Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1861(7): 594-605, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27090939

ABSTRACT

Non-alcoholic steatohepatitis (NASH), is the form of non-alcoholic fatty liver disease posing risk to progress into serious long term complications. Human and pre-clinical models implicate cellular cholesterol dysregulation playing important role in its development. Mouse model studies suggest synergism between dietary cholesterol and fat in contributing to NASH but the mechanisms remain poorly understood. Our laboratory previously reported the primary importance of hepatic endoplasmic reticulum cholesterol (ER-Chol) in regulating hepatic ER stress by comparing the responses of wild type, Ldlr-/-xLcat+/+ and Ldlr-/-xLcat-/- mice, to a 2% high cholesterol diet (HCD). Here we further investigated the roles of ER-Chol and ER stress in HFHS diet-induced NASH using the same strains. With HFHS diet feeding, both WT and Ldlr-/-xLcat+/+ accumulate ER-Chol in association with ER stress and inflammasome activation but the Ldlr-/-xLcat-/- mice are protected. By contrast, all three strains accumulate cholesterol crystal, in correlation with ER-Chol, albeit less so in Ldlr-/-xLcat-/- mice. By comparison, HCD feeding per se (i) is sufficient to promote steatosis and activate inflammasomes, and (ii) results in dramatic accumulation of cholesterol crystal which is linked to inflammasome activation in Ldlr-/-xLcat-/- mice, independent of ER-Chol. Our data suggest that both dietary fat and cholesterol each independently promote steatosis, cholesterol crystal accumulation and inflammasome activation through distinct but complementary pathways. In vitro studies using palmitate-induced hepatic steatosis in HepG2 cells confirm the key roles by cellular cholesterol in the induction of steatosis and inflammasome activations. These novel findings provide opportunities for exploring a cellular cholesterol-focused strategy for treatment of NASH.


Subject(s)
Cholesterol, Dietary/metabolism , Endoplasmic Reticulum Stress/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Receptors, LDL/genetics , Animals , Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation , Hep G2 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lipid Metabolism/genetics , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Oxidation-Reduction , Palmitic Acid/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Receptors, LDL/deficiency , Signal Transduction
2.
Arthritis Rheumatol ; 68(1): 127-37, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26315393

ABSTRACT

OBJECTIVE: With no effective therapies to attenuate cartilage degeneration in osteoarthritis (OA), the result is pain and disability. Activation of hedgehog (HH) signaling causes changes related to the progression of OA, with higher levels of Gli-mediated transcriptional activation associated with increased disease severity. To elucidate the mechanism through which this occurs, this study sought to identify genes regulated by HH signaling in human OA chondrocytes. METHODS: Using human OA cartilage samples, microarray analyses were performed to detect changes in gene expression when the HH pathway was modulated. Results were analyzed for differentially expressed genes, grouped into functional networks, and validated in independent samples. To investigate the effects of chondrocyte-specific sterol accumulation, we generated mice lacking Insig1 and Insig2, which are major negative regulators of cholesterol homeostasis, under Col2a1 regulatory elements. RESULTS: HH signaling was found to regulate genes that govern cholesterol homeostasis, and this led to alterations in cholesterol accumulation in chondrocytes. A higher level of Gli-mediated transcription resulted in accumulation of intracellular cholesterol. In genetically modified mice, chondrocyte-specific cholesterol accumulation was associated with an OA phenotype. Reducing cholesterol accumulation attenuated the severity of OA in mice in vivo and decreased the expression of proteases in human OA cartilage in vitro. CONCLUSION: HH signaling regulates cholesterol homeostasis in chondrocytes, and intracellular cholesterol accumulation contributes to the severity of OA. Our findings have therapeutic implications, since reduction of HH signaling reversed cholesterol accumulation and statin treatment attenuated cartilage degeneration.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Hedgehog Proteins/metabolism , Homeostasis/genetics , Osteoarthritis/genetics , Sterols/metabolism , Stifle/metabolism , ADAM Proteins/metabolism , ADAMTS5 Protein , Animals , Anticholesteremic Agents/pharmacology , Blotting, Western , Cartilage, Articular/drug effects , Cholesterol/metabolism , Chondrocytes/drug effects , Collagen Type II/genetics , Gene Expression Regulation , Hedgehog Proteins/antagonists & inhibitors , Humans , In Vitro Techniques , Kruppel-Like Transcription Factors/genetics , Lovastatin/pharmacology , Matrix Metalloproteinase 13/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Osteoarthritis/metabolism , Radiography , Severity of Illness Index , Signal Transduction , Stifle/diagnostic imaging , Stifle/pathology , Zinc Finger Protein Gli2
3.
Arterioscler Thromb Vasc Biol ; 35(5): 1092-100, 2015 May.
Article in English | MEDLINE | ID: mdl-25675997

ABSTRACT

OBJECTIVE: Intestinal overproduction of atherogenic chylomicron particles postprandially is an important component of diabetic dyslipidemia in insulin-resistant states. In addition to enhancing insulin secretion, peripheral glucagon-like peptide-1 (GLP-1) receptor stimulation has the added benefit of reducing this chylomicron overproduction in patients with type 2 diabetes mellitus. Given the presence of central GLP-1 receptors and GLP-1-producing neurons, we assessed whether central GLP-1 exerts an integral layer of neuronal control during the production of these potentially atherogenic particles. APPROACH AND RESULTS: Postprandial production of triglyceride-rich lipoproteins was assessed in Syrian hamsters administered a single intracerebroventricular injection of the GLP-1 receptor agonist exendin-4. Intracerebroventricular exendin-4 reduced triglyceride-rich lipoprotein-triglyceride and -apolipoprotein B48 accumulation relative to vehicle-treated controls. This was mirrored by intracerebroventricular MK-0626, an inhibitor of endogenous GLP-1 degradation, and prevented by central exendin9-39, a GLP-1 receptor antagonist. The effects of intracerebroventricular exendin-4 were also lost during peripheral adrenergic receptor and central melanocortin-4 receptor inhibition, achieved using intravenous propranolol and phentolamine and intracerebroventricular HS014, respectively. However, central exendin9-39 did not preclude the effects of peripheral exendin-4 treatment on chylomicron output. CONCLUSIONS: Central GLP-1 is a novel regulator of chylomicron production via melanocortin-4 receptors. Our findings point to the relative importance of central accessibility of GLP-1-based therapies and compel further studies examining the status of this brain-gut axis in the development of diabetic dyslipidemia and chylomicron overproduction.


Subject(s)
Central Nervous System/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Lipoproteins/metabolism , Peptides/pharmacology , Receptors, Glucagon/metabolism , Venoms/pharmacology , Animals , Central Nervous System/drug effects , Chylomicrons/drug effects , Chylomicrons/metabolism , Cricetinae , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Exenatide , Glucagon-Like Peptide-1 Receptor , Intestinal Mucosa/metabolism , Intestines/innervation , Lipid Metabolism/drug effects , Lipoproteins/drug effects , Random Allocation
4.
Mol Metab ; 3(9): 823-33, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25506548

ABSTRACT

BACKGROUND/OBJECTIVES: Fasting dyslipidemia is commonly observed in insulin resistant states and mechanistically linked to hepatic overproduction of very low density lipoprotein (VLDL). Recently, the incretin hormone glucagon-like peptide-1 (GLP-1) has been implicated in ameliorating dyslipidemia associated with insulin resistance and reducing hepatic lipid stores. Given that hepatic VLDL production is a key determinant of circulating lipid levels, we investigated the role of both peripheral and central GLP-1 receptor (GLP-1R) agonism in regulation of VLDL production. METHODS: The fructose-fed Syrian golden hamster was employed as a model of diet-induced insulin resistance and VLDL overproduction. Hamsters were treated with the GLP-1R agonist exendin-4 by intraperitoneal (ip) injection for peripheral studies or by intracerebroventricular (ICV) administration into the 3rd ventricle for central studies. Peripheral studies were repeated in vagotomised hamsters. RESULTS: Short term (7-10 day) peripheral exendin-4 enhanced satiety and also prevented fructose-induced fasting dyslipidemia and hyperinsulinemia. These changes were accompanied by decreased fasting plasma glucose levels, reduced hepatic lipid content and decreased levels of VLDL-TG and -apoB100 in plasma. The observed changes in fasting dyslipidemia could be partially explained by reduced respiratory exchange ratio (RER) thereby indicating a switch in energy utilization from carbohydrate to lipid. Additionally, exendin-4 reduced mRNA markers associated with hepatic de novo lipogenesis and inflammation. Despite these observations, GLP-1R activity could not be detected in primary hamster hepatocytes, thus leading to the investigation of a potential brain-liver axis functioning to regulate lipid metabolism. Short term (4 day) central administration of exendin-4 decreased body weight and food consumption and further prevented fructose-induced hypertriglyceridemia. Additionally, the peripheral lipid-lowering effects of exendin-4 were negated in vagotomised hamsters implicating the involvement of parasympathetic signaling. CONCLUSION: Exendin-4 prevents fructose-induced dyslipidemia and hepatic VLDL overproduction in insulin resistance through an indirect mechanism involving altered energy utilization, decreased hepatic lipid synthesis and also requires an intact parasympathetic signaling pathway.

5.
Am J Physiol Endocrinol Metab ; 306(11): E1264-73, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24735884

ABSTRACT

Emerging evidence demonstrates a close interplay between disturbances in mitochondrial function and ER homeostasis in the development of the metabolic syndrome. The present investigation sought to advance our understanding of the communication between mitochondrial dysfunction and ER stress in the onset of hepatic steatosis in male rodents with defective peroxisome proliferator-activated receptor-α (PPARα) signaling. Genetic depletion of PPARα or perturbation of PPARα signaling by high-fructose diet compromised the functional activity of metabolic enzymes involved in mitochondrial fatty acid ß-oxidation and induced hepatic mitochondrial stress in rats and mice. Inhibition of PPARα activity further enhanced the expression of apolipoprotein B (apoB) mRNA and protein, which was associated with reduced mRNA expression of the sarco/endoplasmic reticulum calcium ATPase (SERCA), the induction of hepatic ER stress, and hepatic steatosis. Restoration of PPARα activity recovered the metabolic function of the mitochondria and ER, alleviated systemic hypertriglyceridemia, and improved hepatic steatosis. These findings unveil novel roles for PPARα in mediating stress signals between hepatic subcellular stress-responding machinery and in the onset of hepatic steatosis under conditions of metabolic stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Fatty Liver/metabolism , Mitochondria, Liver/metabolism , PPAR alpha/physiology , Signal Transduction/physiology , Animals , Apolipoproteins B/metabolism , Blotting, Western , Dyslipidemias/etiology , Dyslipidemias/genetics , Fatty Liver/pathology , Fructose/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Homeostasis/physiology , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Lipoproteins, VLDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , PPAR alpha/genetics , Primary Cell Culture , Rats , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ultracentrifugation
6.
Arterioscler Thromb Vasc Biol ; 33(11): 2481-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990208

ABSTRACT

OBJECTIVE: The hypothesis that cholesterol that enters the cell within low-density lipoprotein (LDL) particles rapidly equilibrates with the regulatory pool of intracellular cholesterol and maintains cholesterol homeostasis by reducing cholesterol and LDL receptor synthesis was validated in the fibroblast but not in the hepatocyte. Accordingly, the present studies were designed to compare the effects of cholesterol that enters the hepatocyte within an LDL particle with those of cholesterol that enters via other lipoprotein particles. APPROACH AND RESULTS: We measured cholesterol synthesis and esterification in hamster hepatocytes treated with LDL and other lipoprotein particles, including chylomicron remnants and VLDL. Endogenous cholesterol synthesis was not significantly reduced by uptake of LDL, but cholesterol esterification (280%) and acyl CoA:cholesterol acyltransferase 2 expression (870%) were increased. In contrast, cholesterol synthesis was significantly reduced (70% decrease) with other lipoprotein particles. Furthermore, more cholesterol that entered the hepatocyte within LDL particles was secreted within VLDL particles (480%) compared with cholesterol from other sources. CONCLUSIONS: Much of the cholesterol that enters the hepatocyte within LDL particles is shunted through the cell and resecreted within VLDL particles without reaching equilibrium with the regulatory pool.


Subject(s)
Cholesterol, LDL/metabolism , Cholesterol, VLDL/metabolism , Hepatocytes/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , Animals , Cholesterol Esters/biosynthesis , Cholesterol Esters/metabolism , Cholesterol, HDL/biosynthesis , Cholesterol, HDL/metabolism , Cholesterol, LDL/biosynthesis , Cholesterol, VLDL/biosynthesis , Chylomicrons/metabolism , Cricetinae , Fibroblasts/metabolism , Homeostasis/genetics , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipid Metabolism/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism
7.
Atherosclerosis ; 228(1): 98-109, 2013 May.
Article in English | MEDLINE | ID: mdl-23466071

ABSTRACT

Sphingolipids have emerged as important bioactive lipid species involved in the pathogenesis of type 2 diabetes and cardiovascular disease. However, little is known of the regulatory role of sphingolipids in dyslipidemia of insulin-resistant states. We employed hamster models of dyslipidemia and insulin resistance to investigate the role of sphingolipids in hepatic VLDL overproduction, induction of insulin resistance, and inflammation. Hamsters were fed either a control chow diet, a high fructose diet, or a diet high in fat, fructose and cholesterol (FFC diet). They were then treated for 2 weeks with vehicle or 0.3 mg/kg myriocin, a potent inhibitor of de novo sphingolipid synthesis. Both fructose and FFC feeding induced significant increases in hepatic sphinganine, which was normalized to chow-fed levels with myriocin (P < 0.05); myriocin also lowered hepatic ceramide content (P < 0.05). Plasma TG and cholesterol as well as VLDL-TG and -apoB100 were similarly reduced with myriocin treatment in all hamsters, regardless of diet. Myriocin treatment also led to improved insulin sensitivity and reduced hepatic SREBP-1c mRNA, though it did not appear to ameliorate the activation of hepatic inflammatory pathways. Importantly, direct treatment of primary hamster hepatocytes ex vivo with C2 ceramide or sphingosine led to an increased secretion of newly synthesized apoB100. Taken together, these data suggest that a) hepatic VLDL-apoB100 overproduction may be stimulated by ceramides and sphingosine and b) inhibition of sphingolipid synthesis can reduce circulating VLDL in hamsters and improve circulating lipids--an effect that is possibly due to improved insulin signaling and reduced lipogenesis but is independent of changes in inflammation.


Subject(s)
Apolipoprotein B-100/metabolism , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Fatty Acids, Monounsaturated/pharmacology , Lipoproteins, VLDL/metabolism , Sphingolipids/biosynthesis , Animal Feed , Animals , Cricetinae , Dietary Fats/pharmacology , Disease Models, Animal , Fructose/pharmacokinetics , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Hepatitis/drug therapy , Hepatitis/metabolism , Immunosuppressive Agents/pharmacology , Insulin Resistance/physiology , Liver/drug effects , Liver/metabolism , Male , Mesocricetus , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
8.
J Lipid Res ; 54(1): 134-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23118444

ABSTRACT

ETC-1002 (8-hydroxy-2,2,14,14-tetramethylpentadecanedioic acid) is a novel investigational drug being developed for the treatment of dyslipidemia and other cardio-metabolic risk factors. The hypolipidemic, anti-atherosclerotic, anti-obesity, and glucose-lowering properties of ETC-1002, characterized in preclinical disease models, are believed to be due to dual inhibition of sterol and fatty acid synthesis and enhanced mitochondrial long-chain fatty acid ß-oxidation. However, the molecular mechanism(s) mediating these activities remained undefined. Studies described here show that ETC-1002 free acid activates AMP-activated protein kinase in a Ca(2+)/calmodulin-dependent kinase ß-independent and liver kinase ß 1-dependent manner, without detectable changes in adenylate energy charge. Furthermore, ETC-1002 is shown to rapidly form a CoA thioester in liver, which directly inhibits ATP-citrate lyase. These distinct molecular mechanisms are complementary in their beneficial effects on lipid and carbohydrate metabolism in vitro and in vivo. Consistent with these mechanisms, ETC-1002 treatment reduced circulating proatherogenic lipoproteins, hepatic lipids, and body weight in a hamster model of hyperlipidemia, and it reduced body weight and improved glycemic control in a mouse model of diet-induced obesity. ETC-1002 offers promise as a novel therapeutic approach to improve multiple risk factors associated with metabolic syndrome and benefit patients with cardiovascular disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , ATP Citrate (pro-S)-Lyase/metabolism , Carbohydrate Metabolism/drug effects , Dicarboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acids/pharmacology , Lipid Metabolism/drug effects , Molecular Targeted Therapy/methods , AMP-Activated Protein Kinase Kinases , ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcium/metabolism , Cricetinae , Dicarboxylic Acids/chemistry , Dicarboxylic Acids/therapeutic use , Diet/adverse effects , Dyslipidemias/blood , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Fatty Acids/biosynthesis , Fatty Acids/chemistry , Fatty Acids/therapeutic use , Female , Glucagon/metabolism , Glucose/biosynthesis , Hep G2 Cells , Humans , Liver/cytology , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Mice , Obesity/blood , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Signal Transduction/drug effects , Sterols/biosynthesis
9.
Circ Res ; 110(10): 1345-54, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22474253

ABSTRACT

RATIONALE: Dysregulation of hepatic triglyceride (TG)-rich very low-density lipoproteins (VLDL-TG) in obesity and type 2 diabetes contributes to the dyslipidemia that leads to cardiovascular morbidity. The central nervous system (CNS), particularly the hypothalamus, regulates hepatic lipid metabolism. Although the underlying neurocircuitry remains elusive, glycine has been documented to enhance CNS N-methyl-d-aspartate (NMDA) receptor-mediated transmission. OBJECTIVE: We tested the hypothesis that glycine regulates hepatic VLDL-TG secretion by potentiating NMDA receptor-mediated transmission in the CNS. METHODS AND RESULTS: Using 10-hour fasted male Sprague-Dawley rats implanted with stereotaxic cannulae into an extrahypothalamic region termed the dorsal vagal complex (DVC) and vascular catheters to enable direct DVC infusion and blood sampling, respectively, the rate of hepatic VLDL-TG secretion was measured following tyloxapol (an inhibitor of lipoprotein lipase) injection. Direct DVC infusion of glycine lowered VLDL-TG secretion, whereas NMDA receptor blocker MK-801 fully negated glycine's effect. NR1 subunit of NMDA receptor antagonist 7-chlorokynurenic acid, adenoviral injection of NR1 short hairpin RNA (shRNA), and hepatic vagotomy also nullified glycine's effect. Finally, DVC glycine normalized the hypersecretion of VLDL-TG induced by high-fat feeding. CONCLUSIONS: Molecular and pharmacological inhibition of the NR1-containing NMDA receptors in the DVC negated the ability of glycine to inhibit hepatic secretion of VLDL-TG in vivo. Importantly, the hypersecretion of VLDL-TG from the liver induced by a model of high-fat feeding was restored by the hepatic lipid control of CNS glycine sensing. These findings collectively suggest that glycine or glycine analogues may have therapeutic benefits in lowering plasma lipid levels in diabetes and obesity by triggering the CNS.


Subject(s)
Cholesterol, VLDL/metabolism , Glycine/metabolism , Hypothalamus/metabolism , Liver/metabolism , Triglycerides/metabolism , Adiponectin/blood , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dietary Fats/pharmacology , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Fatty Acids, Nonesterified/blood , Glycine/pharmacology , Insulin/blood , Leptin/blood , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Liver/drug effects , Male , Obesity/drug therapy , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/metabolism , Vagotomy
10.
J Biol Chem ; 287(24): 20755-68, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22500017

ABSTRACT

We recently reported that lecithin:cholesterol acyltransferase (LCAT) knock-out mice, particularly in the LDL receptor knock-out background, are hypersensitive to insulin and resistant to high fat diet-induced insulin resistance (IR) and obesity. We demonstrated that chow-fed Ldlr-/-xLcat+/+ mice have elevated hepatic endoplasmic reticulum (ER) stress, which promotes IR, compared with wild-type controls, and this effect is normalized in Ldlr-/-xLcat-/- mice. In the present study, we tested the hypothesis that hepatic ER cholesterol metabolism differentially regulates ER stress using these models. We observed that the Ldlr-/-xLcat+/+ mice accumulate excess hepatic total and ER cholesterol primarily attributed to increased reuptake of biliary cholesterol as we observed reduced biliary cholesterol in conjunction with decreased hepatic Abcg5/g8 mRNA, increased Npc1l1 mRNA, and decreased Hmgr mRNA and nuclear SREBP2 protein. Intestinal NPC1L1 protein was induced. Expression of these genes was reversed in the Ldlr-/-xLcat-/- mice, accounting for the normalization of total and ER cholesterol and ER stress. Upon feeding a 2% high cholesterol diet (HCD), Ldlr-/-xLcat-/- mice accumulated a similar amount of total hepatic cholesterol compared with the Ldlr-/-xLcat+/+ mice, but the hepatic ER cholesterol levels remained low in conjunction with being protected from HCD-induced ER stress and IR. Hepatic ER stress correlates strongly with hepatic ER free cholesterol but poorly with hepatic tissue free cholesterol. The unexpectedly low ER cholesterol seen in HCD-fed Ldlr-/-xLcat-/- mice was attributable to a coordinated marked up-regulation of ACAT2 and suppressed SREBP2 processing. Thus, factors influencing the accumulation of ER cholesterol may be important for the development of hepatic insulin resistance.


Subject(s)
Cholesterol/metabolism , Endoplasmic Reticulum Stress , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Liver/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase , Receptors, LDL/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Cholesterol/genetics , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Gene Expression Regulation/genetics , Insulin Resistance/genetics , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Lipoproteins/biosynthesis , Lipoproteins/genetics , Liver/pathology , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, LDL/genetics , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Sterol O-Acyltransferase 2
11.
Am J Physiol Gastrointest Liver Physiol ; 302(9): G1043-52, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22345552

ABSTRACT

Ezetimibe is a cholesterol uptake inhibitor that targets the Niemann-Pick C1-like 1 cholesterol transporter. Ezetimibe treatment has been shown to cause significant decreases in plasma cholesterol levels in patients with hypercholesterolemia and familial hypercholesterolemia. A recent study in humans has shown that ezetimibe can decrease the release of atherogenic postprandial intestinal lipoproteins. In the present study, we evaluated the mechanisms by which ezetimibe treatment can lower postprandial apoB48-containing chylomicron particles, using a hyperlipidemic and insulin-resistant hamster model fed a diet rich in fructose and fat (the FF diet) and fructose, fat, and cholesterol (the FFC diet). Male Syrian Golden hamsters were fed either chow or the FF or FFC diet ± ezetimibe for 2 wk. After 2 wk, chylomicron production was assessed following intravenous triton infusion. Tissues were then collected and analyzed for protein and mRNA content. FFC-fed hamsters treated with ezetimibe showed improved glucose tolerance, decreased fasting insulin levels, and markedly reduced circulating levels of TG and cholesterol in both the LDL and VLDL fractions. Examination of triglyceride (TG)-rich lipoprotein (TRL) fractions showed that ezetimibe treatment reduced postprandial cholesterol content in TRL lipoproteins as well as reducing apoB48 content. Although ezetimibe did not decrease TRL-TG levels in FFC hamsters, ezetimibe treatment in FF hamsters resulted in decreases in TRL-TG. Jejunal apoB48 protein expression was lower in ezetimibe-treated hamsters. Reductions in jejunal protein levels of scavenger receptor type B-1 (SRB-1) and fatty acid transport protein 4 were also observed. In addition, ezetimibe-treated hamsters showed significantly lower jejunal mRNA expression of a number of genes involved in lipid synthesis and transport, including srebp-1c, sr-b1, ppar-γ, and abcg1. These data suggest that treatment with ezetimibe not only inhibits cholesterol uptake, but may also alter intestinal function to promote improved handling of dietary lipids and reduced chylomicron production. These, in turn, promote decreases in fasting and postprandial lipid levels and improvements in glucose homeostasis.


Subject(s)
Azetidines/administration & dosage , Blood Glucose/metabolism , Chylomicrons/biosynthesis , Disease Models, Animal , Glucose Tolerance Test , Metabolic Syndrome/drug therapy , Metabolic Syndrome/physiopathology , Animals , Anticholesteremic Agents/administration & dosage , Blood Glucose/drug effects , Cricetinae , Diet, High-Fat , Ezetimibe , Male , Mesocricetus , Treatment Outcome
12.
J Biol Chem ; 287(13): 10277-10288, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22275361

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is becoming the leading cause of chronic liver disease and is now considered to be the hepatic manifestation of the metabolic syndrome. However, the role of steatosis per se and the precise factors required in the progression to steatohepatitis or insulin resistance remain elusive. The JAK-STAT pathway is critical in mediating signaling of a wide variety of cytokines and growth factors. Mice with hepatocyte-specific deletion of Janus kinase 2 (L-JAK2 KO mice) develop spontaneous steatosis as early as 2 weeks of age. In this study, we investigated the metabolic consequences of jak2 deletion in response to diet-induced metabolic stress. To our surprise, despite the profound hepatosteatosis, deletion of hepatic jak2 did not sensitize the liver to accelerated inflammatory injury on a prolonged high fat diet (HFD). This was accompanied by complete protection against HFD-induced whole-body insulin resistance and glucose intolerance. Improved glucose-stimulated insulin secretion and an increase in ß-cell mass were also present in these mice. Moreover, L-JAK2 KO mice had progressively reduced adiposity in association with blunted hepatic growth hormone signaling. These mice also exhibited increased resting energy expenditure on both chow and high fat diet. In conclusion, our findings indicate a key role of hepatic JAK2 in metabolism such that its absence completely arrests steatohepatitis development and confers protection against diet-induced systemic insulin resistance and glucose intolerance.


Subject(s)
Dietary Fats/adverse effects , Fatty Liver/enzymology , Glucose Intolerance/enzymology , Hepatocytes/enzymology , Janus Kinase 2/metabolism , Adiposity/drug effects , Adiposity/genetics , Animals , Dietary Fats/pharmacology , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Gene Deletion , Glucose Intolerance/chemically induced , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Hepatocytes/pathology , Insulin Resistance/genetics , Janus Kinase 2/genetics , Mice , Mice, Knockout
13.
Hepatology ; 53(1): 127-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20967757

ABSTRACT

UNLABELLED: Plasma C-reactive protein (CRP) concentration is increased in the metabolic syndrome, which consists of a cluster of cardiovascular disease risk factors, including insulin resistance. It is not known, however, whether CRP is merely a marker of accompanying inflammation or whether it contributes causally to insulin resistance. The objective of this study is to investigate the role that CRP may play in the development of insulin resistance. We examined the effect of single-dose intravenous administration of purified human (h)CRP on insulin sensitivity in Sprague-Dawley rats using the euglycemic, hyperinsulinemic clamp technique. hCRP was associated with impaired insulin suppression of endogenous glucose production with no reduction in peripheral tissue glucose uptake, suggesting that hCRP mediated insulin resistance in the liver but not extrahepatic tissues. We further assessed components of the insulin signaling pathway and mitogen-activated protein kinases (MAPKs) in the liver. Liver tissues derived from hCRP-treated rats showed reduced insulin-stimulated insulin receptor substrate (IRS) tyrosine phosphorylation, IRS/phosphatidylinositol 3-kinase (PI3K) association, and Akt phosphorylation, consistent with hCRP-induced impairment of hepatic insulin signaling. Furthermore, hCRP enhanced phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and p38 MAPK as well as IRS-1 Ser(612) . Finally, we observed in primary cultured rat hepatocytes that U0126 (a selective inhibitor of MAPK/ERK kinase1/2) corrected hCRP-induced impairment of insulin signaling. CONCLUSIONS: hCRP plays an active role in inducing hepatic insulin resistance in the rat, at least in part by activating ERK1/2, with downstream impairment in the insulin signaling pathway.


Subject(s)
C-Reactive Protein/pharmacology , Insulin Resistance , Insulin/physiology , Liver/metabolism , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/physiology , Adiponectin/blood , Animals , Butadienes/pharmacology , Glucose Clamp Technique , Humans , Imidazoles/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Interleukin-6/blood , Leptin/blood , Male , Nitriles/pharmacology , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
14.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G323-32, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19497957

ABSTRACT

Liver X receptor-alpha (LXRalpha) is considered a master regulator of hepatic lipid metabolism; however, little is known about the link between LXR activation, hepatic insulin signaling, and very low-density lipoprotein (VLDL)-apolipoprotein B (apoB) assembly and secretion. Here, we examined the effect of LXRalpha activation on hepatic insulin signaling and apoB-lipoprotein production. In vivo activation of LXRalpha for 7 days using a synthetic LXR agonist, TO901317, in hamsters led to increased plasma triglyceride (TG; 3.6-fold compared with vehicle-treated controls, P = 0.006), apoB (54%, P < 0.0001), and VLDL-TG (eightfold increase compared with vehicle). As expected, LXR stimulation activated maturation of sterol response element binding protein-1c (SREBP-1c) as well as the SREBP-1c target genes steroyl CoA desaturase (SCD) and fatty acid synthase (FAS). Metabolic pulse-chase labeling experiments in primary hamster hepatocytes showed increased stability and secretion of newly synthesized apoB following LXR activation. Microsomal triglyceride transfer protein (MTP) mRNA and protein were unchanged, however, likely because of the relatively short period of treatment and long half-life of MTP mRNA. Examination of hepatic insulin-signaling molecules revealed LXR-mediated reductions in insulin receptor (IR)beta subunit mass (39%, P = 0.014) and insulin receptor substrate (IRS)-1 tyrosine phosphorylation (24%, P = 0.023), as well as increases in protein tyrosine phosphatase (PTP)1B (29%, P < 0.001) protein mass. In contrast to IRS-1, a twofold increase in IRS-2 mass (228%, P = 0.0037) and a threefold increase in IRS-2 tyrosine phosphorylation (321%, P = 0.012) were observed. In conclusion, LXR activation dysregulates hepatic insulin signaling and leads to a considerable increase in the number of circulating TG-rich VLDL-apoB particles, likely due to enhanced hepatic assembly and secretion of apoB-containing lipoproteins.


Subject(s)
Apolipoproteins B/metabolism , DNA-Binding Proteins/agonists , Hepatocytes/drug effects , Hydrocarbons, Fluorinated/pharmacology , Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Signal Transduction/drug effects , Sulfonamides/pharmacology , Administration, Oral , Animals , Apolipoprotein B-100/metabolism , Apolipoproteins B/blood , Carrier Proteins/metabolism , Cells, Cultured , Cricetinae , DNA-Binding Proteins/metabolism , Fatty Acid Synthases/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Hydrocarbons, Fluorinated/administration & dosage , Insulin Receptor Substrate Proteins/metabolism , Lipoproteins, VLDL/blood , Liver X Receptors , Male , Mesocricetus , Orphan Nuclear Receptors , Phosphorylation , Protein Stability , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/genetics , Stearoyl-CoA Desaturase/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sulfonamides/administration & dosage , Time Factors , Triglycerides/blood
15.
Am J Physiol Endocrinol Metab ; 297(2): E462-73, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19509184

ABSTRACT

Although the atherogenic role of dietary cholesterol has been well established, its diabetogenic potential and associated metabolic disturbances have not been reported. Diet-induced hamster models of insulin resistance and dyslipidemia were employed to determine lipogenic and diabetogenic effects of dietary cholesterol. Metabolic studies were conducted in hamsters fed diets rich in fructose (40%), fat (30%), and cholesterol (0.05-0.25%) (FFC) and other test diets. Short-term feeding of the FFC diet induced insulin resistance, glucose intolerance, hypertriglyceridemia, and hypercholesterolemia. Prolonged feeding (6-22 wk) of the FFC diet led to severe hepatic steatosis, glucose intolerance, and mild increases in fasting blood glucose, suggesting progression toward type 2 diabetes, but did not induce beta-cell dysfunction. Metabolic changes induced by the diet, including dyslipidemia and insulin resistance, were cholesterol concentration dependent and were only markedly induced on a high-fructose and high-fat dietary background. There were significant increases in hepatic and plasma triglyceride with FFC feeding, likely due to a 10- to 15-fold induction of hepatic stearoyl-CoA desaturase compared with chow levels (P < 0.03). Hepatic insulin resistance was evident based on reduced tyrosine phosphorylation of the insulin receptor-beta, IRS-1, and IRS-2 as well as increased protein mass of protein tyrosine phosphatase 1B. Interestingly, nuclear liver X receptor (LXR) target genes such as ABCA1 were upregulated on the FFC diet, and dietary supplementation with an LXR agonist (instead of dietary cholesterol) worsened dyslipidemia, glucose intolerance, and upregulation of target mRNA and proteins similar to that of dietary cholesterol. In summary, these data clearly implicate dietary cholesterol, synergistically acting with dietary fat and fructose, as a major determinant of the severity of metabolic disturbances in the hamster model. Dietary cholesterol appears to induce hepatic cholesterol ester and triglyceride accumulation, and diet-induced LXR activation (via cholesterol-derived oxysterols) may possibly be one key underlying mechanism.


Subject(s)
Cholesterol, Dietary/pharmacology , Fatty Liver/metabolism , Insulin Resistance , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cricetinae , Dietary Fats/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Fatty Liver/pathology , Fructose/pharmacology , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Lipids/blood , Male , Mesocricetus , Metabolic Diseases/etiology , Triglycerides/blood , Triglycerides/metabolism
16.
Am J Physiol Gastrointest Liver Physiol ; 296(6): G1287-98, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19342510

ABSTRACT

Insulin-resistant states are commonly associated with chronic inflammation and hepatic overproduction of apolipoprotein B100 (apoB100), leading to hypertriglyceridemia and a metabolic dyslipidemic profile. Molecular mechanisms linking hepatic inflammatory cascades and the pathways of apoB100-lipoprotein production are, however, unknown. In the present study, we employed a diet-induced, insulin-resistant hamster model, as well as cell culture studies, to investigate the potential link between activation of hepatic inflammatory nuclear factor-kappaB (NF-kappaB) signaling cascade and the synthesis and secretion of apoB100-containing lipoproteins. Using an established insulin-resistant animal model, the fructose-fed hamster, we found that feeding fructose (previously shown to induce hepatic inflammation) for as little as 4 days reduced hepatic IkappaB (inhibitor of NF-kappaB) level, indicating activation of the inflammatory NF-kappaB cascade. Importantly, IKK (IkappaB kinase) inhibition was found to suppress apoB100 overproduction in fructose-fed hamster hepatocytes. As IKK, the upstream activator of NF-kappaB has been shown to inhibit insulin signaling, and insulin is a major regulator of apoB100, we modulated IKK activity in primary hamster hepatocytes and HepG2 cells and assessed the effects on hepatic apoB100 biosynthesis. Inhibition of the IKK-NF-kappaB pathway by BMS345541 and activation of the pathway by adenoviral-mediated IKK overexpression decreased and increased newly synthesized apoB100 levels, respectively. Pulse-chase and metabolic labeling experiments revealed that IKK activation regulates apoB100 levels at the levels of apoB100 biosynthesis and protein stability. Inhibition of the IKK-NF-kappaB pathway significantly enhanced proteasomal degradation of hepatic apoB100, while direct IKK activation led to reduced degradation and increased apoB100 mRNA translation. Together, our results reveal important links between modulation of the inflammatory IKK-NF-kappaB signaling cascade and hepatic synthesis and secretion of apoB100-containing lipoproteins. Hepatic inflammation may be an important underlying factor in hepatic apoB100 overproduction observed in insulin resistance.


Subject(s)
Apolipoprotein B-100/metabolism , Inflammation/metabolism , Lipoproteins/biosynthesis , Liver/metabolism , NF-kappa B/metabolism , Administration, Oral , Animals , Apolipoprotein B-100/biosynthesis , Apolipoprotein B-100/genetics , Cell Line, Tumor , Cell-Free System/drug effects , Cell-Free System/metabolism , Cricetinae , Cysteine Proteinase Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fructose/administration & dosage , Fructose/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Imidazoles/pharmacology , Inflammation/chemically induced , Leupeptins/pharmacology , Lipid Metabolism/drug effects , Lipoproteins, VLDL/metabolism , Male , Mesocricetus , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , Quinoxalines/pharmacology , Transfection , Tumor Necrosis Factor-alpha/metabolism
17.
Endocrinology ; 150(8): 3483-92, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19359385

ABSTRACT

The carbohydrate response element binding protein (ChREBP) has been recognized as a key controller of hepatic lipogenesis. Whereas the function of ChREBP has been extensively investigated, mechanisms underlying its transcription remain largely unknown, although ChREBP production is elevated in a hyperinsulinemic mouse model. We located a conserved Pit-1, Oct-1/Oct-2, and Unc-86 (POU) protein binding site (ATGCTAAT) within the proximal promoter region of human ChREBP. This site interacts with the POU homeodomain protein octamer transcription factor-1 (Oct-1), as detected by gel shift and chromatin immunoprecipitation assays. Oct-1 cotransfection in the human HepG2 cell line repressed ChREBP promoter activity approximately 50-75% (P < 0.01 to P < 0.001), and this repression was dependent on the existence of the POU binding site. Furthermore, overexpression of Oct-1 repressed endogenous ChREBP mRNA and protein expression, whereas knockdown of Oct-1 expression, using a lentivirus-based small hairpin RNA approach, led to increased ChREBP mRNA and protein expression. In contrast, HepG2 cells treated with 10 or 100 nM insulin for 4 or 8 h resulted in an approximately 2-fold increase of ChREBP promoter activity (P < 0.05 to P < 0.01). Insulin (10 nM) also stimulated endogenous ChREBP expression in HepG2 and primary hamster hepatocytes. More importantly, we found that the stimulatory effect of insulin on ChREBP promoter activity was dependent on the presence of the POU binding site, and insulin treatment reduced Oct-1 expression levels. Our observations therefore identify Oct-1 as a transcriptional repressor of ChREBP and suggest that insulin stimulates ChREBP expression via attenuating the repressive effect of Oct-1.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Octamer Transcription Factor-1/physiology , POU Domain Factors/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Cricetinae , Electrophoretic Mobility Shift Assay , Gene Expression/drug effects , Humans , Lentivirus/genetics , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Hepatology ; 48(6): 1799-809, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19026012

ABSTRACT

Hepatic apolipoprotein B (apoB) lipoprotein production is metabolically regulated via the phosphoinositide 3-kinase cascade; however, the role of the key negative regulator of this pathway, the tumor suppressor phosphatase with tensin homology (PTEN), is unknown. Here, we demonstrate that hepatic protein levels of apoB100 and microsomal triglyceride transfer protein (MTP) are significantly down-regulated (73% and 36%, respectively) in the liver of PTEN liver-specific knockout (KO) mice, and this is accompanied by increased triglyceride (TG) accumulation and lipogenic gene expression, and reduced hepatic apoB secretion in freshly isolated hepatocytes. MTP protein mass and lipid transfer activity were also significantly reduced in liver of PTEN KO mice. Overexpression of the dominant negative mutant PTEN C/S124 (adenovirus expressing PTEN C/S mutant [AdPTENC/S]) possessing constitutive phospoinositide 3-kinase activity in HepG2 cells led to significant reductions in both secreted apoB100 and cellular MTP mass (76% and 34%, respectively), and increased messenger RNA (mRNA) levels of sterol regulatory element binding protein 1c (SREBP-1c), fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC). Reduced apoB100 secretion induced by AdPTENC/S was associated with increased degradation of newly-synthesized cellular apoB100, in a lactacystin-sensitive manner, suggesting enhanced proteasomal degradation. AdPTENC/S also reduced apoB-lipoprotein production in McA-RH7777 and primary hamster hepatocytes. Our findings suggest a link between PTEN expression and hepatic production of apoB-containing lipoproteins. We postulate that perturbations in PTEN not only may influence hepatic insulin signaling and hepatic lipogenesis, but also may alter hepatic apoB-lipoprotein production and the MTP stability. On loss of PTEN activity, increased lipid substrate availability in the face of reduced hepatic lipoprotein production capacity can rapidly lead to hepatosteatosis and fatty liver.


Subject(s)
Apolipoproteins B/metabolism , Carrier Proteins/metabolism , Fatty Liver/metabolism , Lipogenesis/physiology , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Apolipoprotein B-100/metabolism , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Acid Synthases/metabolism , Fatty Liver/pathology , Insulin/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
19.
Atherosclerosis ; 198(1): 94-103, 2008 May.
Article in English | MEDLINE | ID: mdl-18093597

ABSTRACT

Statin-treatment of fructose-fed/insulin resistant hamsters was recently shown to ameliorate metabolic dyslipidemia and hepatic VLDL overproduction. Here, we provide evidence that rosuvastatin treatment of insulin resistant hamsters can induce improvements in hepatic and whole body insulin sensitivity. Treatment with 10 mg/kg/day rosuvastatin for 10 days significantly reduced fasting insulin (-59%) and triglyceride (-50%) levels in fructose-fed hamsters (p<0.05). Following an intraperitoneal (IP) glucose challenge, rosuvastatin-treated hamsters exhibited enhanced glucose clearance compared to untreated hamsters maintained on the high-fructose diet (area under curve (AUC)=1772+/-223 mM min vs. 2413+/-253 mM min, respectively; p<0.002) with a significant reduction in 2h post-challenge glucose (n=5, p<0.02). Rosuvastatin-treatment also significantly improved sensitivity to an IP insulin challenge (AUC=314+/-39 mM min vs. 195+/-22 mM min for rosuvastatin-treated and fructose-fed hamsters, respectively; p<0.04, n=3). At the molecular level, significant increases in tyrosine-phosphorylation of the hepatic insulin receptor and IRS-1 were observed for rosuvastatin-treated hamsters (+37% and +58%, respectively) compared to fructose-fed controls following an intravenous (IV) bolus of insulin (p<0.05). Increases in insulin receptor and IRS-1 phosphorylation were also observed in muscle and adipose tissue. Analysis of hepatic Akt phosphorylation and mass revealed a small (25%) increase in serine phosphorylation of Akt with no significant change in Akt mass, although serine-phosphorylation and mass of Akt2 were significantly increased (+32%, p=0.03, and +42%, p=0.01, respectively). Interestingly, expression of PTP-1B, a key negative regulator of insulin signaling, showed a non-significant trend toward reduction in liver and was significantly reduced in adipose tissue (-20% and -37%, respectively). Taken together, these data suggest that statin-treatment increases whole body and peripheral tissue insulin sensitivity via improved cellular insulin signal transduction.


Subject(s)
Fluorobenzenes/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Insulin Resistance , Liver/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cricetinae , Disease Models, Animal , Fructose/pharmacology , Injections, Intravenous , Insulin/blood , Insulin Receptor Substrate Proteins , Male , Mesocricetus , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Rosuvastatin Calcium , Triglycerides/blood
20.
Am J Physiol Endocrinol Metab ; 293(2): E587-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17551001

ABSTRACT

We previously reported that LCAT-deficient mice develop not only low HDL-cholesterol but also hypertriglyceridemia, hepatic triglyceride (TG) overproduction, and, unexpectedly, improved hepatic insulin sensitivity and reduced hepatic TG content. Here, we examined the mechanistic links underlying this apparent paradox. The LDL receptor-deficient (Ldlr)(-/-)xLcat(-/-) mouse model and age- and sex-matched Ldlr(-/-)xLcat(+/+) littermates, both in C57Bl/6 background, were employed. Studies of hepatic insulin signal transduction showed an upregulation of hepatic Irs2 mRNA level (5.3-fold, P = 0.02), IRS-2 protein mass level (1.5-fold, P = 0.009) and pIRS-2 (1.8-fold. P = 0.02) in the Ldlr(-/-)xLcat(-/-) mice. There was a 1.2-fold increase in pAkt (P = 0.03) with a nonsignificant change in total Akt. We observed a significant shift in its downstream transcription factor FoxO-1 to the cytosolic compartment (2.3-fold increase in cytosolic/nuclear ratio, P = 0.04). We also observed a significant 3.1-fold increase in nuclear abundance of FoxA-2 mass (P = 0.017) and a 1.5-fold upregulation of its coactivator PGC-1beta (P = 0.002), the coordinated actions of which promotes hepatic TG production and beta-oxidation. Increased hepatic insulin signaling in the Ldlr(-/-)xLcat(-/-) mice was associated with an upregulation of the Tcfe3 gene (1.7-fold, P = 0.024), a selective downregulation of the Socs-1 gene by 60% (P = 0.01), and no change in PTP-1B protein mass. These data suggest that LCAT deficiency induces complex alterations in hepatic signal transduction cascades, which explain, at least in part, the observed enhanced insulin signaling in association with hepatic TG overproduction and reduced hepatic TG content.


Subject(s)
Insulin Resistance/genetics , Liver/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Insulin/metabolism , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...