Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Metabolism ; 64(9): 982-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26024755

ABSTRACT

OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD) is a common disorder characterized by excessive hepatic fat accumulation, production of reactive oxygen species (ROS), inflammation and potentially resulting in non-alcoholic steatohepatitis (NASH), cirrhosis and end-stage liver disease. Recently, we have shown that niacin significantly prevented hepatic steatosis and regressed pre-existing steatosis in high-fat fed rat model of NAFLD. To gain further insight into the cellular mechanisms, this study investigated the effect of niacin on human hepatocyte fat accumulation, ROS production, and inflammatory mediator IL-8 secretion. MATERIALS AND METHODS: Human hepatoblastoma cell line HepG2 or human primary hepatocytes were first stimulated with palmitic acid followed by treatment with niacin or control for 24 h. RESULTS: The data indicated that niacin (at 0.25 and 0.5 mmol/L doses) significantly inhibited palmitic acid-induced fat accumulation in human hepatocytes by 45-62%. This effect was associated with inhibition of diacylglycerol acyltransferase 2 (DGAT2) mRNA expression without affecting the mRNA expression of fatty acid synthase (FAS) and carnitine palmitoyltransferase 1 (CPT1). Niacin attenuated hepatocyte ROS production and it also inhibited NADPH oxidase activity. Niacin reduced palmitic acid-induced IL-8 levels. CONCLUSIONS: These findings suggest that niacin, through inhibiting hepatocyte DGAT2 and NADPH oxidase activity, attenuates hepatic fat accumulation and ROS production respectively. Decreased ROS production, at least in part, may have contributed to the inhibition of pro-inflammatory IL-8 levels. These mechanistic studies may be useful for the clinical development of niacin and niacin-related compounds for the treatment of NAFLD/NASH and its complications.


Subject(s)
Hypolipidemic Agents/pharmacology , Interleukin-8/metabolism , Lipid Metabolism/drug effects , Niacin/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress/drug effects , Diacylglycerol O-Acyltransferase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Palmitic Acid/pharmacology , Primary Cell Culture , Reactive Oxygen Species/metabolism
2.
Atherosclerosis ; 235(2): 554-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24956528

ABSTRACT

OBJECTIVES: Leukocyte myeloperoxidase (MPO) is a major player in the pathogenesis of various chronic diseases including atherosclerosis. This study proposes the novel concept that niacin, through reactive oxygen species (ROS)-mediated signaling, decreases neutrophil MPO release and its activity, protects apolipoprotein-AI (apo-AI) modification and improves HDL function. METHODS: Human blood leukocytes and leukocytic cell line HL-60 cells were treated with niacin, and stimulated with phorbol myristate acetate (PMA). Cellular and released MPO activity in the medium was measured by assessing chlorination of MPO-specific substrate. MPO protein release in the medium and apo-AI degradation was measured by Western blot analysis. Monocyte adhesion to human aortic primary endothelial cells was measured to assess biological function of HDL/apo-AI. RESULTS: PMA significantly increased leukocyte MPO activity in both intracellular extract and medium. Niacin (0.25-0.5 mM) decreased PMA-induced MPO activity (cellular and released in the media). Niacin also decreased MPO protein mass in the medium without affecting its mRNA expression. Increased NADPH oxidase and ROS production by PMA were also significantly inhibited by niacin. Studies with specific inhibitors suggest that ROS-dependent Src and p38MAP kinase mediate decreased MPO activity by niacin. Niacin blocked apo-AI degradation, and apo-AI from niacin treated cells decreased monocyte adhesion to aortic endothelial cells. CONCLUSIONS: These findings identify niacin as a potent inhibitor of leukocyte MPO release and MPO-mediated formation of dysfunctional HDL. Niacin and niacin-related chemical entities may form important therapeutic agents for MPO-mediated inflammatory diseases.


Subject(s)
Niacin/pharmacology , Peroxidase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism , Apolipoprotein A-I/metabolism , HL-60 Cells , Humans , Leukocytes/drug effects , Leukocytes/enzymology , NADPH Oxidases/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Tetradecanoylphorbol Acetate/pharmacology
3.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G320-7, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24356885

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), a leading cause of liver damage, comprises a spectrum of liver abnormalities including the early fat deposition in the liver (hepatic steatosis) and advanced nonalcoholic steatohepatitis. Niacin decreases plasma triglycerides, but its effect on hepatic steatosis is elusive. To examine the effect of niacin on steatosis, rats were fed either a rodent normal chow, chow containing high fat (HF), or HF containing 0.5% or 1.0% niacin in the diet for 4 wk. For regression studies, rats were first fed the HF diet for 6 wk to induce hepatic steatosis and were then treated with niacin (0.5% in the diet) while on the HF diet for 6 wk. The findings indicated that inclusion of niacin at 0.5% and 1.0% doses in the HF diet significantly decreased liver fat content, liver weight, hepatic oxidative products, and prevented hepatic steatosis. Niacin treatment to rats with preexisting hepatic steatosis induced by the HF diet significantly regressed steatosis. Niacin had no effect on the mRNA expression of fatty acid synthesis or oxidation genes (including sterol-regulatory element-binding protein 1, acetyl-CoA carboxylase 1, fatty acid synthase, and carnitine palmitoyltransferase 1) but significantly inhibited mRNA levels, protein expression, and activity of diacylglycerol acyltrasferase 2, a key enzyme in triglyceride synthesis. These novel findings suggest that niacin effectively prevents and causes the regression of experimental hepatic steatosis. Approved niacin formulation(s) for other indications or niacin analogs may offer a very cost-effective opportunity for the clinical development of niacin for treating NAFLD and fatty liver disease.


Subject(s)
Antioxidants/pharmacology , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Lipid Metabolism/drug effects , Liver/drug effects , Niacin/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Disease Progression , Fatty Acids, Nonesterified/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation, Enzymologic/drug effects , Inflammation Mediators/metabolism , Lipid Metabolism/genetics , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Non-alcoholic Fatty Liver Disease , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Curr Opin Lipidol ; 24(3): 239-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23619367

ABSTRACT

PURPOSE OF REVIEW: This review focuses on the current understanding of the physiological mechanisms of action of niacin on lipid metabolism and atherosclerosis. RECENT FINDINGS: Emerging findings indicate that niacin decreases hepatic triglyceride synthesis and subsequent VLDL/LDL secretion by directly and noncompetitively inhibiting hepatocyte diacylglycerol acyltransferase 2. Recent studies in mice lacking niacin receptor GPR109A and human clinical trials with GPR109A agonists disproved the long believed hypothesis of adipocyte triglyceride lipolysis as the mechanism for niacin's effect on serum lipids. Niacin, through inhibiting hepatocyte surface expression of ß-chain ATP synthase, inhibits the removal of HDL-apolipoprotein (apo) AI resulting in increased apoAI-containing HDL particles. Additional recent findings suggest that niacin by increasing hepatic ATP-binding cassette transporter A1-mediated apoAI lipidation increases HDL biogenesis, thus stabilizing circulation of newly secreted apoAI. New concepts have also emerged on lipid-independent actions of niacin on vascular endothelial oxidative and inflammatory events, myeloperoxidase release from neutrophils and its impact on HDL function, and GPR109A-mediated macrophage inflammatory events involved in atherosclerosis. SUMMARY: Recent advances have provided physiological mechanisms of action of niacin on lipid metabolism and atherosclerosis. Better understanding of niacin's actions on multiple tissues and targets may be helpful in designing combination therapy and new treatment strategies for atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Hypolipidemic Agents/therapeutic use , Lipid Metabolism/drug effects , Niacin/therapeutic use , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol, HDL/agonists , Cholesterol, HDL/metabolism , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/metabolism , Cholesterol, VLDL/antagonists & inhibitors , Cholesterol, VLDL/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Gene Expression Regulation/drug effects , Humans , Mice , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis
5.
J Lipid Res ; 53(5): 941-950, 2012 May.
Article in English | MEDLINE | ID: mdl-22389325

ABSTRACT

The lipidation of apoA-I in liver greatly influences HDL biogenesis and plasma HDL levels by stabilizing the secreted apoA-I. Niacin is the most effective lipid-regulating agent clinically available to raise HDL. This study was undertaken to identify regulatory mechanisms of niacin action in hepatic lipidation of apoA-I, a critical event involved in HDL biogenesis. In cultured human hepatocytes (HepG2), niacin increased: association of apoA-I with phospholipids and cholesterol by 46% and 23% respectively, formation of lipid-poor single apoA-I molecule-containing particles up to ~2.4-fold, and pre ß 1 and α migrating HDL particles. Niacin dose-dependently stimulated the cell efflux of phospholipid and cholesterol and increased transcription of ABCA1 gene and ABCA1 protein. Mutated DR4, a binding site for nuclear factor liver X receptor alpha (LXR α ) in the ABCA1 promoter, abolished niacin stimulatory effect. Further, knocking down LXR α or ABCA1 by RNA interference eliminated niacin-stimulated apoA-I lipidation. Niacin treatment did not change apoA-I gene expression. The present data indicate that niacin increases apoA-I lipidation by enhancing lipid efflux through a DR4-dependent transcription of ABCA1 gene in HepG2 cells. A stimulatory role of niacin in early hepatic formation of HDL particles suggests a new mechanism that contributes to niacin action to increase the stability of newly synthesized circulating HDL.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Apolipoprotein A-I/metabolism , Cholesterol, HDL/biosynthesis , Niacin/pharmacology , Repetitive Sequences, Nucleic Acid/genetics , Transcription, Genetic/drug effects , ATP Binding Cassette Transporter 1 , Biological Transport/drug effects , Culture Media/metabolism , Gene Expression Regulation/drug effects , Hep G2 Cells , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Movement/drug effects , Phospholipids/metabolism , Repetitive Sequences, Nucleic Acid/drug effects
6.
J Cardiovasc Pharmacol Ther ; 17(3): 334-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22308547

ABSTRACT

OBJECTIVE: Synthetic class A amphipathic helical peptide analogs of apolipoprotein-AI (apoAI; with varied phenylalanine residues) are emerging therapeutic approaches under investigation for atherosclerosis. Utilizing retroinverso sequencing, we designed reverse-D4F (Rev-D4F) peptide with 18 d-amino acids containing 4 phenylalanine residues and reverse order that allows the side chain residues to be of exact alignment and superimposable to those of the parent l-amino acid peptide. This study examined the effect of Rev-D4F on atherosclerosis in apolipoprotein E (apoE)-null mice and the underlying mechanisms. MATERIALS/METHODS: ApoE-null mice were fed a chow diet and administered water (control), Rev-D4F, or L4F mimetic peptides (0.4 mg/mL, equivalent to 1.6 mg/d) orally in drinking water for 6 weeks. Aortic root atherosclerotic lesion area, lesion macrophage content, and the ability of plasma high-density lipoprotein (HDL) to influence monocyte chemotaxis were measured. RESULTS: Rev-D4F significantly decreased aortic sinus atherosclerotic lesion area and lesion macrophage content without affecting plasma total and HDL-cholesterol levels in apoE-null mice. The HDL from Rev-D4F-treated mice showed enhanced anti-inflammatory monocyte chemotactic activity, while low-density lipoprotein (LDL) exhibited reduced proinflammatory activity. In in vitro studies, Rev-D4F inhibited LDL oxidation, endothelial cell vascular cell adhesion molecule 1 (VCAM-1), and monocyte chemotactic factor 1 (MCP-1) expression, and monocyte adhesion to aortic endothelial cells. CONCLUSIONS: The Rev-D4F inhibits atherosclerosis by inhibiting endothelial inflammatory/oxidative events and improving HDL function. The data suggest that Rev-D4F may be an effective apoAI mimetic peptide for further development in preventing atherosclerosis.


Subject(s)
Apolipoprotein A-I/pharmacology , Apolipoproteins E/metabolism , Atherosclerosis/prevention & control , Peptides/pharmacology , Animals , Aorta/cytology , Apolipoproteins E/genetics , Body Weight/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Coculture Techniques , Endothelial Cells , Female , Humans , Lipids/blood , Macrophages/drug effects , Mice , Mice, Knockout
7.
Am J Nephrol ; 35(2): 114-9, 2012.
Article in English | MEDLINE | ID: mdl-22212390

ABSTRACT

BACKGROUND/AIMS: Intravenous (IV) iron preparations are widely used in the management of anemia in ESRD populations. Recent changes in reimbursement policy have dramatically increased the use of IV iron to lower the use of costly erythropoiesis-stimulating agents. These preparations are frequently administered with insufficient attention to the total body iron stores or presence of inflammation which is aggravated by excess iron. Endothelial injury and dysfunction are critical steps in atherosclerosis, thrombosis and cardiovascular disease. IV iron preparations raise plasma non-transferrin-bound iron which can promote oxidative stress, endothelial damage and dysfunction. We explored the effect of an IV iron preparation on endothelial cells, monocytes and isolated arteries. METHODS: Primary cultures of human aortic endothelial cells (HAEC) were treated with pharmacologically relevant concentrations of iron sucrose (10-100 µg/ml) for 4-24 h. Endothelial cell morphology, viability, and monocyte adhesion were tested. Endothelial function was assessed by measuring the vasorelaxation response to acetylcholine in normal rat thoracic aorta rings preincubated with iron sucrose (200 µg/ml). RESULTS: In contrast to the control HAEC which showed normal cobblestone appearance, cells treated with iron sucrose (50-100 µg/ml) for 4 h showed loss of normal morphological characteristics, cellular fragmentation, shrinkage, detachment, monolayer disruption and nuclear condensation/fragmentation features signifying apoptosis. HAEC exposure to iron sucrose (10-100 µg/ml) increased monocyte adhesion 5- to 25-fold. Incubation in media containing 200 µg/ml iron sucrose for 3 h caused marked reduction in the acetylcholine-mediated relaxation in phenylephrine-precontracted rat aorta. CONCLUSION: Pharmacologically relevant concentration of iron sucrose results in endothelial injury and dysfunction and marked increase in monocyte adhesion.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Ferric Compounds/pharmacology , Glucaric Acid/pharmacology , Hematinics/pharmacology , Monocytes/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelium, Vascular/injuries , Endothelium, Vascular/physiopathology , Ferric Oxide, Saccharated , Humans , In Vitro Techniques , Male , Monocytes/immunology , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects
8.
J Lipid Res ; 51(8): 2211-22, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20371549

ABSTRACT

Pioglitazone, a hypoglycemic agent, has been shown to increase plasma HDL cholesterol, but the mechanism is incompletely understood. We further investigated effects of pioglitazone on transcriptional regulation of apolipoprotein (apo)A-I gene and functional properties of pioglitazone-induced apoA-I-containing particles. Pioglitazone dose-dependently stimulated apoA-I promoter activities in HepG2 cells. A peroxisome proliferator-activated receptor (PPAR)-response element located in site A (-214 to -192 bp, upstream of the transcription start site) of the promoter is required for pioglitazone-induced apoA-I gene transcription. Deletion of site A (-214 to -192 bp), B (-169 to -146 bp), or C (-134 to -119 bp), which clusters a number of cis-acting elements for binding of different transcription factors, reduced the basal apoA-I promoter activities, and no additional pioglitazone-sensitive elements were found within this region. Overexpression or knock-down of liver receptor homolog-1, a newly identified nuclear factor with strong stimulatory effect on apoA-I transcription, did not alter pioglitazone-induced apoA-I transcription. Pioglitazone-induced apoA-I transcription is mainly mediated through PPARalpha but not PPARgamma in hepatocytes. Pioglitazone induced production of HDL enriched in its subfraction containing apoA-I without apoA-II, which inhibited monocyte adhesion to endothelial cells in vitro. In conclusion, pioglitazone increases apoA-I production by directly enhancing PPAR-response element-dependent transcription, resulting in generation of apoA-I-containing HDL particles with increased anti-inflammatory property.


Subject(s)
Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Hypoglycemic Agents/pharmacology , Peroxisome Proliferator-Activated Receptors/genetics , Response Elements , Thiazolidinediones/pharmacology , Transcription, Genetic/drug effects , Aorta/cytology , Base Sequence , Cell Adhesion/drug effects , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Hep G2 Cells , Humans , Lipoproteins, HDL/chemistry , Monocytes/cytology , Monocytes/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Pioglitazone
9.
Atherosclerosis ; 202(1): 68-75, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18550065

ABSTRACT

In pharmacological doses, nicotinic acid (niacin) reduces myocardial infarction, stroke and atherosclerosis. The beneficial effects of niacin on lipoproteins are thought to mediate these effects. We hypothesized that niacin inhibits oxidative stress and redox-sensitive inflammatory genes that play a critical role in early atherogenesis. In cultured human aortic endothelial cells (HAEC), niacin increased nicotinamide adenine dinucleotide phosphate (NAD(P)H) levels by 54% and reduced glutathione (GSH) by 98%. Niacin inhibited: (a) angiotensin II (ANG II)-induced reactive oxygen species (ROS) production by 24-86%, (b) low density lipoprotein (LDL) oxidation by 60%, (c) tumor necrosis factor alpha (TNF-alpha)-induced NF-kappaB activation by 46%, vascular cell adhesion molecule-1 (VCAM-1) by 77-93%, monocyte chemotactic protein-1 (MCP-1) secretion by 34-124%, and (d) in a functional assay TNF-alpha-induced monocyte adhesion to HAEC (41-54%). These findings indicate for the first time that niacin inhibits vascular inflammation by decreasing endothelial ROS production and subsequent LDL oxidation and inflammatory cytokine production, key events involved in atherogenesis. Initial data presented herein support the novel concept that niacin has vascular anti-inflammatory and potentially anti-atherosclerotic properties independent of its effects on lipid regulation.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/drug effects , Monocytes/cytology , Niacin/pharmacology , Oxidation-Reduction , Oxidative Stress , Aorta/cytology , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/pathology , Glutathione/metabolism , Humans , Lipoproteins, LDL/metabolism , Monocytes/metabolism , Reactive Oxygen Species , Vascular Cell Adhesion Molecule-1/metabolism , Vasodilator Agents/pharmacology
10.
Curr Atheroscler Rep ; 11(1): 45-51, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19080727

ABSTRACT

Niacin has long been used in the treatment of dyslipidemia and cardiovascular disease. Recent research on niacin has been focused on understanding the mechanism of action of niacin and preparation of safer niacin formulations. New findings indicate that niacin does the following: 1) it inhibits hepatic diacylglycerol acyltransferase 2, resulting in inhibition of triglyceride synthesis and decreased apolipoprotein B-containing lipoproteins; 2) it decreases the surface expression of hepatic adenosine triphosphate synthase beta-chain, leading to decreased holoparticle high-density lipoprotein catabolism and increased high-density lipoprotein levels; and 3) it increases redox potential in arterial endothelial cells, resulting in inhibition of redox-sensitive genes. Flushing, an adverse effect of niacin, results from niacin receptor GPR109A-mediated production of prostaglandin D2 and E2 via DP1 and EP2/4 receptors. DP1 receptor antagonist (laropiprant) attenuates the niacin flush. A reformulated preparation of extended-release niacin (Niaspan; Abbott, Abbott Park, IL) lowers flushing compared with an older Niaspan formulation. These advancements in niacin research have rejuvenated its use for the treatment of dyslipidemia and cardiovascular disease.


Subject(s)
Hypolipidemic Agents/therapeutic use , Niacin/therapeutic use , Adipocytes/metabolism , Anti-Inflammatory Agents , Antioxidants , Apolipoprotein A-I/blood , Cardiovascular Diseases/drug therapy , Cholesterol/blood , Dyslipidemias/drug therapy , Flushing/chemically induced , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Triglycerides/blood
11.
Life Sci ; 83(17-18): 595-601, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18805430

ABSTRACT

Hyperlipidemia and the glomerular accumulation of atherogenic lipoproteins (low density lipoprotein, LDL; and its oxidatively-modified variants, ox-LDL) are commonly associated with the development of glomerular mesangial proliferative diseases. However, cellular signaling mechanisms by which atherogenic lipoproteins stimulate mesangial cell proliferation are poorly defined. In this study, we examined the effect of atherogenic lipoproteins on the activation of mesangial cell epidermal growth factor (EGF) receptor, mitogen activated protein kinase (MAP kinase), Ras, and mesangial cell proliferation. Stimulation of mesangial cells with LDL, and with greater activity, ox-LDL, markedly induced the transactivation of EGF receptor within 5 min of stimulation; the effect persisted up to at least 60 min LDL, and with a greater degree, ox-LDL, increased the activation of Ras, MAP kinase, and mesangial cell proliferation. Inhibition of EGF receptor kinase activity and/or MAP kinase activation blocked both LDL- and ox-LDL-induced mesangial cell proliferation. We suggest that the accumulation of LDL and more potently its oxidized forms within the glomerulus, through the transactivation of EGF receptor, stimulate down-stream Ras-MAP kinase signaling cascade leading to mesangial cell proliferation. Regulation of glomerular accumulation of atherogenic lipoproteins and/or EGF receptor signaling may provide protective environment against mesangial hypercellularity seen in glomerular diseases.


Subject(s)
ErbB Receptors/physiology , Lipoproteins, LDL/toxicity , Mesangial Cells/cytology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Humans , MAP Kinase Signaling System , Mice , Oxidative Stress , Phosphorylation , Quinazolines , Tyrphostins/pharmacology , ras Proteins/metabolism
12.
Metabolism ; 57(7): 973-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18555840

ABSTRACT

Hydroxymethylglutaryl-coenzyme A reductase inhibitors (statins) are extensively used to regulate dyslipidemia and to reduce atherosclerotic cardiovascular disease. In addition to effectively lowering cholesterol and low-density lipoprotein levels, rosuvastatin and certain other statins can also increase plasma high-density lipoprotein (HDL) cholesterol modestly. However, the mechanism of action of rosuvastatin on HDL metabolic processes is not understood. Using cultured human hepatoblastoma cells (Hep G2) as an in vitro model system, we assessed the effect of rosuvastatin on apolipoprotein (apo) A-I and apo A-II (the major proteins of HDL) synthesis and HDL catabolic processes. Rosuvastatin dose-dependently increased messenger RNA expression and de novo synthesis of apo A-I but not apo A-II. Rosuvastatin selectively increased the synthesis of HDL particles containing only apo A-I (LP A-I) but not particles containing both apo A-I and A-II (LP A-I + A-II). The HDL(3)-protein or HDL(3)-cholesterol ester uptake by Hep G2 cells was not affected by rosuvastatin. The apo A-I-containing particles secreted by rosuvastatin-treated Hep G2 significantly increased cholesterol efflux from fibroblasts. The data indicate that rosuvastatin increases hepatic apo A-I but not apo A-II messenger RNA transcription, thereby selectively increasing the synthesis of functionally active apo A-I-containing HDL particles, which mediate cholesterol efflux from peripheral tissues. We suggest that this mechanism of action of rosuvastatin to increase apo A-I production without apo A-I/HDL removal may result in increased apo A-I turnover that results in accelerated reverse cholesterol transport.


Subject(s)
Apolipoprotein A-II/biosynthesis , Apolipoprotein A-I/biosynthesis , Fluorobenzenes/pharmacology , Hepatocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Cell Line, Tumor , Cholesterol, HDL/metabolism , Fibroblasts/metabolism , Hepatocytes/drug effects , Humans , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium , Stimulation, Chemical , Transcription, Genetic
13.
Biochim Biophys Acta ; 1771(11): 1364-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17950662

ABSTRACT

Lysophosphatidylcholine (LPC), a major component of oxidized-low density lipoproteins (ox-LDL), modulates various pathobiological processes involved in vascular and glomerular diseases. Although several studies have shown increased plasma concentrations of ox-LDL as well as LPC in patients with renal disease, the role of LPC in mesangial cell proliferation and associated signaling mechanisms are not clearly understood. In this study, we have shown that LPC induced the phosphorylation of epidermal growth factor receptor (EGFR), as well as the p42/44 MAP kinases. LPC activated Src-kinase and protein kinase C (PKC), and both Src kinase inhibitor PP-2 and PKC inhibitor inhibited the activation of EGFR by LPC. LPC (5-25 microM) stimulated human mesangial cell proliferation by 4-5 fold. Preincubation of mesangial cells with the Src inhibitor (PP-2), or PKC inhibitor (bisindolylmaleimide GF109203-X), or EGF receptor kinase inhibitor (AG1478), or MEK inhibitor (PD98059) significantly inhibited LPC-mediated mesangial cell proliferation. The data suggest that LPC, by activating Src and PKC signaling pathways, stimulates EGF receptor transactivation and down-stream MAP kinase signaling resulting in mesangial hypercellularity, which is a characteristic feature of diverse renal diseases.


Subject(s)
ErbB Receptors/drug effects , ErbB Receptors/metabolism , Lysophosphatidylcholines/metabolism , Lysophosphatidylcholines/pharmacology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Protein Kinase C/metabolism , src-Family Kinases/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Maleimides/pharmacology , Mesangial Cells/cytology , Protein Kinase C/antagonists & inhibitors , Quinazolines , Tyrphostins/pharmacology , src-Family Kinases/antagonists & inhibitors
14.
Am J Physiol Renal Physiol ; 287(6): F1250-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15292052

ABSTRACT

Lysophosphatidic acid (LPA), a major member of the bioactive lysophospholipids in serum, possesses diverse physiological activities including cell proliferation. Recently, three endothelial differentiation gene (EDG) family receptors, including EDG-2 (LPA1), EDG-4 (LPA2), and EDG-7 (LPA3), have been identified as LPA receptors. The role of LPA and their receptors in mesangial cell physiology is not clearly understood. This study examined the expression profile of EDG receptors as a function of cell density and the participation of EDG receptors in human mesangial cell proliferation by LPA. We showed that mesangial cells express all three EDG family LPA receptors in a cell density-dependent manner. EDG-7 maximally expressed at sparse cell density and minimally expressed in dense cell population. The EDG-2 expression pattern was opposite to the EDG-7. No changes in EDG-4 expression as a function of cell density were noted. DNA synthetic rate was greater in sparse cell density compared with dense cell population and followed a similar pattern with EDG-7 expression. Comparative studies in sparse and dense cell density indicated that EDG-7 was positively associated, whereas EDG-2 was negatively associated with cell proliferation rate. LPA induced mesangial cell proliferation by 1.5- to 3.5-fold. Dioctanoylglycerol pyrophosphate, an antagonist for EDG-7, almost completely inhibited mesangial cell proliferation induced by LPA. We suggest that EDG-7 regulates LPA-mediated mesangial cell proliferation. Additionally, these data suggest that EDG-7 and EDG-2 LPA receptors play a diverse role as proliferative and antiproliferative, respectively, in mesangial cells. Regulation of EDG family receptors may be importantly linked to mesangial cell-proliferative processes.


Subject(s)
Cell Count , Cell Division/drug effects , Gene Expression Regulation , Glomerular Mesangium/cytology , Lysophospholipids/pharmacology , Receptors, Lysosphingolipid/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/physiology , Receptors, Lysosphingolipid/physiology , Reverse Transcriptase Polymerase Chain Reaction
15.
J Lipid Res ; 45(10): 1835-45, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15258194

ABSTRACT

Niacin is a widely used lipid-regulating agent in dyslipidemic patients. Previously, we have shown that niacin inhibits triacylglycerol synthesis. In this report, using HepG2 cells, we have examined the effect of niacin on the mRNA expression and microsomal activity of diacylglycerol acyltransferase 1 and 2 (DGAT1 and DGAT2), the last committed but distinctly different enzymes for triglyceride synthesis. Addition of niacin to the DGAT assay reaction mixture dose-dependently (0-3 mM) inhibited DGAT activity by 35-50%, and the IC(50) was found to be 0.1 mM. Enzyme kinetic studies showed apparent K(m) values of 8.3 microM and 100 microM using [(14)C]oleoyl-CoA and sn-1,2-dioleoylglycerol as substrates, respectively. A decrease in apparent V(max) was observed with niacin, whereas the apparent K(m) remained constant. A Lineweaver-Burk plot of DGAT inhibition by niacin showed a noncompetitive type of inhibition. Niacin selectively inhibited DGAT2 but not DGAT1 activity. Niacin inhibited overt DGAT activity. Niacin had no effect on the expression of DGAT1 and DGAT2 mRNA. These data suggest that niacin directly and noncompetitively inhibits DGAT2 but not DGAT1, resulting in decreased triglyceride synthesis and hepatic atherogenic lipoprotein secretion, thus indicating a major target site for its mechanism of action.


Subject(s)
Acyltransferases/antagonists & inhibitors , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/metabolism , Niacin/pharmacology , Acyltransferases/drug effects , Acyltransferases/genetics , Acyltransferases/metabolism , Cell Line, Tumor , Diacylglycerol O-Acyltransferase , Dose-Response Relationship, Drug , Humans , Kinetics , Lipoproteins/metabolism , RNA, Messenger/analysis , Triglycerides/biosynthesis
16.
J Nutr Biochem ; 14(6): 298-305, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12873710

ABSTRACT

Niacin has been widely used as a pharmacologic agent to regulate abnormalities in plasma lipid and lipoprotein metabolism and in the treatment of atherosclerotic cardiovascular disease. Although the use of niacin in the treatment of dyslipidemia has been reported as early as 1955, only recent studies have yielded an understanding about the cellular and molecular mechanism of action of niacin on lipid and lipoprotein metabolism. In brief, the beneficial effect of niacin to reduce triglycerides and apolipoprotein-B containing lipoproteins (e.g., VLDL and LDL) are mainly through: a) decreasing fatty acid mobilization from adipose tissue triglyceride stores, and b) inhibiting hepatocyte diacylglycerol acyltransferase and triglyceride synthesis leading to increased intracellular apo B degradation and subsequent decreased secretion of VLDL and LDL particles. The mechanism of action of niacin to raise HDL is by decreasing the fractional catabolic rate of HDL-apo AI without affecting the synthetic rates. Additionally, niacin selectively increases the plasma levels of Lp-AI (HDL subfraction without apo AII), a cardioprotective subfraction of HDL in patients with low HDL. Using human hepatocytes (Hep G2 cells) as an in vitro model system, recent studies indicate that niacin selectively inhibits the uptake/removal of HDL-apo AI (but not HDL-cholesterol ester) by hepatocytes, thereby increasing the capacity of retained HDL-apo AI to augment cholesterol efflux through reverse cholesterol transport pathway. The studies discussed in this review provide evidence to extend the role of niacin as a lipid-lowering drug beyond its role as a vitamin.


Subject(s)
Cardiovascular Diseases , Cholesterol , Niacin/physiology , Acyltransferases/antagonists & inhibitors , Adipose Tissue/metabolism , Apolipoprotein A-I/metabolism , Apolipoproteins B/blood , Apolipoproteins B/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Cholesterol/blood , Cholesterol/metabolism , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Coronary Disease/prevention & control , Diacylglycerol O-Acyltransferase , Enzyme Inhibitors , Humans , Liver/enzymology , Liver/metabolism , Niacin/administration & dosage , Triglycerides/biosynthesis , Triglycerides/blood
17.
Atherosclerosis ; 164(2): 221-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12204791

ABSTRACT

The mechanism of action of a widely used drug gemfibrozil to reduce triglycerides (TG) and apolipoprotein B (apo B) is incompletely understood. Using human hepatoblastoma (HepG2) cells, we examined the effect of gemfibrozil on apo B secretion and TG synthesis catalyzed by diacylglycerol acyltransferase (DGAT), primary processes associated with the secretion of LDL. Gemfibrozil significantly decreased apo B secretion by HepG2 cells. It decreased oleate-induced stimulation of apo B secretion, suggesting that gemfibrozil-mediated inhibition of apo B secretion may be dependent on the synthesis of TG catalyzed by DGAT. Pre-incubation of HepG2 cells with gemfibrozil (200-400 micromol/l for 48 h) significantly inhibited microsomal DGAT activity. When added directly to the DGAT assay system containing control microsomes, gemfibrozil significantly inhibited the activity of DGAT by 14-25%. Gemfibrozil (200-400 micromol/l) inhibited TG synthesis by 47-50% as measured by the incorporation of 3H-oleic acid into TG. The data indicate that gemfibrozil inhibits DGAT activity resulting in decreased synthesis of TG and its availability for apo B lipidation rendering it susceptible to intracellular apo B degradation leading to the decreased secretion. These in-vitro data suggest a novel additional mechanism by which gemfibrozil lowers plasma TG and atherogenic apo B lipoproteins in dyslipidemic patients.


Subject(s)
Acyltransferases/metabolism , Apolipoproteins B/drug effects , Apolipoproteins B/metabolism , Gemfibrozil/pharmacology , Liver/cytology , Triglycerides/metabolism , Acyltransferases/drug effects , Diacylglycerol O-Acyltransferase , Hepatoblastoma , Humans , Liver Neoplasms , Microsomes/drug effects , Microsomes/metabolism , Sensitivity and Specificity , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...