Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(3): 105684, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272231

ABSTRACT

Eukaryotic elongation factor 1A1 (EEF1A1) is canonically involved in protein synthesis but also has noncanonical functions in diverse cellular processes. Previously, we identified EEF1A1 as a mediator of lipotoxicity and demonstrated that chemical inhibition of EEF1A1 activity reduced mouse liver lipid accumulation. These findings suggested a link between EEF1A1 and metabolism. Therefore, we investigated its role in regulating metabolic substrate preference. EEF1A1-deficient Chinese hamster ovary (2E2) cells displayed reduced media lactate accumulation. These effects were also observed with EEF1A1 knockdown in human hepatocyte-like HepG2 cells and in WT Chinese hamster ovary and HepG2 cells treated with selective EEF1A inhibitors, didemnin B, or plitidepsin. Extracellular flux analyses revealed decreased glycolytic ATP production and increased mitochondrial-to-glycolytic ATP production ratio in 2E2 cells, suggesting a more oxidative metabolic phenotype. Correspondingly, fatty acid oxidation was increased in 2E2 cells. Both 2E2 cells and HepG2 cells treated with didemnin B exhibited increased neutral lipid content, which may be required to support elevated oxidative metabolism. RNA-seq revealed a >90-fold downregulation of a rate-limiting glycolytic enzyme, hexokinase 2, which we confirmed through immunoblotting and enzyme activity assays. Pathway enrichment analysis identified downregulations in TNFA signaling via NFKB and MYC targets. Correspondingly, nuclear abundances of RELB and MYC were reduced in 2E2 cells. Thus, EEF1A1 deficiency may perturb glycolysis by limiting NFKB- and MYC-mediated gene expression, leading to decreased hexokinase expression and activity. This is the first evidence of a role for a translation elongation factor, EEF1A1, in regulating metabolic substrate utilization in mammalian cells.


Subject(s)
Hexokinase , Peptide Elongation Factor 1 , Animals , Cricetinae , Humans , Adenosine Triphosphate , Cell Line , Cricetulus , Hexokinase/genetics , Hexokinase/metabolism , Lipids , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor 1/metabolism , Glycolysis , Oxidation-Reduction , Cell Movement , Cell Proliferation , Lipid Metabolism
2.
Front Endocrinol (Lausanne) ; 13: 938680, 2022.
Article in English | MEDLINE | ID: mdl-36093092

ABSTRACT

Obesity plays a major role in type II diabetes (T2DM) progression because it applies metabolic and oxidative stress resulting in dysfunctional beta-cells and activation of intra-islet pancreatic stellate cells (PaSCs) which cause islet fibrosis. Administration of antioxidant N-acetyl-L-cysteine (NAC) in vivo improves metabolic outcomes in diet-induced obese diabetic mice, and in vitro inhibits PaSCs activation. However, the effects of NAC on diabetic islets in vivo are unknown. This study examined if dosage and length of NAC treatment in HFD-induced diabetic mice effect metabolic outcomes associated with maintaining healthy beta-cells and quiescent PaSCs, in vivo. Male C57BL/6N mice were fed normal chow (ND) or high-fat (HFD) diet up to 30 weeks. NAC was administered in drinking water to HFD mice in preventative treatment (HFDpNAC) for 23 weeks or intervention treatment for 10 (HFDiNAC) or 18 (HFDiNAC+) weeks, respectively. HFDpNAC and HFDiNAC+, but not HFDiNAC, mice showed significantly improved glucose tolerance and insulin sensitivity. Hyperinsulinemia led by beta-cell overcompensation in HFD mice was significantly rescued in NAC treated mice. A reduction of beta-cell nuclear Pdx-1 localization in HFD mice was significantly improved in NAC treated islets along with significantly reduced beta-cell oxidative stress. HFD-induced intra-islet PaSCs activation, labeled by αSMA, was significantly diminished in NAC treated mice along with lesser intra-islet collagen deposition. This study determined that efficiency of NAC treatment is beneficial at maintaining healthy beta-cells and quiescent intra-islet PaSCs in HFD-induced obese T2DM mouse model. These findings highlight an adjuvant therapeutic potential in NAC for controlling T2DM progression in humans.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/etiology , Oxidative Stress , Pancreatic Stellate Cells/metabolism
3.
Int J Mol Sci ; 21(23)2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33271781

ABSTRACT

Prolonged, isocaloric, time-restricted feeding (TRF) protocols can promote weight loss, improve metabolic dysregulation, and mitigate non-alcoholic fatty liver disease (NAFLD). In addition, 3-day, severe caloric restriction can improve liver metabolism and glucose homeostasis prior to significant weight loss. Thus, we hypothesized that short-term, isocaloric TRF would improve NAFLD and characteristics of metabolic syndrome in diet-induced obese male mice. After 26 weeks of ad libitum access to western diet, mice either continued feeding ad libitum or were provided with access to the same quantity of western diet for 8 h daily, over the course of two weeks. Remarkably, this short-term TRF protocol modestly decreased liver tissue inflammation in the absence of changes in body weight or epidydimal fat mass. There were no changes in hepatic lipid accumulation or other characteristics of NAFLD. We observed no changes in liver lipid metabolism-related gene expression, despite increased plasma free fatty acids and decreased plasma triglycerides in the TRF group. However, liver Grp78 and Txnip expression were decreased with TRF suggesting hepatic endoplasmic reticulum (ER) stress and activation of inflammatory pathways may have been diminished. We conclude that two-week, isocaloric TRF can potentially decrease liver inflammation, without significant weight loss or reductions in hepatic steatosis, in obese mice with NAFLD.


Subject(s)
Body Weight , Fasting , Hepatitis/etiology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Animals , Biomarkers , Biopsy , Blood Glucose , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Glucose/metabolism , Hepatitis/metabolism , Hepatitis/pathology , Lipid Metabolism , Liver/metabolism , Liver/pathology , Mice , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism
4.
Pharmacol Res ; 161: 105208, 2020 11.
Article in English | MEDLINE | ID: mdl-32977024

ABSTRACT

Inhibition of eukaryotic elongation factor 1A1 (EEF1A1) with the marine compound didemnin B decreases lipotoxic HepG2 cell death in vitro and improves early stage non-alcoholic fatty liver disease (NAFLD) in young genetically obese mice. However, the effects of didemnin B on NAFLD in a model of long-term diet-induced obesity are not known. We investigated the effects of didemnin B on NAFLD severity and metabolic parameters in western diet-induced obese mice, and on the cell types that contribute to liver inflammation and fibrosis in vitro. Male 129S6 mice were fed either standard chow or western diet for 26 weeks, followed by intervention with didemnin B (50 µg/kg) or vehicle by intraperitoneal (i.p.) injection once every 3 days for 14 days. Didemnin B decreased liver and plasma triglycerides, improved oral glucose tolerance, and decreased NAFLD severity. Moreover, didemnin B moderately increased hepatic expression of genes involved in ER stress response (Perk, Chop), and fatty acid oxidation (Fgf21, Cpt1a). In vitro, didemnin B decreased THP-1 monocyte proliferation, disrupted THP-1 monocyte-macrophage differentiation, decreased THP-1 macrophage IL-1ß secretion, and decreased hepatic stellate cell (HSteC) proliferation and collagen secretion under both basal and lipotoxic (high fatty acid) conditions. Thus, didemnin B improves hepatic steatosis, glucose tolerance, and blood lipids in obesity, in association with moderate, possibly hormetic, upregulation of pathways involved in cell stress response and energy balance in the liver. Furthermore, it decreases the activity of the cell types implicated in liver inflammation and fibrosis in vitro. These findings highlight the therapeutic potential of partial protein synthesis inhibition in the treatment of NAFLD.


Subject(s)
Depsipeptides/pharmacology , Diet, Western , Liver Cirrhosis/prevention & control , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Peptide Elongation Factor 1/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Animals , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Energy Metabolism/drug effects , Hep G2 Cells , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation Mediators/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, 129 Strain , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/etiology , Obesity/metabolism , Peptide Elongation Factor 1/metabolism , Signal Transduction , THP-1 Cells , Triglycerides/blood
5.
J Nutr Biochem ; 70: 65-74, 2019 08.
Article in English | MEDLINE | ID: mdl-31176988

ABSTRACT

Vitamin D appears to either promote or inhibit neovascularization in a disease context-dependent manner. The effects of vitamin D, alone or in combination with niacin, on endothelial cell (EC) angiogenic function and on revascularization in obese animals with peripheral ischemia are unknown. Here, we report that supplementation of high palmitate medium with vitamin D, niacin or both vitamins increased EC tube formation, which relies primarily on cell migration, and also maintained tube stability over time. Transcriptomic analyses revealed that both vitamins increased stress response and anti-inflammatory gene expression. However, vitamin D decreased cell cycle gene expression and inhibited proliferation, while niacin induced stable expression of miR-126-3p and -5p and maintained cell proliferation in high palmitate. To assess vascular regeneration, diet-induced obese mice received vitamin D, niacin or both vitamins following hind limb ischemic injury. Niacin, but not vitamin D or combined treatment, improved recovery of hind limb use. Histology of tibialis anterior sections revealed no improvements in revascularization, regeneration, inflammation or fibrosis with vitamin D or combined treatment. In summary, although both vitamin D and niacin increased angiogenic function of EC cultures in high fat, only niacin improved recovery of hind limb use following ischemic injury in obese mice. It is possible that inhibition of cell proliferation by vitamin D in high-fat conditions limits vascular regeneration and recovery from peripheral ischemia in obesity.


Subject(s)
Diet , Ischemia/pathology , Neovascularization, Physiologic/drug effects , Niacin/pharmacology , Veins/pathology , Vitamin D/pharmacology , Animals , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Gene Expression Profiling , Hindlimb/blood supply , Inflammation , Male , Metabolic Syndrome/pathology , Mice , Mice, Obese , Microcirculation , Neovascularization, Pathologic , Palmitic Acid/pharmacology , Regeneration , Transcriptome
6.
Atherosclerosis ; 286: 60-70, 2019 07.
Article in English | MEDLINE | ID: mdl-31102954

ABSTRACT

BACKGROUND AND AIMS: Naringenin is a citrus-derived flavonoid with lipid-lowering and insulin-sensitizing effects leading to athero-protection in Ldlr-/- mice fed a high-fat diet. However, the ability of naringenin to promote atherosclerosis regression is unknown. In the present study, we assessed the capacity of naringenin to enhance regression in Ldlr-/- mice with diet-induced intermediate atherosclerosis intervened with a chow diet. METHODS: Male Ldlr-/- mice were fed a high-fat, cholesterol-containing (HFHC) diet for 12 weeks to induce intermediate atherosclerosis and metabolic dysfunction. Subsequently, a group of these mice were sacrificed for baseline analyses and the remainder either 1) continued on the HFHC diet, 2) switched to a chow diet or 3) switched to chow diet supplemented with naringenin. RESULTS: After 12 weeks induction, intermediate lesions developed in the aortic sinus. Intervention with chow alone slowed lesion growth, while intervention with naringenin-supplemented chow completely halted lesion growth. Lesions were characterized by features of improved morphology. Compared to chow alone, naringenin reduced plaque macrophages and modestly increased smooth muscle cells. Investigating processes that contributed to improved plaque morphology, we showed naringenin further reduced plasma triglycerides and cholesterol compared to chow alone. Furthermore, elevated monocytosis and myelopoiesis were further corrected by intervention with naringenin compared to chow alone. Metabolically, naringenin enhanced the correction of insulin resistance, hepatic steatosis and obesity compared to chow alone, potentially contributing to enhanced regression. CONCLUSIONS: Naringenin supplementation to chow enhances atherosclerosis regression in male Ldlr-/- mice. These studies further underscore the potential therapeutic utility of naringenin.


Subject(s)
Atherosclerosis/drug therapy , Flavanones/therapeutic use , Animals , Atherosclerosis/etiology , Diet, High-Fat , Male , Mice , Remission Induction
7.
Mol Nutr Food Res ; 63(6): e1800833, 2019 03.
Article in English | MEDLINE | ID: mdl-30578663

ABSTRACT

SCOPE: Naringenin is a citrus-derived flavonoid that has potent lipid-lowering and insulin-sensitizing effects in obese mouse models of metabolic dysfunction. However, in these models, a significant effect of naringenin supplementation is the prevention of weight gain, which in itself can confer metabolic protection. Therefore, in the present study, the effect of naringenin supplementation in lean, chow-fed Ldlr-/- mice is investigated. METHODS AND RESULTS: In Ldlr-/- mice with isocaloric food consumption, treatment with naringenin for 8 weeks reduces body weight and adiposity compared to littermate controls pair-fed the chow diet alone. Furthermore, naringenin treatment reduces plasma lipids and enhances insulin sensitivity compared to chow-fed controls. Metabolic cage studies reveal that naringenin-treated mice have elevated energy expenditure with no change in ambulatory activity. Additionally, naringenin-treated mice have an increased respiratory exchange ratio and food consumption during the dark cycle. Treatment increases the expression of fatty acid oxidation genes in liver, and increased ß-hydroxybutyrate concentrations in plasma, indicating that one mechanism through which naringenin mediates metabolic improvement is enhanced hepatic fatty acid oxidation. CONCLUSIONS: These studies highlight the potential therapeutic utility of naringenin and suggest that this flavonoid maintains potent metabolic properties in the absence of obesity or a high-fat diet.


Subject(s)
Adiposity/drug effects , Energy Metabolism/drug effects , Fatty Acids/metabolism , Flavanones/pharmacology , Adiposity/physiology , Animals , Dietary Supplements , Insulin/blood , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Oxidation-Reduction , Receptors, LDL/genetics
8.
J Lipid Res ; 59(9): 1714-1728, 2018 09.
Article in English | MEDLINE | ID: mdl-30008441

ABSTRACT

Obesity and its associated metabolic dysfunction and cardiovascular disease risk represent a leading cause of adult morbidity worldwide. Currently available pharmacological therapies for obesity have had limited success in reversing existing obesity and metabolic dysregulation. Previous prevention studies demonstrated that the citrus flavonoids, naringenin and nobiletin, protect against obesity and metabolic dysfunction in Ldlr-/- mice fed a high-fat cholesterol-containing (HFHC) diet. However, their effects in an intervention model are unknown. In this report, we show that, in Ldlr-/- mice with diet-induced obesity, citrus flavonoid supplementation to a HFHC diet reversed existing obesity and adipocyte size and number through enhanced energy expenditure and increased hepatic fatty acid oxidation. Caloric intake was unaffected and no evidence of white adipose tissue browning was observed. Reversal of adiposity was accompanied by improvements in hyperlipidemia, insulin sensitivity, hepatic steatosis, and a modest reduction in blood monocytes. Together, this resulted in atherosclerotic lesions that were unchanged in size, but characterized by reduced macrophage content, consistent with a more stable plaque phenotype. These studies further suggest potential therapeutic utility of citrus flavonoids, especially in the context of existing obesity, metabolic dysfunction, and cardiovascular disease.


Subject(s)
Atherosclerosis/complications , Citrus/chemistry , Flavonoids/pharmacology , Metabolic Syndrome/complications , Obesity/complications , Obesity/drug therapy , Receptors, LDL/deficiency , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Body Weight/drug effects , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Flavonoids/therapeutic use , Hyperlipidemias/complications , Insulin Resistance , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Obesity/metabolism , Obesity/pathology
9.
Arterioscler Thromb Vasc Biol ; 38(5): 1178-1190, 2018 05.
Article in English | MEDLINE | ID: mdl-29449335

ABSTRACT

OBJECTIVE: Bempedoic acid (BemA; ETC-1002) is a novel drug that targets hepatic ATP-citrate lyase to reduce cholesterol biosynthesis. In phase 2 studies, BemA lowers elevated low-density lipoprotein cholesterol (LDL-C) in hypercholesterolemic patients. In the present study, we tested the ability of BemA to decrease plasma cholesterol and LDL-C and attenuate atherosclerosis in a large animal model of familial hypercholesterolemia. APPROACH AND RESULTS: Gene targeting has been used to generate Yucatan miniature pigs heterozygous (LDLR+/-) or homozygous (LDLR-/-) for LDL receptor deficiency (ExeGen). LDLR+/- and LDLR-/- pigs were fed a high-fat, cholesterol-containing diet (34% kcal fat; 0.2% cholesterol) and orally administered placebo or BemA for 160 days. In LDLR+/- pigs, compared with placebo, BemA decreased plasma cholesterol and LDL-C up to 40% and 61%, respectively. In LDLR-/- pigs, in which plasma cholesterol and LDL-C were 5-fold higher than in LDLR+/- pigs, BemA decreased plasma cholesterol and LDL-C up to 27% and 29%, respectively. Plasma levels of triglycerides and high-density lipoprotein cholesterol, fasting glucose and insulin, and liver lipids were unaffected by treatment in either genotype. In the aorta of LDLR+/- pigs, BemA robustly attenuated en face raised lesion area (-58%) and left anterior descending coronary artery cross-sectional lesion area (-40%). In LDLR-/- pigs, in which lesions were substantially more advanced, BemA decreased aortic lesion area (-47%) and left anterior descending coronary artery lesion area (-48%). CONCLUSIONS: In a large animal model of LDLR deficiency and atherosclerosis, long-term treatment with BemA reduces LDL-C and attenuates the development of aortic and coronary atherosclerosis in both LDLR+/- and LDLR-/- miniature pigs.


Subject(s)
Anticholesteremic Agents/pharmacology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cholesterol, LDL/blood , Coronary Artery Disease/prevention & control , Dicarboxylic Acids/pharmacology , Fatty Acids/pharmacology , Hyperlipoproteinemia Type II/drug therapy , Receptors, LDL/deficiency , Animals , Animals, Genetically Modified , Anticholesteremic Agents/pharmacokinetics , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Dicarboxylic Acids/pharmacokinetics , Disease Models, Animal , Down-Regulation , Fatty Acids/pharmacokinetics , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/genetics , Male , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/genetics , Swine , Swine, Miniature
10.
Arterioscler Thromb Vasc Biol ; 37(4): 647-656, 2017 04.
Article in English | MEDLINE | ID: mdl-28153881

ABSTRACT

OBJECTIVE: Bempedoic acid (ETC-1002, 8-hydroxy-2,2,14,14-tetramethylpentadecanedioic acid) is a novel low-density lipoprotein cholesterol-lowering compound. In animals, bempedoic acid targets the liver where it inhibits cholesterol and fatty acid synthesis through inhibition of ATP-citrate lyase and through activation of AMP-activated protein kinase. In this study, we tested the hypothesis that bempedoic acid would prevent diet-induced metabolic dysregulation, inflammation, and atherosclerosis. APPROACH AND RESULTS: Ldlr-/- mice were fed a high-fat, high-cholesterol diet (42% kcal fat, 0.2% cholesterol) supplemented with bempedoic acid at 0, 3, 10 and 30 mg/kg body weight/day. Treatment for 12 weeks dose-dependently attenuated diet-induced hypercholesterolemia, hypertriglyceridemia, hyperglycemia, hyperinsulinemia, fatty liver and obesity. Compared to high-fat, high-cholesterol alone, the addition of bempedoic acid decreased plasma triglyceride (up to 64%) and cholesterol (up to 50%) concentrations, and improved glucose tolerance. Adiposity was significantly reduced with treatment. In liver, bempedoic acid prevented cholesterol and triglyceride accumulation, which was associated with increased fatty acid oxidation and reduced fatty acid synthesis. Hepatic gene expression analysis revealed that treatment significantly increased expression of genes involved in fatty acid oxidation while suppressing inflammatory gene expression. In full-length aorta, bempedoic acid markedly suppressed cholesteryl ester accumulation, attenuated the expression of proinflammatory M1 genes and attenuated the iNos/Arg1 ratio. Treatment robustly attenuated atherosclerotic lesion development in the aortic sinus by 44%, with beneficial changes in morphology, characteristic of earlier-stage lesions. CONCLUSIONS: Bempedoic acid effectively prevents plasma and tissue lipid elevations and attenuates the onset of inflammation, leading to the prevention of atherosclerotic lesion development in a mouse model of metabolic dysregulation.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , Atherosclerosis/prevention & control , Dicarboxylic Acids/pharmacology , Diet, High-Fat , Dyslipidemias/prevention & control , Enzyme Inhibitors/pharmacology , Fatty Acids/pharmacology , Inflammation/prevention & control , Liver/drug effects , Obesity/prevention & control , Receptors, LDL/deficiency , ATP Citrate (pro-S)-Lyase/metabolism , Animals , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/genetics , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Disease Models, Animal , Dyslipidemias/blood , Dyslipidemias/enzymology , Dyslipidemias/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Inflammation/blood , Inflammation/enzymology , Inflammation/genetics , Inflammation Mediators/blood , Insulin/blood , Lipids/blood , Liver/enzymology , Male , Mice, Knockout , Obesity/blood , Obesity/enzymology , Obesity/genetics , Phenotype , Receptors, LDL/genetics , Time Factors
11.
Data Brief ; 10: 385-389, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28050580

ABSTRACT

As nonalcoholic fatty liver disease progresses to end-stage diseases, including fibrosis, cirrhosis and hepatocellular carcinoma, fibrotic activated hepatic stellate cells and cancerous epithelial cells can become abundant, changing the cellular composition of this organ. Despite potentially residing within the same diseased tissue, direct comparisons of global gene expression between activated hepatic stellate cells and hepatocellular carcinoma cells are lacking. Here we provide data collected using Affymetrix GeneChip microarrays to identify differential gene expression in cultured primary human activated hepatic stellate cells compared to HepG2 human hepatoma cells. The dataset includes many genes involved in intermediary metabolism which were investigated in greater depth in our associated article (A.M. Hetherington, C.G. Sawyez, E. Zilberman, A.M. Stoianov, D.L. Robson, J.M. Hughes-Large, et al., 2016) [1]. Pathway analyses of known protein coding genes down-regulated or up-regulated by greater than 2.0-fold are also provided.

12.
J Mol Med (Berl) ; 95(4): 431-444, 2017 04.
Article in English | MEDLINE | ID: mdl-28070626

ABSTRACT

Joint homeostasis failure can result in osteoarthritis (OA). Currently, there are no treatments to alter disease progression in OA, but targeting early changes in cellular behavior has great potential. Recent data show that nuclear receptors contribute to the pathogenesis of OA and could be viable therapeutic targets, but their molecular mechanisms in cartilage are incompletely understood. This study examines global changes in gene expression after treatment with agonists for four nuclear receptor implicated in OA (LXR, PPARδ, PPARγ, and RXR). Murine articular chondrocytes were treated with agonists for LXR, PPARδ, PPARγ, or RXR and underwent microarray, qPCR, and cellular lipid analyses to evaluate changes in gene expression and lipid profile. Immunohistochemistry was conducted to compare two differentially expressed targets (Txnip, Gsta4) in control and cartilage-specific PPARδ knockout mice subjected to surgical destabilization of the medial meniscus (DMM). Nuclear receptor agonists induced different gene expression profiles with many responses affecting lipid metabolism. LXR activation downregulated gene expression of proteases involved in OA, whereas RXR agonism decreased expression of ECM components and increased expression of Mmp13. Functional assays indicate increases in cell triglyceride accumulation after PPARγ, LXR, and RXR agonism but a decrease after PPARδ agonism. PPARδ and RXR downregulate the antioxidant Gsta4, and PPARδ upregulates Txnip. Wild-type, but not PPARδ-deficient mice, display increased staining for Txnip after DMM. Collectively, these data demonstrate that nuclear receptor activation in chondrocytes primarily affects lipid metabolism. In the case of PPARδ, this change might lead to increased oxidative stress, possibly contributing to OA-associated changes. KEY MESSAGE: Nuclear receptors regulate metabolic genes in chondrocytes. Nuclear receptors affect triglyceride levels. PPARδ mediates regulation of oxidative stress markers. Nuclear receptors are promising therapeutic targets for osteoarthritis.


Subject(s)
Chondrocytes/metabolism , Lipid Metabolism , Osteoarthritis/metabolism , Oxidative Stress , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/pathology , Gene Expression Regulation , Liver X Receptors/metabolism , Male , Mice, Inbred C57BL , Osteoarthritis/genetics , Osteoarthritis/pathology , PPAR delta/metabolism , PPAR gamma/metabolism , Retinoid X Receptors/metabolism
13.
Cell Physiol Biochem ; 39(4): 1648-62, 2016.
Article in English | MEDLINE | ID: mdl-27626926

ABSTRACT

BACKGROUND/AIMS: Nonalcoholic fatty liver disease (NAFLD) progression to fibrosis, cirrhosis and hepatocellular carcinoma, alters the cellular composition of this organ. During late-stage NAFLD, fibrotic and possibly cancerous cells can proliferate and, like normal hepatocytes, are exposed to high concentrations of fatty acids from both surrounding tissue and circulating lipid sources. We hypothesized that primary human activated hepatic stellate cells and epithelial hepatoma (HepG2) cells respond differently to lipotoxic conditions, and investigated the mechanisms involved. METHODS: Primary activated hepatic stellate cells and HepG2 cells were exposed to pathophysiological concentrations of fatty acids and comparative studies of lipid metabolic and stress response pathways were performed. RESULTS: Both cell types remained proliferative during exposure to a combination of palmitate plus oleate reflective of the general saturated versus unsaturated fatty acid composition of western diets. However, exposure to either high palmitate or high oleate alone induced cytotoxicity in activated stellate cells, while only palmitate caused cytotoxicity in HepG2 cells. mRNA microarray and biochemical comparisons revealed that stellate cells stored markedly less fatty acids as neutral lipids, and had reduced capacity for beta-oxidation. Similar to previous observations in HepG2 cells, palmitate, but not oleate, induced ER stress and actin stress fiber formation in activated stellate cells. In contrast, oleate, but not palmitate, induced the inflammatory signal TXNIP, decreased cytoskeleton proteins, and decreased cell polarity preceding cell death in activated stellate cells. CONCLUSIONS: Palmitate-induced lipotoxicity was associated with ER stress pathways in both primary activated hepatic stellate cells and epithelial hepatoma cells, whereas high oleate caused lipotoxicity only in activated stellate cells, possibly through a distinct mechanism involving disruption of cytoskeleton components. This may have implications for optimal dietary fatty acid compositions during various stages of NAFLD.


Subject(s)
Hepatic Stellate Cells/drug effects , Lipid Metabolism/drug effects , Oleic Acid/toxicity , Palmitic Acid/toxicity , Stress, Physiological/drug effects , Transcriptome , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Polarity/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Lipid Metabolism/genetics , Oligonucleotide Array Sequence Analysis , Organ Specificity , Oxidation-Reduction , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological/genetics
14.
Physiol Rep ; 4(17)2016 09.
Article in English | MEDLINE | ID: mdl-27613825

ABSTRACT

Eukaryotic elongation factor EEF1A1 is induced by oxidative and ER stress, and contributes to subsequent cell death in many cell types, including hepatocytes. We recently showed that blocking the protein synthesis activity of EEF1A1 with the peptide inhibitor, didemnin B, decreases saturated fatty acid overload-induced cell death in HepG2 cells. In light of this and other recent work suggesting that limiting protein synthesis may be beneficial in treating ER stress-related disease, we hypothesized that acute intervention with didemnin B would decrease hepatic ER stress and lipotoxicity in obese mice with nonalcoholic fatty liver disease (NAFLD). Hyperphagic male ob/ob mice were fed semipurified diet for 4 weeks, and during week 5 received i.p. injections of didemnin B or vehicle on days 1, 4, and 7. Interestingly, we observed that administration of this compound modestly decreased food intake without evidence of illness or distress, and thus included an additional control group matched for food consumption with didemnin B-treated animals. Treatment with didemnin B improved several characteristics of hepatic lipotoxicity to a greater extent than the effects of caloric restriction alone, including hepatic steatosis, and some hepatic markers of ER stress and inflammation (GRP78, Xbp1s, and Mcp1). Plasma lipid and lipoprotein profiles and histopathological measures of NAFLD, including lobular inflammation, and total NAFLD activity score were also improved by didemnin B. These data indicate that acute intervention with the EEF1A inhibitor, didemnin B, improves hepatic lipotoxicity in obese mice with NAFLD through mechanisms not entirely dependent on decreased food intake, suggesting a potential therapeutic strategy for this ER stress-related disease.


Subject(s)
Depsipeptides/pharmacology , Hep G2 Cells/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Immunosuppressive Agents/pharmacology , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Peptide Elongation Factor 1/antagonists & inhibitors , Peptide Elongation Factor 1/metabolism , Animals , Cell Death , Depsipeptides/administration & dosage , Depsipeptides/metabolism , Eating/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/metabolism , Gene Expression , Heat-Shock Proteins/metabolism , Hep G2 Cells/metabolism , Hep G2 Cells/ultrastructure , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/metabolism , Injections, Intraperitoneal , Lipid Metabolism/drug effects , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology
15.
Pharmacol Res Perspect ; 4(3): e00233, 2016 06.
Article in English | MEDLINE | ID: mdl-27433343

ABSTRACT

Niacin can reduce vascular disease risk in individuals with metabolic syndrome, but in light of recent large randomized controlled trials outcomes, its biological actions and clinical utility remain controversial. Niacin can improve endothelial function, vascular inflammation, and vascular regeneration, independent of correcting dyslipidemia, in various lean rodent models of vascular injury. Here, we tested whether niacin could directly improve endothelial cell angiogenic function during combined exposure to excess fatty acids and hypoxia, and whether intervention with niacin during continued feeding of western diet could improve revascularization and functional recovery in obese, hyperlipidemic mice with peripheral ischemia. Treatment with niacin (10 µmol/L) increased human microvascular endothelial cell angiogenic function during exposure to high fatty acids and hypoxia (2% oxygen), as determined by tube formation on Matrigel. To assess revascularization in vivo, we used western diet-induced obese mice with unilateral hind limb femoral artery ligation and excision. Treatment for 14 days postinjury with once daily i.p. injections of a low dose of niacin (50 mg/kg) improved recovery of hind limb use, in association with enhanced revascularization and decreased inflammation of the tibialis anterior muscle. These effects were concomitant with decreased plasma triglycerides, but not increased plasma apoAI. Thus, niacin improves endothelial tube formation under lipotoxic and hypoxic conditions, and moreover, promotes revascularization and functional hind limb recovery following ischemic injury in diet-induced obese mice with hyperlipidemia. These data may have implications for niacin therapy in the treatment of peripheral ischemic vascular disease associated with metabolic syndrome.

16.
PLoS One ; 10(6): e0131269, 2015.
Article in English | MEDLINE | ID: mdl-26102086

ABSTRACT

Elongation factor 1A-1 (eEF1A-1) has non-canonical functions in regulation of the actin cytoskeleton and apoptosis. It was previously identified through a promoter-trap screen as a mediator of fatty acid-induced cell death (lipotoxicity), and was found to participate in this process downstream of ER stress. Since ER stress is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), we investigated the mechanism of action of eEF1A-1 in hepatocyte lipotoxicity. HepG2 cells were exposed to excess fatty acids, followed by assessments of ER stress, subcellular localization of eEF1A-1, and cell death. A specific inhibitor of eEF1A-1 elongation activity, didemnin B, was used to determine whether its function in protein synthesis is involved in lipotoxicity. Within 6 h, eEF1A-1 protein was modestly induced by high palmitate, and partially re-localized from its predominant location at the ER to polymerized actin at the cell periphery. This early induction and subcellular redistribution of eEF1A-1 coincided with the onset of ER stress, and was later followed by cell death. Didemnin B did not prevent the initiation of ER stress by high palmitate, as indicated by eIF2α phosphorylation. However, consistent with sustained inhibition of eEF1A-1-dependent elongation activity, didemnin B prevented the recovery of protein synthesis and increase in GRP78 protein that are normally associated with later phases of the response to ongoing ER stress. This resulted in decreased palmitate-induced cell death. Our data implicate eEF1A-1, and its function in protein synthesis, in hepatocyte lipotoxicity.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hepatocytes/drug effects , Palmitates/toxicity , Peptide Chain Elongation, Translational , Peptide Elongation Factor 1/physiology , Animals , Apoptosis/drug effects , Depsipeptides/pharmacology , Dietary Fats/toxicity , Dietary Sucrose/toxicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Hep G2 Cells , Hepatocytes/metabolism , Humans , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Animal , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Protein Transport
17.
Endocrinology ; 156(6): 2087-102, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25774553

ABSTRACT

The molecular mechanisms and metabolic pathways whereby the citrus flavonoid, naringenin, reduces dyslipidemia and improves glucose tolerance were investigated in C57BL6/J wild-type mice and fibroblast growth factor 21 (FGF21) null (Fgf21(-/-)) mice. FGF21 regulates energy homeostasis and the metabolic adaptation to fasting. One avenue of this regulation is through induction of peroxisome proliferator-activated receptor-γ coactivator-1α (Pgc1a), a regulator of hepatic fatty acid oxidation and ketogenesis. Because naringenin is a potent activator of hepatic FA oxidation, we hypothesized that induction of FGF21 might be an integral part of naringenin's mechanism of action. Furthermore, we predicted that FGF21 deficiency would potentiate high-fat diet (HFD)-induced metabolic dysregulation and compromise metabolic protection by naringenin. The absence of FGF21 exacerbated the response to a HFD. Interestingly, naringenin supplementation to the HFD robustly prevented obesity in both genotypes. Gene expression analysis suggested that naringenin was not primarily targeting fatty acid metabolism in white adipose tissue. Naringenin corrected hepatic triglyceride concentrations and normalized hepatic expression of Pgc1a, Cpt1a, and Srebf1c in both wild-type and Fgf21(-/-) mice. HFD-fed Fgf21(-/-) mice displayed greater muscle triglyceride deposition, hyperinsulinemia, and impaired glucose tolerance as compared with wild-type mice, confirming the role of FGF21 in insulin sensitivity; however, naringenin supplementation improved these metabolic parameters in both genotypes. We conclude that FGF21 deficiency exacerbates HFD-induced obesity, hepatic steatosis, and insulin resistance. Furthermore, FGF21 is not required for naringenin to protect mice from HFD-induced metabolic dysregulation. Collectively these studies support the concept that naringenin has potent lipid-lowering effects and may act as an insulin sensitizer in vivo.


Subject(s)
Fatty Liver/prevention & control , Fibroblast Growth Factors/metabolism , Flavanones/therapeutic use , Glucose Intolerance/prevention & control , Obesity/prevention & control , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Fatty Liver/genetics , Fatty Liver/metabolism , Fibroblast Growth Factors/genetics , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism
18.
J Lipid Res ; 55(7): 1254-66, 2014 07.
Article in English | MEDLINE | ID: mdl-24864274

ABSTRACT

PPARδ regulates systemic lipid homeostasis and inflammation, but its role in hepatic lipid metabolism remains unclear. Here, we examine whether intervening with a selective PPARδ agonist corrects hepatic steatosis induced by a high-fat, cholesterol-containing (HFHC) diet. Ldlr(-/-) mice were fed a chow or HFHC diet (42% fat, 0.2% cholesterol) for 4 weeks. For an additional 8 weeks, the HFHC group was fed HFHC or HFHC plus GW1516 (3 mg/kg/day). GW1516-intervention significantly attenuated liver TG accumulation by induction of FA ß-oxidation and attenuation of FA synthesis. In primary mouse hepatocytes, GW1516 treatment stimulated AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) phosphorylation in WT hepatocytes, but not AMPKß1(-/-) hepatocytes. However, FA oxidation was only partially reduced in AMPKß1(-/-) hepatocytes, suggesting an AMPK-independent contribution to the GW1516 effect. Similarly, PPARδ-mediated attenuation of FA synthesis was partially due to AMPK activation, as GW1516 reduced lipogenesis in WT hepatocytes but not AMPKß1(-/-) hepatocytes. HFHC-fed animals were hyperinsulinemic and exhibited selective hepatic insulin resistance, which contributed to elevated fasting FA synthesis and hyperglycemia. GW1516 intervention normalized fasting hyperinsulinemia and selective hepatic insulin resistance and attenuated fasting FA synthesis and hyperglycemia. The HFHC diet polarized the liver toward a proinflammatory M1 state, which was reversed by GW1516 intervention. Thus, PPARδ agonist treatment inhibits the progression of preestablished hepatic steatosis.


Subject(s)
Dietary Fats/adverse effects , Fatty Acids/biosynthesis , Fatty Liver/metabolism , Insulin Resistance , Lipogenesis/drug effects , PPAR delta/metabolism , Receptors, LDL/metabolism , Animals , Dietary Fats/pharmacology , Fatty Acids/genetics , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Lipogenesis/genetics , Mice , Mice, Knockout , Oxidation-Reduction/drug effects , PPAR delta/genetics , Receptors, LDL/genetics
19.
Arterioscler Thromb Vasc Biol ; 34(1): 52-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24158519

ABSTRACT

OBJECTIVE: The peroxisome proliferator-activated receptor (PPAR) δ regulates systemic lipid homeostasis and inflammation. However, the ability of PPARδ agonists to improve the pathology of pre-established lesions and whether PPARδ activation is atheroprotective in the setting of insulin resistance have not been reported. Here, we examine whether intervention with a selective PPARδ agonist corrects metabolic dysregulation and attenuates aortic inflammation and atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor knockout mice were fed a chow or a high-fat, high-cholesterol (HFHC) diet (42% fat, 0.2% cholesterol) for 4 weeks. For a further 8 weeks, the HFHC group was fed either HFHC or HFHC plus GW1516 (3 mg/kg per day). GW1516 significantly attenuated pre-established fasting hyperlipidemia, hyperglycemia, and hyperinsulinemia, as well as glucose and insulin intolerance. GW1516 intervention markedly reduced aortic sinus lesions and lesion macrophages, whereas smooth muscle α-actin was unchanged and collagen deposition enhanced. In aortae, GW1516 increased the expression of the PPARδ-specific gene Adfp but not PPARα- or γ-specific genes. GW1516 intervention decreased the expression of aortic proinflammatory M1 cytokines, increased the expression of the anti-inflammatory M2 cytokine Arg1, and attenuated the iNos/Arg1 ratio. Enhanced mitogen-activated protein kinase signaling, known to induce inflammatory cytokine expression in vitro, was enhanced in aortae of HFHC-fed mice. Furthermore, the HFHC diet impaired aortic insulin signaling through Akt and forkhead box O1, which was associated with elevated endoplasmic reticulum stress markers CCAAT-enhancer-binding protein homologous protein and 78kDa glucose regulated protein. GW1516 intervention normalized mitogen-activated protein kinase activation, insulin signaling, and endoplasmic reticulum stress. CONCLUSIONS: Intervention with a PPARδ agonist inhibits aortic inflammation and attenuates the progression of pre-established atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortitis/prevention & control , Atherosclerosis/prevention & control , Insulin Resistance , PPAR delta/agonists , Receptors, LDL/deficiency , Thiazoles/pharmacology , Animals , Aortitis/blood , Aortitis/etiology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Blood Glucose/metabolism , Cholesterol, Dietary , Diet, High-Fat , Disease Models, Animal , Dyslipidemias/blood , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Dyslipidemias/metabolism , Inflammation Mediators/metabolism , Insulin/blood , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , Time Factors
20.
J Lipid Res ; 54(3): 711-724, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269394

ABSTRACT

Obesity-associated chronic inflammation contributes to metabolic dysfunction and propagates atherosclerosis. Recent evidence suggests that increased dietary cholesterol exacerbates inflammation in adipose tissue and liver, contributing to the proatherogenic milieu. The ability of the citrus flavonoid naringenin to prevent these cholesterol-induced perturbations is unknown. To assess the ability of naringenin to prevent the amplified inflammatory response and atherosclerosis induced by dietary cholesterol, male Ldlr⁻/⁻ mice were fed either a cholesterol-enriched high-fat or low-fat diet supplemented with 3% naringenin for 12 weeks. Naringenin, through induction of hepatic fatty acid (FA) oxidation and attenuation of FA synthesis, prevented hepatic steatosis, hepatic VLDL overproduction, and hyperlipidemia induced by both cholesterol-rich diets. Naringenin attenuated hepatic macrophage infiltration and inflammation stimulated by dietary cholesterol. Insulin resistance, adipose tissue expansion, and inflammation were alleviated by naringenin. Naringenin attenuated the cholesterol-induced formation of both foam cells and expression of inflammatory markers in peritoneal macrophages. Naringenin significantly decreased atherosclerosis and inhibited the formation of complex lesions, which was associated with normalized aortic lipids and a reversal of aortic inflammation. We demonstrate that in mice fed cholesterol-enriched diets, naringenin attenuates peripheral and systemic inflammation, leading to protection from atherosclerosis. These studies offer a therapeutically relevant alternative for the prevention of cholesterol-induced metabolic dysregulation.


Subject(s)
Atherosclerosis/chemically induced , Atherosclerosis/prevention & control , Cholesterol/adverse effects , Flavanones/therapeutic use , Inflammation/chemically induced , Inflammation/prevention & control , Animals , Fatty Liver/chemically induced , Fatty Liver/prevention & control , Flavonoids/metabolism , Insulin Resistance , Male , Mice , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...