Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 8(357): 357ra122, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27655848

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world, and safe and effective therapies are needed. Bile acids (BAs) and their receptors [including the nuclear receptor for BAs, farnesoid X receptor (FXR)] play integral roles in regulating whole-body metabolism and hepatic lipid homeostasis. We hypothesized that interruption of the enterohepatic BA circulation using a luminally restricted apical sodium-dependent BA transporter (ASBT) inhibitor (ASBTi; SC-435) would modify signaling in the gut-liver axis and reduce steatohepatitis in high-fat diet (HFD)-fed mice. Administration of this ASBTi increased fecal BA excretion and messenger RNA (mRNA) expression of BA synthesis genes in liver and reduced mRNA expression of ileal BA-responsive genes, including the negative feedback regulator of BA synthesis, fibroblast growth factor 15. ASBT inhibition resulted in a marked shift in hepatic BA composition, with a reduction in hydrophilic, FXR antagonistic species and an increase in FXR agonistic BAs. ASBT inhibition restored glucose tolerance, reduced hepatic triglyceride and total cholesterol concentrations, and improved NAFLD activity score in HFD-fed mice. These changes were associated with reduced hepatic expression of lipid synthesis genes (including liver X receptor target genes) and normalized expression of the central lipogenic transcription factor, Srebp1c Accumulation of hepatic lipids and SREBP1 protein were markedly reduced in HFD-fed Asbt(-/-) mice, providing genetic evidence for a protective role mediated by interruption of the enterohepatic BA circulation. Together, these studies suggest that blocking ASBT function with a luminally restricted inhibitor can improve both hepatic and whole body aspects of NAFLD.


Subject(s)
Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Ileum/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Ceramides/metabolism , Cholesterol/metabolism , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/pharmacology , Feces , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Ileum/drug effects , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Organic Anion Transporters, Sodium-Dependent/deficiency , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Symporters/deficiency , Symporters/metabolism , Triglycerides/metabolism , Tropanes/administration & dosage , Tropanes/pharmacology
2.
J Nutr Biochem ; 26(3): 285-92, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25577467

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome (MetS). Up to a third of NAFLD subjects are at risk for developing nonalcoholic steatohepatitis (NASH). Many rodent models fail to replicate both MetS and NASH. The purpose of this study was to develop a reliable mouse model of NASH and MetS using a diet containing cholesterol, saturated fat and carbohydrate that is reflective of Western diets of North Americans. EXPERIMENTAL DESIGN: We used adult male C57BL/6 J 4- to 5-week-old mice and administered a solid diet containing 0.2% cholesterol, 45% of its calories from fat, with 30% of the fat in the form of partially hydrogenated vegetable oil. We also provided carbohydrate largely as high-fructose corn syrup equivalent in water. In a separate cohort, we gave the identical diet in the absence of cholesterol. Glucose and insulin tolerance testing was conducted throughout the feeding period. The feeding was conducted for 16 weeks, and the mice were sacrificed for histological analysis, markers of MetS, liver inflammation, circulating lipids, as well as liver staining for fibrosis and alpha smooth muscle actin (α-SMA). RESULTS: We found that cholesterol significantly increased serum leptin, interleukin-6, liver weight and liver weight/body weight ratio, fibrosis and liver α-SMA. CONCLUSIONS: Mice administered a diet accurately reflecting patterns associated with humans afflicted with MetS can reliably replicate features of MetS, NASH and significant liver fibrosis. The model we describe significantly reduces the time by several months for development of stage 3 hepatic fibrosis.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Liver/physiopathology , Metabolic Syndrome/etiology , Non-alcoholic Fatty Liver Disease/etiology , Actins/metabolism , Adipokines/blood , Adipokines/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Glucose Intolerance/etiology , High Fructose Corn Syrup/adverse effects , Hydrogenation , Insulin Resistance , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice, Inbred C57BL , Organ Size , Time Factors
3.
FASEB J ; 28(12): 5172-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25154876

ABSTRACT

Previous evidence indicates that adiponectin possesses antifibrogenic activity in inhibiting liver fibrosis. Therapeutic strategies, however, are limited by adiponectin quaternary structure and effective concentrations in circulation. Here we postulate a novel molecular mechanism, whereby adiponectin targets focal adhesion kinase (FAK) activity and disrupts key features of the fibrogenic response. Adiponectin-null (Ad(-/-)) mice and wild-type littermates were exposed to either saline or carbon tetrachloride (CCl4) for 6 wk. CCl4-gavaged mice were also injected with attenuated adenoviral adiponectin (Ad-Adn) or Ad-LacZ for 2 wk. Hepatic stellate cells (HSCs) were treated with or without adiponectin to elucidate signal transduction mechanisms. In vivo delivery of Ad-Adn markedly attenuates CCl4-induced expression of key integrin proteins and markers of HSC activation: αv, ß3, ß1, α2(I) collagen, and α-smooth muscle actin. Confocal experiments of liver tissues demonstrated that adiponectin delivery also suppressed vinculin and p-FAK activity in activated HSCs. In vitro, adiponectin induced dephosphorylation of FAK, mediated by a physical association with activated tyrosine phosphatase, Shp2. Conversely, Shp2 knockdown by siRNA significantly attenuated adiponectin-induced FAK deactivation, and expression of TIMP1 and α2(I) collagen was abolished in the presence of adiponectin and si-FAK. Finally, we documented that either adiponectin or the synthetic peptide with adiponectin properties, ADP355, suppressed p-FAK in synthetic matrices with stiffness measurements of 9 and 15 kPa, assessed by immunofluorescent imaging and quantitation. The in vivo and in vitro data presented indicate that disassembly of focal adhesion complexes in HSCs is pivotal for hepatic fibrosis therapy, now that small adiponectin-like peptides are available.


Subject(s)
Adiponectin/physiology , Focal Adhesions , Hepatic Stellate Cells/cytology , Liver Cirrhosis/therapy , Animals , Base Sequence , DNA Primers , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Gastrointest Liver Physiol ; 302(2): G225-35, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22038829

ABSTRACT

The aims of this study were designed to determine whether liraglutide, a long-acting glucagon-like peptide, could reverse the adverse effects of a diet high in fat that also contained trans-fat and high-fructose corn syrup (ALIOS diet). Specifically, we examined whether treatment with liraglutide could reduce hepatic insulin resistance and steatosis as well as improve cardiac function. Male C57BL/6J mice were pair fed or fed ad libitum either standard chow or the ALIOS diet. After 8 wk the mice were further subdivided and received daily injections of either liraglutide or saline for 4 wk. Hyperinsulinemic-euglycemic clamp studies were performed after 6 wk, revealing hepatic insulin resistance. Glucose tolerance and insulin resistance tests were performed at 8 and 12 wk prior to and following liraglutide treatment. Liver pathology, cardiac measurements, blood chemistry, and RNA and protein analyses were performed. Clamp studies revealed hepatic insulin resistance after 6 wk of ALIOS diet. Liraglutide reduced visceral adiposity and liver weight (P < 0.001). As expected, liraglutide improved glucose and insulin tolerance. Liraglutide improved hypertension (P < 0.05) and reduced cardiac hypertrophy. Surprisingly, liver from liraglutide-treated mice had significantly higher levels of fatty acid binding protein, acyl-CoA oxidase II, very long-chain acyl-CoA dehydrogenase, and microsomal triglyceride transfer protein. We conclude that liraglutide reduces the harmful effects of an ALIOS diet by improving insulin sensitivity and by reducing lipid accumulation in liver through multiple mechanisms including, transport, and increase ß-oxidation.


Subject(s)
Cardiomegaly/drug therapy , Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Glucagon-Like Peptide 1/analogs & derivatives , Adiposity/drug effects , Animals , Cardiomegaly/etiology , Fatty Liver/etiology , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Glucose Tolerance Test , Hypertension/drug therapy , Hypertension/etiology , Insulin Resistance , Liraglutide , Liver/drug effects , Male , Mice , Mice, Inbred C57BL
5.
PLoS One ; 6(9): e25269, 2011.
Article in English | MEDLINE | ID: mdl-21957486

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a known outcome of hepatosteatosis. Free fatty acids (FFA) induce the unfolded protein response (UPR) or endoplasmic reticulum (ER) stress that may induce apoptosis. Recent data indicate ER stress to be a major player in the progression of fatty liver to more aggressive lesions. Autophagy on the other hand has been demonstrated to be protective against ER stress-induced cell death. We hypothesized that exendin-4 (GLP-1 analog) treatment of fat loaded hepatocytes can reduce steatosis by autophagy which leads to reduced ER stress-related hepatocyte apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: Primary human hepatocytes were loaded with saturated, cis- and trans-unsaturated fatty acids (palmitic, oleic and elaidic acid respectively). Steatosis, induced with all three fatty acids, was significantly resolved after exendin-4 treatment. Exendin-4 sustained levels of GRP78 expression in fat-loaded cells when compared to untreated fat-loaded cells alone. In contrast, CHOP (C/EBP homologous protein); the penultimate protein that leads to ER stress-related cell death was significantly decreased by exendin-4 in hepatocytes loaded with fatty acids. Finally, exendin-4 in fat loaded hepatocytes clearly promoted gene products associated with macroautophagy as measured by enhanced production of both Beclin-1 and LC3B-II, markers for autophagy; and visualized by transmission electron microscopy (TEM). Similar observations were made in mouse liver lysates after mice were fed with high fat high fructose diet and treated with a long acting GLP-1 receptor agonist, liraglutide. CONCLUSIONS/SIGNIFICANCE: GLP-1 proteins appear to protect hepatocytes from fatty acid-related death by prohibition of a dysfunctional ER stress response; and reduce fatty acid accumulation, by activation of both macro-and chaperone-mediated autophagy. These findings provide a novel role for GLP-1 proteins in halting the progression of more aggressive lesions from underlying steatosis in humans afflicted with NAFLD.


Subject(s)
Autophagy/drug effects , Fatty Liver/drug therapy , Glucagon-Like Peptide 1/analogs & derivatives , Hepatocytes/drug effects , Peptides/chemistry , Peptides/pharmacology , Unfolded Protein Response/drug effects , Venoms/chemistry , Venoms/pharmacology , Adipocytes/drug effects , Adipocytes/pathology , Animals , Apoptosis/drug effects , Diet, High-Fat/adverse effects , Dietary Carbohydrates/adverse effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Exenatide , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Fructose/adverse effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Life Style , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Peptides/therapeutic use , Survival Analysis , Venoms/therapeutic use
6.
Biochem J ; 440(3): 385-95, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21846328

ABSTRACT

Adiponectin is protective against hepatic fibrosis, whereas leptin promotes fibrosis. In HSCs (hepatic stellate cells), leptin signals via a JAK2 (Janus kinase 2)/STAT3 (signal transducer and activator of transcription 3) pathway, producing effects that enhance ECM (extracellular matrix) deposition. SOCS-3 (suppressor of cytokine signalling-3) and PTP1B (protein tyrosine phosphatase 1B) are both negative regulators of JAK/STAT signalling, and recent studies have demonstrated a role for adiponectin in regulating SOCS-3 expression. In the present study we investigate mechanisms whereby adiponectin dampens leptin signalling and prevents excess ECM production. We treated culture-activated rat HSCs with recombinant adiponectin, leptin, both or neither, and also treated adiponectin knockout (Ad-/-) and wild-type mice with leptin and/or carbon tetrachloride (CCl4) or saline. We analyse JAK2 and Ob-Rb (long form of the leptin receptor) phosphorylation, and PTP1B expression and activity. We also explore potential mechanisms through which adiponectin regulates SOCS-3-Ob-Rb association. Adiponectin inhibits leptin-stimulated JAK2 activation and Ob-Rb phosphorylation in HSCs, whereas both were increased in Ad-/- mice. Adiponectin stimulates PTP1B expression and activity in vitro, whereas PTP1B expression was lower in Ad-/-mice than in wild-type mice. Adiponectin also promotes SOCS-3-Ob-R association and blocks leptin-stimulated formation of extracellular TIMP-1 (tissue inhibitor of metalloproteinases-1)-MMP-1 (matrix metalloproteinase-1) complexes in vitro. These results suggest two novel mechanisms whereby adiponectin inhibits hepatic fibrosis: (i) by promoting binding of SOCS-3 to Ob-Rb, and (ii) by stimulating PTP1B expression and activity, thus inhibiting JAK2/STAT3 signalling at multiple points.


Subject(s)
Cytoprotection , Hepatic Stellate Cells/metabolism , Leptin/physiology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/physiopathology , Adiponectin/genetics , Adiponectin/pharmacology , Adiponectin/physiology , Animals , Carbon Tetrachloride , Cells, Cultured , Gene Knockout Techniques , Humans , Janus Kinase 2/metabolism , Leptin/pharmacology , Male , Matrix Metalloproteinase 1/metabolism , Mice , Mice, Knockout , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Leptin/metabolism , Recombinant Proteins/pharmacology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism
7.
J Cell Biochem ; 110(5): 1195-207, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20564215

ABSTRACT

Adiponectin is an adipocytokine that was recently shown to be anti-fibrogenic in hepatic fibrosis. Leptin, on the other hand, promotes hepatic fibrosis. The purpose of the present study was to elucidate a mechanism (or mechanisms) whereby adiponectin dampens leptin signaling in activated hepatic stellate cells (HSCs), and prevents excess extracellular matrix production. Activated HSCs, between passages 2 and 5, were cultured and exposed to recombinant human adiponectin and recombinant leptin. Immunoblot analysis for SOCS-3, TIMP-1, and the phosphorylated species of Stat3 and adenosine monophosphate-activated protein kinase (AMPK) were conducted. We also examined MMP-1 activity by immunosorbant fluorimetric analysis. In HSCs, adiponectin-induced phosphorylation of AMPK, and subsequently suppressed leptin-mediated Stat3 phosphorylation and SOCS-3 induction. Adiponectin also blocked leptin-stimulated secretion of TIMP-1, and significantly increased MMP-1 activity, in vitro. To extend this study, we treated adiponectin knockout mice (Ad-/-) daily with 5 mg/kg recombinant leptin and/or carbon tetrachloride (2 ml/kg) for 6 weeks. Post-necropsy analysis was performed to examine for inflammation, and histological changes in the Ad-/- and wild-type mice. There was no significant difference in inflammation, or aminotransferases, between mice receiving carbon tetrachloride and leptin versus carbon tetrachloride alone. As anticipated, the combination of leptin and CCl(4) enhanced hepatic fibrosis in both wild-type and Ad-/- mice, as estimated by amount of collagen in injured livers, but wild-type mice had significantly higher levels of SOCS-3 and significantly lower levels of TIMP-1 mRNA and protein than did adiponectin KO mice exposed to both CCl(4) and leptin. We therefore conclude that the protective effects of adiponectin against liver fibrosis require AMPK activation, and may occur through inhibition of the Jak-Stat signal transduction pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Leptin/metabolism , Liver Cirrhosis/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , AMP-Activated Protein Kinases/genetics , Adiponectin/genetics , Adiponectin/pharmacology , Animals , Blotting, Western , Carbon Tetrachloride , Cells, Cultured , Female , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Leptin/genetics , Leptin/pharmacology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Mice , Mice, Knockout , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...