(-/-) mice the side chain-modified bile acid 24-norursodeoxycholic acid (norUDCA) reversed their liver injury and fibrosis, increased serum levels of lipids, lowered phospholipase and triglyceride hydrolase activities, and restored hepatic LCA-CoA and triglyceride levels. Additional genetic and nutritional studies indicated that lipid metabolism contributed to chronic cholestatic liver injury; crossing peroxisome proliferator-activated receptor (PPAR)-α-deficient mice with Abcb4(-/-) mice (to create double knockouts) or placing Abcb4(-/-) mice on a high-fat diet protected against liver injury, with features similar to those involved in the response to norUDCA. Placing pregnant Abcb4(-/-) mice on high-fat diets prevented liver injury in their offspring. However, fenofibrate, an activator of PPARα, aggravated liver injury in Abcb4(-/-) mice. CONCLUSIONS: Alterations in lipid metabolism contribute to the pathogenesis and progression of cholestatic liver disease in mice.


Subject(s)
Cell Proliferation , Cholestasis, Intrahepatic/metabolism , Hepatitis/metabolism , Lipid Metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Cholestasis, Intrahepatic/drug therapy , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/pathology , Chronic Disease , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Disease Models, Animal , Disease Progression , Fatty Acids/metabolism , Female , Fenofibrate/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation , Hepatitis/drug therapy , Hepatitis/genetics , Hepatitis/pathology , Hypolipidemic Agents/pharmacology , Lipid Metabolism/genetics , Liver/drug effects , Liver/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Metabolomics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , PPAR gamma/deficiency , PPAR gamma/genetics , Pregnancy , Prenatal Exposure Delayed Effects , Triglycerides/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/pharmacology , ATP-Binding Cassette Sub-Family B Member 4
3.
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
4.
Lab Invest ; 90(6): 844-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20368698

ABSTRACT

Proinflammatory and profibrotic cytokines such as osteopontin (OPN) and tumor necrosis factor-alpha receptor-1 (TNFR(1)) may be critically involved in the pathogenesis of cholangiopathies and biliary fibrosis. We therefore aimed to determine the role of genetic loss of either OPN or TNFR(1) in 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-fed mice as a model of xenobiotic-induced sclerosing cholangitis with biliary-type liver fibrosis using respective knock-out mice. OPN and TNFR(1) knock-out mice were fed a 0.1% DDC-supplemented diet for 4 weeks and compared with corresponding wild-type (WT) controls. Liver morphology (H&E staining), serum markers of liver injury and cholestasis (ALT, AP, bilirubin), markers of inflammation in liver (CD11b and F4/80 immunostaining, mRNA expression of iNOS, MCP-1, IL-1beta, INF-gamma, TNF-alpha and OPN), degree of ductular reaction (immunohistochemistry with morphometric analysis and western blotting for cholangiocyte-specific marker keratin 19) and degree of liver fibrosis (Sirius-red staining, hepatic hydroxyproline content for quantification) were compared between groups. DDC feeding in OPN and TNFR(1) knock-out mice and respective WT controls resulted in comparable extent of liver injury, inflammatory response, ductular reaction and liver fibrosis. Our data indicate that genetic loss of neither OPN nor TNFR(1) significantly effects on the pathogenesis of DDC-induced sclerosing cholangitis, ductular reaction and resulting biliary fibrosis.


Subject(s)
Cholangitis/immunology , Gallbladder Diseases/immunology , Osteopontin/physiology , Animals , Chemokine CCL2/immunology , Cholangitis/pathology , Disease Models, Animal , Gallbladder Diseases/pathology , Immunohistochemistry , Inflammation/pathology , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
Gastroenterology ; 138(7): 2499-508, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20193684

ABSTRACT

BACKGROUND & AIMS: Signal transducer and activator of transcription 3 (Stat3) is the main mediator of interleukin-6-type cytokine signaling required for hepatocyte proliferation and hepatoprotection, but its role in sclerosing cholangitis and other cholestatic liver diseases remains unresolved. METHODS: We investigated the role of Stat3 in inflammation-induced cholestatic liver injury and used mice lacking the multidrug resistance gene 2 (mdr2(-/-)) as a model for SC. RESULTS: We show that conditional inactivation of Stat3 in hepatocytes and cholangiocytes (stat3(Deltahc)) of mdr2(-/-) mice strongly aggravated bile acid-induced liver injury and fibrosis. A similar phenotype was observed in mdr2(-/-) mice lacking interleukin-6 production. Biochemical and molecular characterization suggested that Stat3 exerts hepatoprotective functions in both hepatocytes and cholangiocytes. Loss of Stat3 led to increased expression of tumor necrosis factor alpha, which might reduce the barrier function of bile ducts. Moreover, Stat3-deficient hepatocytes displayed up-regulation of bile acid biosynthesis genes and down-regulation of hepatoprotective epidermal growth factor receptor and insulin-like growth factor 1 signaling pathways. Consistently, stat3(Deltahc) mice were more sensitive to cholic acid-induced liver damage than control mice. CONCLUSIONS: Our data suggest that Stat3 prevents cholestasis and liver damage in sclerosing cholangitis via regulation of pivotal functions in hepatocytes and cholangiocytes.


Subject(s)
Cholangitis, Sclerosing/complications , Cytoprotection , Liver Cirrhosis, Experimental/prevention & control , STAT3 Transcription Factor/physiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Bile Acids and Salts/toxicity , Cell Proliferation , Liver/drug effects , Liver Regeneration , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , ATP-Binding Cassette Sub-Family B Member 4
6.
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
7.
Hepatology ; 51(4): 1319-26, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20162728

ABSTRACT

UNLABELLED: Growth hormone (GH) resistance and low serum levels of insulinlike growth factor 1 (IGF-1) are common features in human liver fibrosis and cirrhosis. Signal transducer and activator of transcription 5 (STAT5) controls several vital functions in the liver, including GH-mediated transcription of IGF-1. To investigate the role of STAT5 in liver fibrogenesis, we specifically deleted the Stat5a/b locus both in hepatocytes and cholangiocytes in the multidrug resistance gene 2 knockout (Mdr2(-/-)) mouse model of cholestasis. Double knockout mice develop an early and severe liver fibrosis phenotype, accompanied by perturbed expression of key regulators of bile acid homeostasis. Deletion of Stat5 resulted in GH resistance, and IGF-1 levels in serum were undetectable. We could observe reduced expression of important hepatoprotective genes, such as epidermal growth factor receptor (Egfr), hepatocyte nuclear factor 6 (Hnf6), prolactin receptor (Prlr), and leukemia inhibitory factor receptor (Lifr) as well as increased numbers of apoptotic hepatocytes. CONCLUSION: Our data suggest that loss of STAT5 sensitizes hepatocytes to bile acid-induced damage and apoptosis caused by disruption of GH-induced transcription of Igf-1 and down-regulation of hepatoprotective genes. These findings could contribute to the understanding of liver fibrosis and future treatment strategies for liver fibrosis.


Subject(s)
Cholestasis/complications , Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Liver Cirrhosis, Experimental/etiology , STAT5 Transcription Factor/physiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Apoptosis , Disease Models, Animal , ErbB Receptors/genetics , Hepatocyte Nuclear Factor 6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , ATP-Binding Cassette Sub-Family B Member 4
8.
Trop Med Int Health ; 14(11): 1418-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19747186

ABSTRACT

OBJECTIVES: The murine model of Schistosoma mansoni infection is characterized by strong fibrosis and little hepatocellular injury. The objective of this study was to evaluate the potential link between hepatic schistosomiasis and bile duct injury in relation to the expression of profibrotic cytokines and fibrosis-related genes. METHODS: Hepatic schistosomiasis was induced via percutaneous infection of mice with 50 S. mansoni cercariae. Markers of fibrosis including matrixmetalloproteinases (MMPs) and tissue-inhibitors of metalloproteinases (TIMPs), as well as markers of bile duct injury (keratin-19, VCAM-1) were studied during 24 weeks after infection by RT-PCR and immunohistochemistry. RESULTS: Liver biochemistry revealed no differences in serum transaminase and alkaline phosphatase levels in infected and uninfected mice. Total liver hydroxyproline content was increased 5-fold (P < 0.05) after infection. Gene expression analysis revealed MMP-2 (12-fold, P < 0.05) and TIMP-1 (48-fold, P < 0.05) up-regulation after infection. The balance of MMP and TIMP was shifted towards TIMP. Bile ducts were engulfed by adjacent granulomas resulting in ductular proliferation (keratin-19). VCAM-1 expression and inflammatory infiltrates were reduced. CONCLUSIONS: This study demonstrates that schistosomiasis is associated with (i) an imbalance of MMP-2 and TIMP-1 as key players of fibrogenesis and (ii) with secondary bile duct alterations leading to ductular proliferation possibly contributing to fibrosis.


Subject(s)
Bile Duct Diseases/metabolism , Liver Cirrhosis/genetics , Schistosomiasis mansoni/genetics , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Cytokines/metabolism , Keratin-19/metabolism , Liver Cirrhosis/enzymology , Metalloproteases/analysis , Metalloproteases/metabolism , Mice , Models, Animal , Reverse Transcriptase Polymerase Chain Reaction , Schistosoma mansoni/genetics , Tissue Inhibitor of Metalloproteinases/analysis , Tissue Inhibitor of Metalloproteinases/genetics , Transaminases/metabolism , Vascular Cell Adhesion Molecule-1/genetics
9.
Liver Int ; 29(9): 1316-25, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19558576

ABSTRACT

BACKGROUND AND AIMS: The pathogenetic link between ulcerative colitis and sclerosing cholangitis (SC) is unclear. We hypothesized that colitis induces changes in bile composition via inflammation-induced reduction of hepatobiliary transporter gene expression, ultimately resulting in cholestasis and bile duct injury. METHODS: Alterations in transporter expression and bile secretion in acute dextran sulphate sodium (DSS)-induced colitis were compared with lipopolysaccharide (LPS)-treated mice serving as positive control. Whether chronic DSS-colitis elicits cholangitis in genetically predisposed animals was studied in heterozygous multidrug resistance gene 2 knockout mice (Mdr2(+/-)). RESULTS: LPS but not DSS-colitis changed major hepatobiliary transporters (Ntcp, Bsep, Mrp2-4, Ostalpha/beta, Abcg5/8, Oatp1-4, Mdr1b and Mdr2), enzymes (Cyp3a11 and Cyp7a1), nuclear receptors (RXRalpha, FXR, CAR and PXR) and proinflammatory mediators (tumour necrosis factor alpha and inducible nitric oxide synthase). Formation of toxic bile reflected by an increased bile acid/phospholipid ratio was observed neither in acute nor in chronic colitis, although heterozygous Mdr2(+/-) mice developed mild portal inflammation after chronic colitis. CONCLUSIONS: In contrast to LPS, DSS-colitis has a minor impact on hepatobiliary gene expression and bile secretion. Therefore, intestinal inflammation-associated changes of hepatobiliary transporter expression do not play a pathogenetic role in SC.


Subject(s)
Cholangitis, Sclerosing/etiology , Colitis/metabolism , Animals , Bacterial Translocation , Bile/chemistry , Bile/metabolism , Bile Acids and Salts/analysis , Bile Ducts/metabolism , Carrier Proteins/genetics , Colitis/chemically induced , Colitis/complications , Dextran Sulfate/toxicity , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Phospholipids/analysis , RNA, Messenger/analysis
10.
Hepatology ; 49(6): 1972-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19475687

ABSTRACT

UNLABELLED: 24-norursodeoxycholic acid (norUDCA), a side chain-modified ursodeoxycholic acid derivative, has dramatic therapeutic effects in experimental cholestasis and may be a promising agent for the treatment of cholestatic liver diseases. We aimed to better understand the physiologic and therapeutic properties of norUDCA and to test if they are related to its side chain length and/or relative resistance to amidation. For this purpose, Mdr2(-/-) mice, a model for sclerosing cholangitis, received either a standard diet or a norUDCA-, tauro norursodeoxycholic acid (tauro- norUDCA)-, or di norursodeoxycholic acid (di norUDCA)-enriched diet. Bile composition, serum biochemistry, liver histology, fibrosis, and expression of key detoxification and transport systems were investigated. Direct choleretic effects were addressed in isolated bile duct units. The role of Cftr for norUDCA-induced choleresis was explored in Cftr(-/-) mice. norUDCA had pharmacologic features that were not shared by its derivatives, including the increase in hepatic and serum bile acid levels and a strong stimulation of biliary HCO(3)(-)-output. norUDCA directly stimulated fluid secretion in isolated bile duct units in a HCO(3)(-)-dependent fashion to a higher extent than the other bile acids. Notably, the norUDCA significantly stimulated HCO(3)(-)-output also in Cftr(-/-) mice. In Mdr2(-/-) mice, cholangitis and fibrosis strongly improved with norUDCA, remained unchanged with tauro- norUDCA, and worsened with di norUDCA. Expression of Mrp4, Cyp2b10, and Sult2a1 was increased by norUDCA and di norUDCA, but was unaffected by tauro- norUDCA. CONCLUSION: The relative resistance of norUDCA to amidation may explain its unique physiologic and pharmacologic properties. These include the ability to undergo cholehepatic shunting and to directly stimulate cholangiocyte secretion, both resulting in a HCO(3)(-)-rich hypercholeresis that protects the liver from cholestatic injury.


Subject(s)
Cholestasis, Intrahepatic/prevention & control , Ursodeoxycholic Acid/analogs & derivatives , Animals , Male , Mice , Ursodeoxycholic Acid/chemistry , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use
11.
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
12.
Liver Int ; 27(7): 920-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17696930

ABSTRACT

BACKGROUND: Bile acid synthesis, transport and metabolism are markedly altered in experimental cholestasis. Whether such coordinated regulation exists in human cholestatic diseases is unclear. We therefore investigated expression of genes for bile acid synthesis, detoxification and alternative basolateral export and regulatory nuclear factors in primary biliary cirrhosis (PBC). MATERIAL/METHODS: Hepatic CYP7A1, CYP27A1, CYP8B1 (bile acid synthesis), CYP3A4 (hydroxylation), SULT2A1 (sulphation), UGT2B4/2B7 (glucuronidation), MRP4 (basolateral export), farnesoid X receptor (FXR), retinoid X receptor (RXR), short heterodimer partner (SHP), hepatocyte nuclear factor 1alpha (HNF1alpha) and HNF4alpha expression was determined in 11 patients with late-stage PBC and this was compared with non-cholestatic controls. RESULTS: CYP7A1 mRNA was repressed in PBC to 10-20% of controls, while CYP27 and CYP8B1 mRNA remained unchanged. SULT2A1, UGT2B4/2B7 and CYP3A4 mRNA levels were unaltered or only mildly reduced in PBC. MRP4 protein levels were induced three-fold in PBC, whereas mRNA levels remained unchanged. Expression levels of FXR, RXR, SHP, PXR, CAR, HNF1alpha and HNF4alpha were moderately reduced in PBC without reaching statistical significance. SUMMARY/CONCLUSIONS: Repression of bile acid synthesis and induction of basolateral bile acid export may represent adaptive mechanisms to limit bile acid burden in chronic cholestasis. As these changes do not sufficiently counteract cholestatic liver damage, future therapeutic strategies should aim at stimulation of bile acid detoxification pathways.


Subject(s)
Bile Acids and Salts/metabolism , Cytochrome P-450 Enzyme System/analysis , Gene Expression Regulation , Glucuronosyltransferase/analysis , Liver Cirrhosis, Biliary/metabolism , Liver/metabolism , Multidrug Resistance-Associated Proteins/analysis , Sulfotransferases/analysis , Case-Control Studies , Cytochrome P-450 Enzyme System/genetics , Female , Glucuronosyltransferase/genetics , Humans , Liver/enzymology , Liver Cirrhosis, Biliary/enzymology , Liver Cirrhosis, Biliary/genetics , Male , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Severity of Illness Index , Sulfotransferases/genetics , Transcription Factors/analysis , Transcription Factors/genetics
13.
Drug Metab Dispos ; 35(9): 1694-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17576807

ABSTRACT

Liver injury in intercellular adhesion molecule 1 knockout (ICAM(-/-)) and Fas receptor-deficient (lpr) mice is markedly reduced after common bile duct ligation (CBDL) due to significantly reduced inflammation and oxidative stress. Liver injury in CBDL rodents is counteracted by adaptive hepatobiliary transporter induction. Since hepatobiliary transporter expression in obstructive cholestasis may be regulated not only by accumulating bile acids but also by inflammatory mediators and oxidative stress, we hypothesized that differences in the inflammatory response may affect hepatobiliary transporter expression in CBDL, which would contribute to reduced liver injury. Therefore, expression of major hepatobiliary transporters (Ntcp, Bsep, Mrp2-4, Ost alpha/beta) was determined by Taqman RT-PCR and Western blotting in sham-operated animals and 3 days after CBDL in wild-type, ICAM(-/-) and lpr mice of the endotoxin-sensitive C57BL/6 and the endotoxin-resistant C3H/HeJ strains. CBDL resulted in a significant decrease of Ntcp in all genotypes. Canalicular transporters Bsep and Mrp2 were repressed only in the endotoxin-sensitive strain regardless of the genotype. Mrp3 was moderately induced in ICAM(-/-), lpr, and endotoxin-resistant mice, whereas Mrp4 was only induced in the endotoxin-resistant strain. Ost beta was massively induced in all CBDL mice, whereas Ost alpha was reduced. In conclusion, markedly reduced inflammation and oxidative stress in CBDL ICAM(-/-) and lpr mice does not profoundly affect hepatobiliary transporter expression. Therefore, transporter expression does not account for reduced liver injury in ICAM(-/-) and lpr mice. Induction of the adaptive transporter response after CBDL is independent of the degree of the inflammatory response. Rather, retention of biliary constituents may determine transporter expression in CBDL.


Subject(s)
Bile/metabolism , Carrier Proteins/metabolism , Common Bile Duct/physiology , Inflammation/metabolism , Intercellular Adhesion Molecule-1/genetics , Liver/metabolism , Oxidative Stress/physiology , fas Receptor/deficiency , Animals , Bile Canaliculi/metabolism , Blotting, Western , Common Bile Duct/metabolism , Endotoxins/toxicity , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/genetics
14.
Am J Physiol Gastrointest Liver Physiol ; 290(5): G923-32, 2006 May.
Article in English | MEDLINE | ID: mdl-16357057

ABSTRACT

The bile acid receptor farnesoid X receptor (FXR) is a key regulator of hepatic defense mechanisms against bile acids. A comprehensive study addressing the role of FXR in the coordinated regulation of adaptive mechanisms including biosynthesis, metabolism, and alternative export together with their functional significance is lacking. We therefore fed FXR knockout (FXR(-/-)) mice with cholic acid (CA) and ursodeoxycholic acid (UDCA). Bile acid synthesis and hydroxylation were assessed by real-time RT-PCR for cytochrome P-450 (Cyp)7a1, Cyp3a11, and Cyp2b10 and mass spectrometry-gas chromatography for determination of bile acid composition. Expression of the export systems multidrug resistance proteins (Mrp)4-6 in the liver and kidney and the recently identified basoalteral bile acid transporter, organic solute transporter (Ost-alpha/Ost-beta), in the liver, kidney, and intestine was also investigated. CA and UDCA repressed Cyp7a1 in FXR(+/+) mice and to lesser extents in FXR(-/-) mice and induced Cyp3a11 and Cyp2b10 independent of FXR. CA and UDCA were hydroxylated in both genotypes. CA induced Ost-alpha/Ost-beta in the liver, kidney, and ileum in FXR(+/+) but not FXR(-/-) mice, whereas UDCA had only minor effects. Mrp4 induction in the liver and kidney correlated with bile acid levels and was observed in UDCA-fed and CA-fed FXR(-/-) animals but not in CA-fed FXR(+/+) animals. Mrp5/6 remained unaffected by bile acid treatment. In conclusion, we identified Ost-alpha/Ost-beta as a novel FXR target. Absent Ost-alpha/Ost-beta induction in CA-fed FXR(-/-) animals may contribute to increased liver injury in these animals. The induction of bile acid hydroxylation and Mrp4 was independent of FXR but could not counteract liver toxicity sufficiently. Limited effects of UDCA on Ost-alpha/Ost-beta may jeopardize its therapeutic efficacy.


Subject(s)
Bile Acids and Salts/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Membrane Transport Proteins/metabolism , Transcription Factors/genetics , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/chemistry , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholic Acid/pharmacology , Cholic Acid/toxicity , Cytochrome P-450 CYP3A/metabolism , Cytochrome P450 Family 2 , Kidney/metabolism , Liver/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear , Steroid Hydroxylases/metabolism , Symporters , Ursodeoxycholic Acid/pharmacology
15.
J Lipid Res ; 47(3): 582-92, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16327028

ABSTRACT

Farnesoid X receptor knockout (Fxr(-/-)) mice cannot upregulate the bile salt export pump in bile acid loading or cholestatic conditions. To investigate whether Fxr(-/-) mice differ in bile acid detoxification compared with wild-type mice, we performed a comprehensive analysis of bile acids extracted from liver, bile, serum, and urine of naive and common bile duct-ligated wild-type and Fxr(-/-) mice using electrospray and gas chromatography mass spectrometry. In addition, hepatic and renal gene expression levels of Cyp2b10 and Cyp3a11, and protein expression levels of putative renal bile acid-transporting proteins, were investigated. We found significantly enhanced hepatic bile acid hydroxylation in Fxr(-/-) mice, in particular hydroxylations of cholic acid in the 1beta, 2beta, 4beta, 6alpha, 6beta, 22, or 23 position and a significantly enhanced excretion of these metabolites in urine. The gene expression level of Cyp3a11 was increased in the liver of Fxr(-/-) mice, whereas the protein expression levels of multidrug resistance-related protein 4 (Mrp4) were increased in kidneys of both genotypes during common bile duct ligation. In conclusion, Fxr(-/-) mice detoxify accumulating bile acids in the liver by enhanced hydroxylation reactions probably catalyzed by Cyp3a11. The metabolites formed were excreted into urine, most likely with the participation of Mrp4.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/physiopathology , DNA-Binding Proteins/genetics , Kidney/metabolism , Metabolic Detoxication, Phase I , Transcription Factors/genetics , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Bile Acids and Salts/blood , Carrier Proteins/metabolism , Cholestasis/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P450 Family 2 , DNA-Binding Proteins/metabolism , Liver/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL/genetics , Mice, Inbred C57BL/metabolism , Mice, Knockout , Multidrug Resistance-Associated Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear , Steroid Hydroxylases/metabolism , Transcription Factors/metabolism
16.
J Hepatol ; 44(1): 243-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16288819

ABSTRACT

We report two patients with uncommon Gilbert's syndrome with severe unconjugated hyperbilirubinemia which was reduced from 200 to 60-90 micromol/L by long-term administration of rifampicin. Hepatic induction of bilirubin-glucuronosyltransferase was suggested by increased relative amounts of conjugated serum bilirubin. This molecular mechanism was confirmed in primary cultures of human hepatocytes.


Subject(s)
Enzyme Inhibitors/therapeutic use , Gilbert Disease/drug therapy , Glucuronosyltransferase/antagonists & inhibitors , Rifampin/therapeutic use , Adult , Bilirubin/blood , Cells, Cultured , Female , Follow-Up Studies , Gilbert Disease/blood , Gilbert Disease/pathology , Glucuronosyltransferase/blood , Hepatocytes/pathology , Humans , Male , Time Factors
17.
Gastroenterology ; 129(2): 476-85, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16083704

ABSTRACT

BACKGROUND & AIMS: Rifampicin (RIFA) and ursodeoxycholic acid (UDCA) improve symptoms and biochemical markers of liver injury in cholestatic liver diseases by largely unknown mechanisms. We aimed to study the molecular mechanisms of action of these drugs in humans. METHODS: Thirty otherwise healthy gallstone patients scheduled for cholestectomy were randomized to RIFA (600 mg/day for 1 week) or UDCA (1 g/day for 3 weeks) or no medication before surgery. Routine biochemistry, lipids, and surrogate markers for P450 activity (4beta-hydroxy cholesterol, 4beta-OH-C) and bile acid synthesis (7alpha-hydroxy-4-cholesten-3-one, C-4) were measured in serum. Bile acids were analyzed in serum, urine, and bile. A wedge liver biopsy specimen was taken to study expression of hepatobiliary ABC transporters as well as detoxification enzymes and regulatory transcription factors. RESULTS: RIFA enhanced bile acid detoxification as well as bilirubin conjugation and excretion as reflected by enhanced expression of CYP3A4, UGT1A1, and MRP2. These molecular effects were paralleled by decreased bilirubin and deoxycholic acid concentrations in serum and decreased lithocholic and deoxycholic acid concentrations in bile. UDCA on the other hand stimulated the expression of BSEP, MDR3, and MRP4. UDCA became the predominant bile acid after UDCA treatment and lowered the biliary cholesterol saturation index. CONCLUSIONS: RIFA enhances bile acid detoxification as well as bilirubin conjugation and export systems, whereas UDCA stimulates the expression of transporters for canalicular and basolateral bile acid export as well as the canalicular phospholipid flippase. These independent but complementary effects may justify a combination of both agents for the treatment of cholestatic liver diseases.


Subject(s)
Biological Transport/drug effects , Cholelithiasis/surgery , Liver Circulation/drug effects , Rifampin/therapeutic use , Ursodeoxycholic Acid/therapeutic use , Biological Transport/physiology , Cholelithiasis/diagnosis , Cholestasis, Intrahepatic/diagnosis , Cholestasis, Intrahepatic/surgery , Dose-Response Relationship, Drug , Drug Administration Schedule , Elective Surgical Procedures , Female , Follow-Up Studies , Humans , Inactivation, Metabolic , Male , Middle Aged , Preoperative Care/methods , Reference Values , Rifampin/pharmacokinetics , Risk Assessment , Severity of Illness Index , Treatment Outcome , Ursodeoxycholic Acid/pharmacokinetics
18.
Am J Physiol Gastrointest Liver Physiol ; 289(5): G798-805, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16002565

ABSTRACT

Expression of the main hepatic bile acid uptake system, the Na+-taurocholate cotransporter (Ntcp), is downregulated during cholestasis. Bile acid-induced, farnesoid X receptor (FXR)-mediated induction of the nuclear repressor short heterodimer partner (SHP) has been proposed as a key mechanism reducing Ntcp expression. However, the role of FXR and SHP or other nuclear receptors and hepatocyte-enriched transcription factors in mediating Ntcp repression in obstructive cholestasis is unclear. FXR knockout (FXR-/-) and wild-type (FXR+/+) mice were subjected to common bile duct ligation (CBDL). Cholic acid (CA)-fed and LPS-treated FXR-/- and FXR+/+ mice were studied for comparison. mRNA levels of Ntcp and SHP and nuclear protein levels of hepatocyte nuclear factor (HNF)-1alpha, HNF-3beta, HNF-4alpha, retinoid X receptor (RXR)-alpha, and retinoic acid receptor (RAR)-alpha and their DNA binding were assessed. Hepatic cytokine mRNA levels were also measured. CBDL and CA led to Ntcp repression in FXR+/+, but not FXR-/-, mice, whereas LPS reduced Ntcp expression in both genotypes. CBDL and LPS but not CA induced cytokine expression and reduced levels of HNF-1alpha, HNF-3beta, HNF-4alpha, RXRalpha, and RARalpha to similar extents in FXR+/+ and FXR-/-. DNA binding of these transactivators was unaffected by CA in FXR+/+ mice but was markedly reduced in FXR-/- mice. In conclusion, Ntcp repression by CBDL and CA is mediated by accumulating bile acids via FXR and does not depend on cytokines, whereas Ntcp repression by LPS is independent of FXR. Reduced levels of HNF-1alpha, RXRalpha, and RARalpha in CBDL FXR-/- mice and reduced DNA binding in CA-fed FXR-/- mice, despite unchanged Ntcp levels, indicate that these factors may have a minor role in regulation of mouse Ntcp during cholestasis.


Subject(s)
Cholestasis/genetics , Down-Regulation , Hepatocytes/metabolism , Liver/metabolism , Membrane Transport Proteins/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Chemokines/genetics , Cholestasis/metabolism , Cholic Acid/pharmacology , Cytokines/genetics , Down-Regulation/drug effects , Lipopolysaccharides/pharmacology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Symporters
SELECTION OF CITATIONS
SEARCH DETAIL
...