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










Publication year range
1.
Clin Liver Dis (Hoboken) ; 18(5): 237-242, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34840725
2.
Biol Methods Protoc ; 6(1): bpab002, 2021.
Article in English | MEDLINE | ID: mdl-33655078

ABSTRACT

Quantifying the ratio of alternatively spliced mRNA variants of genes with known alternative splicing variants is highly relevant for many applications. Herein, we describe the validation of a quantitative PCR design for the simplified quantification of known mRNA splice variants. The assay uses a single-common primer pair, dual probe design for the determination of splicing variants in a single well configuration. We used murine XBP-1 splicing variants, XBP-1S and XBP-1U, to validate and demonstrate the performance characteristics of this approach. Using synthetic XBP-1S and XBP-1U cDNA as well as cDNA synthesized from mouse beta-cell line MIN6, we established the performance parameters and dynamic range of the assay. Reliable quantification of both variants at varying concentration gradients was shown. No cross detection of XBP-1U by the XBP-1S probe was detected and only marginal XBP-1S cross detection by the XBP-1U probe was detected at high concentration gradients that are unlikely to be relevant. We demonstrated that the assay accurately detected changes of XBP-1 splice variants in mouse liver subjected to pharmacologically induced ER stress without the need for normalization to a reference gene.

3.
Obesity (Silver Spring) ; 29(4): 713-720, 2021 04.
Article in English | MEDLINE | ID: mdl-33594826

ABSTRACT

OBJECTIVE: This study investigates the therapeutic potential of a small molecule inhibitor of plasminogen activator inhibitor-1 (PAI-1), TM5441, in reversing diet-induced obesity in mice. METHODS: Wild-type C57BL/6J mice were fed a high-fat high-sugar (HFHS) diet for 8 weeks to induce obesity. After the first 8 weeks, TM5441 was added to the diet for an additional 8 weeks. In order to determine the efficacy of PAI-1 inhibition in conjunction with dietary modification, mice were fed an HFHS diet for 8 weeks to induce obesity and were then switched to a low-fat diet with or without TM5441 for an additional 2 to 8 weeks. RESULTS: Obese mice showed weight reduction and significant improvement in hepatic steatosis when TM5441 was added to the HFHS diet. Obese mice that were treated with TM5441 in conjunction with dietary modification showed enhanced weight loss and a more rapid reversal of hepatic steatosis compared with obese mice treated with dietary modification alone. The enhanced weight loss among mice treated with TM5441 was associated with increased adipose tissue expression of adipose triglyceride lipase, phosphorylated hormone-sensitive lipase, and phosphorylated perilipin-1 as well as induction of adipose tissue lipolysis. CONCLUSIONS: Pharmacologic PAI-1 inhibition stimulates adipose tissue lipolysis and enhances weight loss in obese mice.


Subject(s)
Lipolysis/physiology , Plasminogen Activator Inhibitor 1/therapeutic use , Weight Loss/drug effects , Animals , Male , Mice , Mice, Obese
4.
FASEB Bioadv ; 2(12): 695-704, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33336157

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1) is a stress-responsive gene that is highly induced in nonalcoholic steatohepatitis (NASH). Endoplasmic reticulum (ER) stress is a salient feature of NASH, yet it is unknown whether ER stress contributes to hepatic PAI-1 induction in this disorder. Therefore, we aimed to (a) establish the role of ER stress in the regulation of hepatic Pai-1 expression, and (b) determine whether induction of Pai-1 in murine NASH is driven by ER stress. Hepatic Pai-1 expression was measured in C57BL/6 J mice and human HepG2 cells subjected to acute or prolonged pharmacologic ER stress. We found that hepatic Pai-1 expression was acutely suppressed in murine liver in response to severe ER stress followed by marked induction during the recovery phase of the ER stress response. Hepatic Pai-1 expression was induced in response to prolonged low-grade ER stress in mice. Induction of PAI-1 by ER stress in HepG2 cells was prevented by pharmacologic inhibition of MEK1/ERK signaling or by siRNA-mediated knockdown of XBP1, mediators of the recovery response to ER stress. Inhibiting ER stress with 4-phenylbutyric acid prevented hepatic Pai-1 induction in mice with diet-induced steatohepatitis. We conclude that hepatic Pai-1 is induced by ER stress via a pathway involving XBP1 and MEK1/ERK signaling, and induction of hepatic Pai-1 in murine NASH is mediated by ER stress. These data implicate ER stress as a novel mechanistic link between Pai-1 induction and NASH.

5.
J Lipid Res ; 60(2): 353-359, 2019 02.
Article in English | MEDLINE | ID: mdl-30482806

ABSTRACT

Refeeding mice after a prolonged fast is a potent stimulus of hepatic lipogenesis, but is also associated with induction of the hepatic unfolded protein response (UPR). The X-box binding protein 1 (Xbp1), a key regulator of the adaptive UPR, transcriptionally activates hepatic lipogenesis genes. We therefore determined whether hepatic Xbp1 mediates the hepatic lipogenic response to refeeding. Mice bearing a hepatocyte-specific deletion of Xbp1 and littermate controls were fasted overnight, followed by refeeding for up to 6 h. Among control mice, refeeding induced hepatic expression of activated Xbp1 and, as expected, induced hepatic expression of genes controlling de novo lipogenesis of fatty acids. Unexpectedly, deletion of hepatic Xbp1 allowed for normal induction of hepatic lipogenesis genes, yet impaired translation of SREBP1c and its targets in response to refeeding. Impaired protein translation was associated with enhanced postprandial activation of the global translational arrest protein, eukaryotic initiation factor 2α, among mice lacking hepatic Xbp1 Deletion of hepatic Xbp1 prevented postprandial induction of genes regulating protein folding and processing and shifted the pattern of postprandial UPR activation to favor proapoptotic signals. We conclude that activation of hepatic Xbp1 in the postprandial states serves the dual roles of restoring postprandial hepatic lipogenesis and proteostasis.


Subject(s)
Feeding Behavior , Liver/metabolism , X-Box Binding Protein 1/metabolism , Animals , Gene Deletion , Lipogenesis/genetics , Mice , Proteostasis/genetics , Transcriptional Activation , X-Box Binding Protein 1/deficiency , X-Box Binding Protein 1/genetics
6.
Hepatol Commun ; 2(12): 1479-1492, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30556037

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1), an essential regulator of fibrinolysis, is increasingly implicated in the pathogenesis of metabolic disorders, such as obesity and nonalcoholic fatty liver disease (NAFLD). Pharmacologic inhibition of PAI-1 is emerging as a highly promising therapeutic strategy for obesity and its sequelae. Given the well-established profibrotic function of PAI-1, we considered whether PAI-1 may serve as a target for antifibrotic therapy in nonalcoholic steatohepatitis (NASH). We therefore determined the effect of genetic Pai-1 deletion and pharmacologic PAI-1 inhibition on the development of NASH-related fibrosis in mice. Pai-1 knockout (Pai-1 -/-) and wild-type control (Pai-1 +/+) mice were fed a high-fat/high-cholesterol high-sugar (HFHS) diet or a methionine- and choline-deficient (MCD) diet to induce steatohepatitis with fibrosis. PAI-1 was pharmacologically inhibited using the small molecule inhibitor TM5441 in wild-type C57BL/6 mice fed an HFHS or MCD diet. Either genetic deletion of Pai-1 or pharmacologic inhibition of PAI-1 attenuated MCD diet-induced hepatic steatosis but did not prevent hepatic inflammation or fibrosis. Targeted inhibition of PAI-1 conferred transient protection from HFHS diet-induced obesity and hepatic steatosis, an effect that was lost with prolonged exposure to the obesigenic diet. Neither genetic deletion of Pai-1 nor pharmacologic inhibition of PAI-1 prevented HFHS diet-induced hepatic inflammation or fibrosis. Conclusion: Pai-1 regulates hepatic lipid accumulation but does not promote NASH progression. The PAI-1 inhibitor TM5441 effectively attenuates diet-induced obesity and hepatic steatosis but does not prevent NASH-related fibrosis in mice.

7.
Semin Liver Dis ; 38(4): 320-332, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30357769

ABSTRACT

Activation of the hepatic unfolded protein response (UPR), a highly conserved cellular response to endoplasmic reticulum (ER) stress, is a firmly established feature of nonalcoholic fatty liver disease (NAFLD). ER stress is now widely accepted as both a cause and a consequence of hepatic steatosis. Moreover, the accumulation of hepatic lipids induces ER stress, which, in turn, disrupts hepatic lipid metabolism thus creating a vicious cycle that potentiates hepatic lipid accumulation. Additionally, there is interplay between the UPR and the inflammatory cascades associated with progressive nonalcoholic steatohepatitis. Understanding the molecular mechanisms by which the UPR regulates hepatic lipid metabolism and lipotoxic liver injury may lead to the identification of novel therapeutic targets for the treatment of NAFLD.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Lipid Metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Unfolded Protein Response/physiology , Animals , Humans , Lipogenesis , Mice
9.
Mol Metab ; 6(12): 1616-1624, 2017 12.
Article in English | MEDLINE | ID: mdl-29157602

ABSTRACT

OBJECTIVE: Fibroblast growth factor 21 (FGF21), a key regulator of the metabolic response to fasting, is highly induced by endoplasmic reticulum (ER) stress. The X-box binding protein 1 (Xbp1) is one of several ER stress proteins that has been shown to directly activate the FGF21 promoter. We aimed to determine whether hepatic Xbp1 is required for induction of hepatic FGF21 in vivo. METHODS: Mice bearing a hepatocyte-specific deletion of Xbp1 (Xbp1LKO) were subjected to fasting, pharmacologic ER stress, or a ketogenic diet, all potent stimuli of Fgf21 expression. RESULTS: Hepatocyte-specific Xbp1 knockout mice demonstrated normal induction of FGF21 in response to fasting or pharmacologic ER stress and enhanced induction of FGF21 in response to a ketogenic diet. Consistent with preserved induction of FGF21, Xbp1LKO mice exhibited normal induction of FGF21 target genes and normal ketogenesis in response to fasting or a ketogenic diet. CONCLUSION: Hepatic Xbp1 is not required for induction of FGF21 under physiologic or pathophysiologic conditions in vivo.


Subject(s)
Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Diet, Ketogenic , Endoplasmic Reticulum Stress , Fasting/metabolism , Fibroblast Growth Factors/genetics , Humans , Mice , Mice, Inbred C57BL , X-Box Binding Protein 1/genetics
10.
Cell Mol Gastroenterol Hepatol ; 3(2): 261-271, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28275692

ABSTRACT

BACKGROUND & AIMS: Cholestasis promotes endoplasmic reticulum (ER) stress in the liver, however, the effect of ER stress on hepatic bile acid metabolism is unknown. We aim to determine the effect of ER stress on hepatic bile acid synthesis and transport in mice. METHODS: ER stress was induced pharmacologically in C57BL/6J mice and human hepatoma (HepG2) cells. The hepatic expression of genes controlling bile acid synthesis and transport was determined. To measure the activity of the primary bile acid synthetic pathway, the concentration of 7α-hydroxy-4-cholesten-3-1 was measured in plasma. RESULTS: Induction of ER stress in mice and HepG2 cells rapidly suppressed the hepatic expression of the primary bile acid synthetic enzyme, cholesterol 7α-hydroxylase. Plasma levels of 7α-hydroxy-4-cholesten-3-1 were reduced in mice subjected to ER stress, indicating impaired bile acid synthesis. Induction of ER stress in mice and HepG2 cells increased expression of the bile salt export pump (adenosine triphosphate binding cassette [Abc]b11) and a bile salt efflux pump (Abcc3). The observed regulation of Cyp7a1, Abcb11, and Abcc3 occurred in the absence of hepatic inflammatory cytokine activation and was not dependent on activation of hepatic small heterodimer partner or intestinal fibroblast growth factor 15. Consistent with suppressed bile acid synthesis and enhanced bile acid export from hepatocytes, prolonged ER stress decreased the hepatic bile acid content in mice. CONCLUSIONS: Induction of ER stress in mice suppresses bile acid synthesis and enhances bile acid removal from hepatocytes independently of established bile acid regulatory pathways. These data show a novel function of the ER stress response in regulating bile acid metabolism.

11.
J Lipid Res ; 58(3): 504-511, 2017 03.
Article in English | MEDLINE | ID: mdl-28039331

ABSTRACT

The unfolded protein response (UPR) is an adaptive response to endoplasmic reticulum stress and the inositol-requiring enzyme 1α/X-box binding protein 1 (IRE1α/XBP1) pathway of the UPR is important in lipid metabolism. However, its role in bile acid metabolism remains unknown. We demonstrate that liver-specific Xbp1 knockout (LS-Xbp1-/-) mice had a 45% reduction in total bile acid pool. LS-Xbp1-/- mice had lower serum 7α-hydroxy-4-cholesten-3-one (C4) levels compared with Xbp1fl/fl mice, indicating reduced cholesterol 7α-hydroxylase (CYP7A1) synthetic activity. This occurred without reductions of hepatic CYP7A1 protein expression. Feeding LS-Xbp1-/- mice cholestyramine increased hepatic CYP7A1 protein expression to levels 2-fold and 8-fold greater than cholestyramine-fed and chow-fed Xbp1fl/fl mice, respectively. However, serum C4 levels remained unchanged and were lower than both groups of Xbp1fl/fl mice. In contrast, although feeding LS-Xbp1-/- mice cholesterol did not increase CYP7A1 expression, serum C4 levels increased significantly up to levels similar to chow-fed Xbp1fl/fl mice and the total bile acid pool normalized. In conclusion, loss of hepatic XBP1 decreased the bile acid pool and CYP7A1 synthetic activity. Cholesterol feeding, but not induction of CYP7A1 with cholestyramine, increased CYP7A1 synthetic activity and corrected the genotype-specific total bile acid pools. These data demonstrate a novel role of IRE1α/XBP1 regulating bile acid metabolism.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/genetics , Lipid Metabolism/genetics , Liver/metabolism , X-Box Binding Protein 1/genetics , Animals , Bile Acids and Salts/metabolism , Cholestenones/blood , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Unfolded Protein Response/genetics
12.
J Biol Chem ; 290(50): 30142-51, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26504083

ABSTRACT

Endoplasmic reticulum (ER) stress activates the unfolded protein response (UPR), a highly conserved signaling cascade that functions to alleviate stress and promote cell survival. If, however, the cell is unable to adapt and restore homeostasis, then the UPR activates pathways that promote apoptotic cell death. The molecular mechanisms governing the critical transition from adaptation and survival to initiation of apoptosis remain poorly understood. We aim to determine the role of hepatic Xbp1, a key mediator of the UPR, in controlling the adaptive response to ER stress in the liver. Liver-specific Xbp1 knockout mice (Xbp1(LKO)) and Xbp1(fl/fl) control mice were subjected to varying levels and durations of pharmacologic ER stress. Xbp1(LKO) and Xbp1(fl/fl) mice showed robust and equal activation of the UPR acutely after induction of ER stress. By 24 h, Xbp1(fl/fl) controls showed complete resolution of UPR activation and no liver injury, indicating successful adaptation to the stress. Conversely, Xbp1(LKO) mice showed ongoing UPR activation associated with progressive liver injury, apoptosis, and, ultimately, fibrosis by day 7 after induction of ER stress. These data indicate that hepatic XBP1 controls the adaptive response of the UPR and is critical to restoring homeostasis in the liver in response to ER stress.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Gene Deletion , Liver/metabolism , Oxidative Stress , Transcription Factors/genetics , Animals , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
13.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G965-74, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26472223

ABSTRACT

Fatty liver is associated with endoplasmic reticulum stress and activation of the hepatic unfolded protein response (UPR). Reduced hepatic expression of the UPR regulator X-box binding protein 1 spliced (XBP1s) is associated with human nonalcoholic steatohepatitis (NASH), and feeding mice a high-fat diet with fructose/sucrose causes progressive, fibrosing steatohepatitis. This study examines the role of XBP1 in nonalcoholic fatty liver injury and fatty acid-induced cell injury. Hepatocyte-specific Xbp1-deficient (Xbp1(-/-)) mice were fed a high-fat/sugar (HFS) diet for up to 16 wk. HFS-fed Xbp1(-/-) mice exhibited higher serum alanine aminotransferase levels compared with Xbp1(fl/fl) controls. RNA sequencing and Gene Ontogeny pathway analysis of hepatic mRNA revealed that apoptotic process, inflammatory response, and extracellular matrix structural constituent pathways had enhanced activation in HFS-fed Xbp1(-/-) mice. Liver histology demonstrated enhanced injury and fibrosis but less steatosis in the HFS-fed Xbp1(-/-) mice. Hepatic Col1a1 and Tgfß1 gene expression, as well as Chop and phosphorylated JNK (p-JNK), were increased in Xbp1(-/-) compared with Xbp1(fl/fl) mice after HFS feeding. In vitro, stable XBP1-knockdown Huh7 cells (Huh7-KD) and scramble control cells (Huh7-SCR) were generated and treated with palmitic acid (PA) for 24 h. PA-treated Huh7-KD cells had increased cytotoxicity measured by lactate dehydrogenase release, apoptotic nuclei, and caspase3/7 activity assays compared with Huh7-SCR cells. CHOP and p-JNK expression was also increased in Huh7-KD cells following PA treatment. In conclusion, loss of XBP1 enhances injury in both in vivo and in vitro models of fatty liver injury. We speculate that hepatic XBP1 plays an important protective role in pathogenesis of NASH.


Subject(s)
DNA-Binding Proteins/deficiency , Diet, High-Fat , Dietary Sucrose , Hepatocytes/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Transcription Factors/deficiency , Alanine Transaminase/blood , Animals , Apoptosis , Cell Line, Tumor , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Palmitic Acid/toxicity , Phosphorylation , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Time Factors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , X-Box Binding Protein 1
14.
PLoS One ; 9(7): e103828, 2014.
Article in English | MEDLINE | ID: mdl-25077945

ABSTRACT

BACKGROUND AND AIMS: Endoplasmic reticulum (ER) stress is induced in many forms of chronic liver disease and may promote the development of hepatocellular carcinoma. The activator protein 1 (AP-1) complex is a transcription factor that promotes hepatic carcinogenesis in response to cellular stress. The aim of this study was to determine the role of ER stress in the regulation of the hepatic AP-1 complex. METHODS: Human hepatocellular carcinoma (HepG2) cells and C57BL/6J mice were subjected to pharmacologic ER stress and the expression of AP-1-associated genes and proteins was assessed. To determine the role of MAPK signaling in ER stress-induced AP-1 activation, ER stress was induced in JNK- and ERK-inhibited HepG2 cells. RESULTS: Induction of ER stress promoted the activation of both Jun- and Fos-related genes and proteins of the AP-1 complex in HepG2 cells and murine liver. Inhibition of ERK phosphorylation in HepG2 cells completely prevented ER stress-induced activation of the fos-related components of AP-1 whereas activation of Jun-related components was only partially attenuated. Conversely, inhibition of JNK phosphorylation in HepG2 cells reduced ER stress-induced activation of Jun-related components but did not prevent activation of fos-related components. CONCLUSIONS: ER stress activates the hepatic AP-1 complex via MAPK-dependent signaling pathways. ER stress-induced activation of Fos-related components is dependent primarily on ERK activation whereas ER stress-induced activation of Jun-related components is dependent primarily on JNK activation, although there is interplay between these regulatory pathways. These data implicate a novel signaling pathway by which sustained ER stress, as observed in many chronic liver diseases, may promote hepatic carcinogenesis.


Subject(s)
Endoplasmic Reticulum Stress , MAP Kinase Signaling System , Transcription Factor AP-1/metabolism , Animals , Cell Proliferation , Hep G2 Cells , Humans , Liver/metabolism , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcriptional Activation
15.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G221-6, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23139217

ABSTRACT

The bile salt export pump, encoded by ABCB11, is the predominant canalicular transport protein for biliary bile acid secretion. The level of ABCB11 expression in humans is widely variable yet the impact of this variability on human disease is not well defined. We aim to determine the effect of hepatic Abcb11 overexpression on the enterohepatic circulation (EHC) in mice. We used a stable isotope dilution technique in transgenic mice overexpressing hepatic Abcb11 (TTR-Abcb11) to determine the pool size, fractional turnover rate (FTR), and synthesis rate of the primary bile acid, cholic acid (CA). The gallbladder was cannulated to determine bile flow, bile acid composition, and the biliary secretion rates of CA, total bile acids, phospholipid, and cholesterol. The combined data allowed for estimation of the CA cycling time and the fraction of CA lost per cycle. Hepatic and intestinal gene and protein expression were determined by qPCR and Western blot. Abcb11 overexpression strongly decreased FTR and synthesis rate of CA. Abcb11 overexpression decreased the fraction of CA that was lost per cycle of the EHC. Hepatic expression of Cyp7a1 was suppressed by nearly 50% and ileal expression of FGF15 was increased more than eightfold in TTR-Abcb11 mice. Despite the increased intestinal reabsorption of bile acids, ileal Asbt expression was suppressed. Hepatic Abcb11 overexpression in mice increases the conservation of bile acids within the enterohepatic circulation. These data provide strong evidence for the existence of feed-forward communication between hepatic expression of a bile acid transport protein and the intestine.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bile Acids and Salts/metabolism , Bile/metabolism , Enterohepatic Circulation , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Animals , Blotting, Western , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholic Acid/metabolism , Feedback, Physiological , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Ileum/metabolism , Indicator Dilution Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organic Anion Transporters, Sodium-Dependent/metabolism , Phospholipids/metabolism , Prealbumin/genetics , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Symporters/metabolism , Up-Regulation
16.
Article in English | MEDLINE | ID: mdl-22556147

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of nonalcoholic steatohepatitis. The ER stress response is activated in the livers of mice fed a methionine- and choline-deficient (MCD) diet, yet the role of ER stress in the pathogenesis of MCD diet-induced steatohepatitis is unknown. Using chemical chaperones on hepatic steatosis and markers of inflammation and fibrosis in mice fed a MCD diet, we aim to determine the effects of reducing ER stress. C57BL/6J mice were fed a MCD diet with or without the ER chemical chaperones 4-phenylbutyric acid (PBA) and tauroursodeoxycholic acid (TUDCA) for 2 wk. TUDCA and PBA effectively attenuated the ER stress response in MCD diet-fed mice, as evidenced by reduced protein levels of phosphorylated eukaryotic initiation factor 2α and phosphorylated JNK and suppression of mRNA levels of CCAAT/enhancer binding protein homologous protein, glucose-regulated protein 78 kDa, and X-box binding protein 1. However, PBA and TUDCA did not decrease MCD diet-induced hepatic steatosis. MCD diet-induced hepatic inflammation, as evidenced by increased plasma alanine aminotransferase and induction of hepatic TNFα expression, was also not reduced by PBA or TUDCA. PBA and TUDCA did not attenuate MCD diet-induced upregulation of the fibrosis-associated genes tissue inhibitor of metalloproteinase-1 and matrix metalloproteinase-9. ER chemical chaperones reduce MCD diet-induced ER stress, yet they do not improve MCD diet-induced hepatic steatosis, inflammation, or activation of genes associated with fibrosis. These data suggest that although the ER stress response is activated by the MCD diet, it does not have a primary role in the pathogenesis of MCD diet-induced steatohepatitis.


Subject(s)
Choline Deficiency/physiopathology , Diet , Endoplasmic Reticulum/physiology , Fatty Liver/physiopathology , Methionine/deficiency , Stress, Physiological/physiology , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/physiology , Cholesterol/metabolism , Gene Expression/genetics , Gene Expression/physiology , Inflammation/pathology , Liver/chemistry , Liver/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones/therapeutic use , Phenylbutyrates/therapeutic use , Real-Time Polymerase Chain Reaction , Taurochenodeoxycholic Acid/therapeutic use
17.
Gastroenterology ; 141(4): 1404-11, 1411.e1-2, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21726512

ABSTRACT

BACKGROUND & AIMS: ABCB11 is a canalicular transport protein that controls the rate-limiting step in hepatic bile acid secretion. Its expression levels vary in humans, and it is not clear how these variations affect lipid metabolism. We investigated whether overexpression of Abcb11 in mice increases lipid absorption in the intestine and affects the development of obesity or hypercholesterolemia. METHODS: Transgenic mice that overexpress Abcb11 in liver (TTR-Abcb11) and FVB/NJ mice (controls) were fed a high-cholesterol or high-fat diet for 12 weeks. Intestinal lipid absorption was measured by the dual fecal isotope method. Energy expenditure was measured by indirect calorimetry. The bile acid pool was analyzed by high-performance liquid chromatography. RESULTS: TTR-Abcb11 mice had a nearly 2-fold increase in intestinal cholesterol absorption compared with controls. TTR-Abcb11 mice fed a high-cholesterol diet had greater increases in plasma and hepatic levels of cholesterol and became more obese than controls; they also had increased intestinal absorption of fatty acids and decreased energy expenditure. In the TTR-Abcb11 mice, the sizes of plasma and total bile acid pools were reduced; the bile acid pool contained more species of hydrophobic bile acids compared with controls. CONCLUSIONS: Hepatic overexpression of Abcb11 in mice promotes diet-induced obesity and hypercholesterolemia; increased intestinal cholesterol absorption by hydrophobic bile acids might cause these features. Increased absorption of fatty acids in the intestine and reduced expenditure of energy could increase weight gain in TTR-Abcb11 mice. In humans, variations in expression of ABCB11 might confer genetic susceptibility to diet-induced hyperlipidemia and obesity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Hypercholesterolemia/metabolism , Liver/metabolism , Obesity/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Animals , Bile Acids and Salts/metabolism , Calorimetry, Indirect , Cholesterol, Dietary/metabolism , Chromatography, High Pressure Liquid , Energy Metabolism , Fatty Acids/metabolism , Genotype , Hydrophobic and Hydrophilic Interactions , Hypercholesterolemia/genetics , Intestinal Absorption , Intestinal Mucosa/metabolism , Male , Mice , Mice, Transgenic , Obesity/genetics , Phenotype , Prealbumin/genetics , Time Factors , Up-Regulation , Weight Gain
18.
J Lipid Res ; 52(2): 289-98, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21097822

ABSTRACT

Cholesterol 7α-hydroxylase (CYP7A1) encodes for the rate-limiting step in the conversion of cholesterol to bile acids in the liver. In response to acute cholesterol feeding, mice upregulate CYP7A1 via stimulation of the liver X receptor (LXR) α. However, the effect of a chronic high-cholesterol diet on hepatic CYP7A1 expression in mice is unknown. We demonstrate that chronic cholesterol feeding (0.2% or 1.25% w/w cholesterol for 12 weeks) in FVB/NJ mice results in a >60% suppression of hepatic CYP7A1 expression associated with a >2-fold increase in hepatic cholesterol content. In contrast, acute cholesterol feeding induces a >3-fold upregulation of hepatic CYP7A1 expression. We show that chronic, but not acute, cholesterol feeding increases the expression of hepatic inflammatory cytokines, tumor necrosis factor (TNF)α, and interleukin (IL)-1ß, which are known to suppress hepatic CYP7A1 expression. Chronic cholesterol feeding also results in activation of the mitogen activated protein (MAP) kinases, c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Furthermore, we demonstrate in vitro that suppression of CYP7A1 by TNFα and IL-1ß is dependent on JNK and ERK signaling. We conclude that chronic high-cholesterol feeding suppresses CYP7A1 expression in mice. We propose that chronic cholesterol feeding induces inflammatory cytokine activation and liver damage, which leads to suppression of CYP7A1 via activation of JNK and ERK signaling pathways.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cholesterol, Dietary/administration & dosage , Liver/metabolism , Animals , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/biosynthesis , Hep G2 Cells , Humans , Interleukin-1beta/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
J Biol Chem ; 284(46): 31807-16, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19762918

ABSTRACT

Hyperhomocysteinemia has been correlated with hepatic steatosis and activation of the unfolded protein response (UPR), yet a causal relationship has not been established. Although methionine and choline are essential components of homocysteine metabolism, the role of homocysteine in the pathogenesis of a methionine- and choline-deficient (MCD) diet remains unknown. We explored the effects of homocysteine supplementation on hepatic steatosis and the UPR in mice fed a control or MCD diet. Mice fed the MCD diet developed severe hyperhomocysteinemia and activation of the hepatic UPR. Supplementing the MCD diet with homocysteine attenuated the MCD diet-induced hepatic UPR activation and other injurious effects of the MCD diet including hepatic cholesterol accumulation, weight loss, and plasma ALT elevation. Homocysteine supplementation replenished the MCD diet-induced depletion of hepatic S-adenosylmethionine (SAM). Depleting SAM in HepG2 cells using MAT1alpha siRNA or cycloleucine resulted in enhanced activation of the UPR upon exposure to thapsigargin. Mice fed a control diet supplemented with homocysteine had a 3-fold elevation in plasma homocysteine level by 2 weeks and 6-fold elevation by 6 weeks but demonstrated no other pathophysiologic change. In summary, we found that homocysteine attenuates MCD diet-induced hepatic UPR activation, likely via repletion of hepatic SAM. Furthermore, homocysteine supplementation alone does not cause hepatic steatosis or UPR activation despite inducing hyperhomocysteinemia. These studies indicate that although hyperhomocysteinemia is often associated with hepatic steatosis and UPR activation, these effects may be a secondary response rather than a direct effect of homocysteine.


Subject(s)
Dietary Supplements , Disease Models, Animal , Fatty Liver/pathology , Homocysteine/administration & dosage , Unfolded Protein Response/drug effects , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Choline Deficiency , Fatty Liver/etiology , Fatty Liver/metabolism , Female , Homocysteine/pharmacology , Liver/drug effects , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Methionine/deficiency , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylmethionine/metabolism
20.
Article in English | MEDLINE | ID: mdl-16582962

ABSTRACT

Malnutrition is highly prevalent among patients with chronic liver disease and is nearly universal among patients awaiting liver transplantation. Malnutrition in patients with cirrhosis leads to increased morbidity and mortality rates. Furthermore, patients who are severely malnourished before transplant surgery have a higher rate of complications and a decreased overall survival rate after liver transplantation. In light of the high incidence of malnutrition among patients with chronic liver disease and the complications that result from malnutrition in these patients, it is essential to assess the nutritional status of all patients with liver disease, and to initiate treatment as indicated. This review addresses the etiologies of malnutrition, methods used to assess nutritional status, and appropriate treatment strategies.


Subject(s)
Liver Diseases/epidemiology , Liver Diseases/therapy , Malnutrition/epidemiology , Nutritional Support , Chronic Disease , Energy Intake , Enteral Nutrition , Humans , Liver Diseases/surgery , Liver Transplantation , Nutrition Assessment , Nutritional Status , Parenteral Nutrition , Protein-Energy Malnutrition/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...