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










Publication year range
1.
Clin Nephrol ; 74(4): 273-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20875379

ABSTRACT

UNLABELLED: End-stage renal disease (ESRD) causes accelerated atherosclerosis which is mediated by oxidative stress and inflammation. Activation and infiltration of monocytes represent the critical steps in atherogenesis which is advanced by oxidized LDL and inhibited by HDL. Via its main apolipoprotein (apoA-I) and constituent enzymes (paraoxonase; glutathione peroxidase (GPX), LCAT) HDL exerts potent antioxidant/anti-inflammatory functions. We have found marked reduction of HDL antioxidant/anti-inflammatory and heightened LDL pro-oxidant/pro-inflammatory activities in ESRD patients. Given the inseparable link between oxidative stress and inflammation, we tested the hypothesis that antioxidant therapy may improve anti-inflammatory (monocyte adhesion-promoting capacity) properties of plasma in ESRD patients. METHODS: We studied 20 hemodialysis patients who after a 4-week wash-out period were treated with a potent antioxidant cocktail (vitamin (v) E, 800 IU; vC, 250 mg; vB6, 100 mg; vB12, 250 µg and folic acid 10 mg daily) for 8 weeks. Twelve healthy volunteers served as control. Pre-dialysis plasma samples were obtained at the onset and conclusion of the study. Markers of oxidative stress and inflammation, apoA-I, HDL-associated enzymes and monocyte adhesion assay were measured using cultured aortic endothelial cells. RESULTS: ESRD patients exhibited reduced plasma level of apoA-1 and antioxidant enzymes, elevated markers of oxidative stress and inflammation and heightened monocyte adhesion-promoting capacity. Antioxidant therapy failed to improve these abnormalities. CONCLUSIONS: High doses of antioxidant vitamins fail to improve oxidative stress, inflammation or plasma monocyte adhesion-promoting capacity in ESRD patients. Thus, high doses of vitamins beyond the routinely-prescribed supplements do not appear to be beneficial in this patient population.


Subject(s)
Antioxidants/therapeutic use , Inflammation/drug therapy , Kidney Failure, Chronic/drug therapy , Monocytes/physiology , Atherosclerosis/etiology , Cell Adhesion/drug effects , Female , Glutathione Peroxidase/blood , Humans , Inflammation/etiology , Kidney Failure, Chronic/metabolism , Lipids/blood , Male , Middle Aged , Oxidative Stress
2.
Int J Clin Pract ; 63(9): 1369-77, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19691622

ABSTRACT

AIMS: To summarise the metabolic responses to niacin that can lead to flushing and to critically evaluate flushing mitigation research. METHODS AND RESULTS: This comprehensive review of the mechanism of action of niacin-induced flushing critically evaluates research regarding flushing mitigating formulations and agents. Niacin induces flushing through dermal Langerhans cells where the activation of G protein-coupled receptor 109A (GPR109A) increases arachidonic acid and prostaglandins, such as prostaglandin D(2) (PGD(2)) and prostaglandin E(2) (PGE(2)), subsequently activating prostaglandin D(2) receptor (DP(1)), prostaglandin E(2) receptor (EP(2)) and prostaglandin E receptor 4 (EP(4)) in capillaries and causing cutaneous vasodilatation. Controlling niacin absorption rates, inhibiting prostaglandin production, or blocking DP(1), EP(2) and EP(4) receptors can inhibit flushing. Niacin extended-release (NER) formulations have reduced flushing incidence, duration and severity relative to crystalline immediate-release niacin with similar lipid efficacy. Non-steroidal anti-inflammatory drugs (NSAIDs), notably aspirin given 30 min before NER at bedtime, further reduce flushing. An antagonist to the DP(1) receptor (laropiprant) combined with an ER niacin formulation can reduce flushing; however, significant residual flushing occurs with clinically-relevant dosages. CONCLUSIONS: Niacin is an attractive option for treating dyslipidemic patients, and tolerance to niacin-induced flushing develops rapidly. Healthcare professionals should particularly address flushing during niacin dose titration.


Subject(s)
Dyslipidemias/drug therapy , Flushing/chemically induced , Hypolipidemic Agents/pharmacology , Niacin/pharmacology , Arachidonic Acid/metabolism , Delayed-Action Preparations , Flushing/prevention & control , Humans , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Niacin/administration & dosage , Niacin/adverse effects , Patient Education as Topic , Prostaglandin Antagonists/therapeutic use , Prostaglandins , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Vasoconstriction/drug effects , Vasodilation/drug effects
3.
Histol Histopathol ; 20(2): 603-13, 2005 04.
Article in English | MEDLINE | ID: mdl-15736064

ABSTRACT

Lysophosphatidic acid (LPA), lyso-phosphatidylcholine (LPC), and sphingosine-1-phosphate (S1P) are major biologically active lysophospholipids (LPLs) that are produced by activated platelets, monocyte/macrophages, and many types of mammalian cells. LPLs have been shown to induce a wide array of physiological and pathophysiological properties including cellular differentiation, proliferation, migration, extracellular matrix deposition, change in morphology, and chemotactic responses. The recent cloning and identification of G protein-coupled receptors as specific receptors for LPLs created a great deal of interest in LPLs signaling and diverse biological responses. The pathobiological role of LPLs has been implicated in a number of pathological states and human diseases including atherosclerosis, glomerulosclerosis, post-ischemic renal failure, polycystic kidney disease, and ovarian cancer. Although the research in this area is growing at an enormous rate, this review is specifically focused on the recent understanding of the pathophysiological properties of LPA and LPC with special reference to kidney diseases, and their specific G-protein-coupled receptors and intracellular signaling pathways.


Subject(s)
Glomerular Mesangium/metabolism , Kidney Diseases/etiology , Kidney Diseases/metabolism , Lysophospholipids/metabolism , Phosphorylcholine/analogs & derivatives , Sphingosine/analogs & derivatives , Animals , Cell Proliferation , Glomerular Mesangium/pathology , Humans , Kidney Diseases/pathology , Lysophosphatidylcholines , Models, Biological , Phosphorylcholine/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophospholipid/metabolism , Signal Transduction , Sphingosine/metabolism
4.
Histol Histopathol ; 17(2): 497-505, 2002 04.
Article in English | MEDLINE | ID: mdl-11962755

ABSTRACT

Abnormalities in lipid and lipoprotein metabolism are commonly observed in patients with chronic renal disease. Specifically, hyperlipidemia and the glomerular deposition of atherogenic lipoproteins (e.g., Low density lipoprotein, LDL; and its oxidized variants) are implicated in key pathobiological processes involved in the development of glomerular disease, including stimulation of monocyte infiltration into the mesangial space, mesangial cell hypercellularity, and mesangial extracellular matrix deposition. This review discusses recent understanding of glomerular mitogenic responses, intracellular signaling events associated with mesangial hypercellularity in renal diseases, and the participation of cholesterol and atherogenic lipoproteins in intracellular signaling pathways involved in mesangial cell proliferation. Generally, the mitogenic intracellular signaling pathways are regulated by the activation of series of transmembrane and cytoplasmic protein tyrosine kinases that converge into the activation of Ras and down-stream mitogen-activated protein kinase (MAP kinase). Activated MAP kinase, through translocating into the nucleus and the activation of various transcription factors and protooncogenes, regulate cell proliferation. The importance of mitogenic intracellular signaling in mesangial proliferative disease has only recently been recognized and showed that the activation of MAP kinase and/or cyclin/cyclin-dependent kinases play crucial role in different phases of cell growth cycle and hypercellularity of glomerular cells in various experimental renal diseases. Using glomerular mesangial cells as an in-vitro model system, studies from our laboratory indicated that the accumulation of LDL and more potently its oxidized forms within the glomerulus, through the activation of membrane receptor tyrosine kinases (e.g., EGF receptor), activate Ras and MAP kinase signaling cascade leading to DNA synthesis and subsequent mesangial cell proliferation. These data suggest that atherogenic lipoproteins may act as one of the major endogenous modulators for mitogenic signaling response and cell proliferation within the glomerulus. It is reasonable to speculate that the correction or reduction of hyperlipidemia, glomerular lipid deposition, and the pro-oxidative milieu within the glomerulus, through the inhibition of mitogenic signaling events, may provide protective environment against mesangial hypercellularity and subsequent matrix deposition, and the progression of renal disease.


Subject(s)
Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Lipoproteins, LDL/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Animals , Cell Division , Cholesterol/metabolism , ErbB Receptors/metabolism , Humans , Kidney Diseases/metabolism , Kidney Glomerulus/cytology , Lipid Metabolism , Lipids/physiology , Lipoproteins/metabolism , Lipoproteins/physiology , Lipoproteins, LDL/physiology , Lysophosphatidylcholines/metabolism , Phosphorylation , ras Proteins/metabolism
5.
Arterioscler Thromb Vasc Biol ; 21(11): 1783-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701466

ABSTRACT

Evidence indicates that the high density lipoprotein (HDL) subfraction containing apolipoprotein A-I without apolipoprotein AII (LP-AI) is more antiatherogenic than HDL particles containing apolipoprotein A-I and apolipoprotein A-II (LP-AI+AII). This study examined the effect of extended-release niacin (niacin-ER) and gemfibrozil on LP-AI and LP-AI+AII particles in patients with low levels of HDL cholesterol (HDL-C). Mechanisms by which these agents modulate HDL particles were investigated by in vitro studies using human hepatoblastoma (Hep G2) cells. A total of 139 patients with low HDL-C (

Subject(s)
Apolipoprotein A-I/biosynthesis , Arteriosclerosis/metabolism , Gemfibrozil/pharmacology , Hypolipidemic Agents/pharmacology , Lipoproteins, HDL/chemistry , Niacin/pharmacology , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/blood , Arteriosclerosis/drug therapy , Cholesterol, HDL/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Hepatocytes/metabolism , Humans , Lipids/blood , Tumor Cells, Cultured
6.
Curr Atheroscler Rep ; 2(1): 36-46, 2000 Jan.
Article in English | MEDLINE | ID: mdl-11122723

ABSTRACT

It is generally accepted that the increased concentrations of apolipoprotein (apo) B containing very low-density lipoproteins (VLDL) and low-density lipoproteins (LDL), and decreased levels of apo AI containing high-density lipoproteins (HDL) are correlated to atherosclerotic cardiovascular disease. Current evidence indicates that the post-translational apo-B degradative processes regulate the hepatic assembly and secretion of VLDL and the subsequent generation of LDL particles. The availability of triglycerides (TG) for the addition to apo B during intracellular processing appears to play a central role in targeting apo B for either intracellular degradation or assembly and secretion as VLDL particles. Based on the availability of TG, the liver secretes either dense TG-poor VLDL2 or large TG-rich VLDL1 particles, and these particles serve as precursors for the formation of more buoyant or small, dense LDL particles by lipid transfer protein- and hepatic lipase-mediated processes. HDLs are a heterogenous class of lipoproteins, and apo AI (the major protein of HDL) participates in reverse cholesterol transport, a process by which excess cholesterol is eliminated. Recent studies indicate that HDL particles containing only apo A-I (LPA-I) are more effective in reverse cholesterol transport and more anti-atherogenic than HDL particles containing both apo A-I and apo A-II (LPA-I + A-II).


Subject(s)
Apolipoproteins A/metabolism , Apolipoproteins B/metabolism , Arteriosclerosis/prevention & control , Niacin/pharmacology , Arteriosclerosis/physiopathology , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism
7.
Am J Physiol Endocrinol Metab ; 279(1): E11-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10893317

ABSTRACT

Diabetes is associated with endothelial dysfunction and increased risk of hypertension, cardiovascular disease, and renal complications. Earlier studies have revealed that hyperglycemia impairs nitric oxide (NO) production and diabetes causes endothelial dysfunction in humans and experimental animals. This study was designed to test the effects of altered concentrations of glucose, insulin, and glucagon, the principal variables in types I and II diabetes, on NO production and endothelial NO synthase (eNOS) expression in cultured human coronary endothelial cells. Cultured endothelial cells were incubated in the presence of glucose at either normal (5.6 mM) or high (25 mM) concentrations for 7 days. The rates of basal and bradykinin-stimulated NO production (nitrate + nitrite) and eNOS protein expression (Western blot) were then determined at the basal condition and in the presence of insulin (10(-8) and 10(-7) M), glucagon (10(-8) and 10(-7) M), or both. Incubation with a high-glucose concentration for 7 days significantly downregulated, whereas insulin significantly upregulated, basal and bradykinin-stimulated NO production and eNOS expression in cultured endothelial cells. The stimulatory action of insulin was mitigated by high-glucose concentration and abolished by cotreatment of cells with glucagon. Thus hyperglycemia, insulinopenia, and hyperglucagonemia, which frequently coexist in diabetes, can work in concert to suppress NO production by human coronary artery endothelial cells.


Subject(s)
Glucagon/pharmacology , Hyperglycemia/enzymology , Insulin Antagonists/pharmacology , Insulin/pharmacology , Nitric Oxide Synthase/metabolism , Cells, Cultured , Glucose , Humans , Hyperglycemia/chemically induced , Nitric Oxide Synthase Type III , Osmolar Concentration
8.
Atherosclerosis ; 149(1): 43-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10704613

ABSTRACT

Although the role of multiple humoral agents (such as plasma albumin, glucose, hormones etc.) are implicated in lipoprotein metabolism, the mechanism of action of these agents on various steps of the synthesis and secretion of lipoproteins and apolipoproteins (protein moieties of lipoproteins) are not completely understood. Specifically, the hepatocellular mechanisms of the effect of albumin and fatty acids on apolipoprotein (apo) AI and AII [major proteins of high density lipoproteins (HDL)] synthesis and secretion are not known. Using human hepatoblastoma cells (Hep G2) as an in vitro model system, this study examined the effect of albumin and fatty acids on the synthesis, secretion, and the steady-state mRNA expression of apo AI and AII. The data indicated that the incubation of Hep G2 cells with albumin, dose-dependently, inhibited apo AI and AII accumulation (secretion) in the media, de novo synthesis, and the steady-state mRNA expression. Albumin did not alter total protein synthesis; thus the effect of albumin appeared to be specific for the synthesis and secretion of apo AI and apo AII. Free fatty acids (FFA) are transported by albumin and diseases characterized by enhanced FFA mobilization (e.g. diabetes mellitus) are associated with low HDL levels. Studies were therefore performed to examine the effect of albumin-bound-oleic acid on apo AI and apo AII production. The results showed that the albumin-oleate complex further increased the inhibitory effects of albumin on apo AI and apo AII production. These data suggest how HDL metabolism may be affected at the hepatocellular level by alterations in plasma albumin concentrations and/or fatty acid mobilization in clinical situations characterized by altered HDL levels.


Subject(s)
Albumins/pharmacology , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-I/biosynthesis , Oleic Acid/pharmacology , Apolipoprotein A-I/drug effects , Apolipoprotein A-I/genetics , Apolipoprotein A-II/drug effects , Apolipoprotein A-II/genetics , Dose-Response Relationship, Drug , Drug Interactions , Hepatoblastoma/chemistry , Humans , Liver Neoplasms/chemistry , Probability , RNA, Messenger/analysis , Tumor Cells, Cultured
9.
Am J Physiol ; 277(3): F328-37, 1999 09.
Article in English | MEDLINE | ID: mdl-10484515

ABSTRACT

Although lysophosphatidylcholine (LPC)-mediated cellular responses are attributed to the activation of protein kinase C (PKC), relatively little is known about the upstream signaling mechanisms that regulate the activation of PKC and downstream mitogen-activated protein (MAP) kinase. LPC activated p42 MAP kinase and PKC in mesangial cells. LPC-mediated MAP kinase activation was inhibited (but not completely) by PKC inhibition, suggesting additional signaling events. LPC stimulated protein tyrosine kinase (PTK) activity and induced Ras-GTP binding. LPC-induced MAP kinase activity was blocked by the PTK inhibitor genistein. Because LPC increased PTK activity, we examined the involvement of phospholipase Cgamma-1 (PLCgamma-1) as a key participant in LPC-induced PKC activation. LPC stimulated the phosphorylation of PLCgamma-1. PTK inhibitors suppressed LPC-induced PKC activity, whereas the same had no effect on phorbol 12-myristate 13-acetate-mediated PKC activity. Other lysophospholipids [e.g., lysophosphatidylinositol and lysophosphatidic acid (LPA)] also induced MAP kinase activity, and only LPA-induced MAP kinase activation was sensitive to pertussis toxin. These results indicate that LPC-mediated PKC activation may be regulated by PTK-dependent activation of PLCgamma-1, and both PKC and PTK-Ras pathways are involved in LPC-mediated downstream MAP kinase activation.


Subject(s)
Glomerular Mesangium/physiology , Lysophosphatidylcholines/pharmacology , Signal Transduction/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Enzyme Activation , Glomerular Mesangium/drug effects , Isoenzymes/physiology , Mice , Phospholipase C gamma , Phosphorylation , Protein Kinase C/physiology , Protein-Tyrosine Kinases/physiology , Type C Phospholipases/physiology , ras Proteins/physiology
10.
Kidney Int Suppl ; 71: S70-5, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10412742

ABSTRACT

BACKGROUND: Mesangial hypercellularity is a critical early histopathological finding seen in human and experimental glomerular diseases. Hyperlipidemia and the glomerular deposition of atherogenic lipoproteins [for example, low-density lipoprotein (LDL) and its oxidized variants, minimally oxidized/modified LDL (mm-LDL)] are commonly associated with mesangial hypercellularity and the development of glomerular disease. This article reviews signal transduction pathways involved in cell proliferation and provides evidence for the participation of atherogenic lipoproteins in intracellular signaling pathways for mesangial cell proliferation. The mitogenic intracellular signaling pathways are regulated by the activation of a series of transmembrane and cytoplasmic protein tyrosine kinases that converge into the activation of Ras and downstream mitogen-activated protein (MAP) kinase. Activated MAP kinase, through translocating into the nucleus and the activation of various transcription factors and proto-oncogenes, regulates cellular proliferation. METHODS: Murine mesangial cells were stimulated with LDL and mm-LDL and were analyzed for the tyrosine kinase activity, phosphorylation of membrane proteins, activation of Ras and MAP kinase, and cell proliferation. RESULTS: The results indicated that the stimulation of mesangial cells with LDL and, with greater activity, mm-LDL induced the phosphorylation of membrane platelet-derived growth factor (PDGF) and epidermal growth factor (EGF) receptors, activated Ras, and resulted in sustained (up to 24 hr) activation of MAP kinase. LDL/mm-LDL-mediated mesangial cell proliferation and MAP kinase activation were dependent on the activation of tyrosine kinases. CONCLUSIONS: We suggest that the accumulation of LDL and more potently its oxidized forms within the glomerulus, through the activation of membrane receptor tyrosine kinases, activate the Ras and MAP kinase signaling cascade leading to DNA synthesis and subsequent cell proliferation.


Subject(s)
Arteriosclerosis/etiology , Cell Division/drug effects , Glomerular Mesangium/drug effects , Lipoproteins, LDL/pharmacology , Protein-Tyrosine Kinases/physiology , Animals , Cell Division/physiology , Enzyme Activation/drug effects , Glomerular Mesangium/cytology , Mice , Protein-Tyrosine Kinases/metabolism , Signal Transduction
11.
Kidney Int ; 55(6): 2192-202, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10354268

ABSTRACT

BACKGROUND: Abnormalities in lipid and lipoprotein metabolism have been implicated in the pathogenesis of glomerulosclerosis. Atherogenic lipoproteins [for example, low-density lipoprotein (LDL) and oxidized LDL (ox-LDL)] have been shown to stimulate glomerular monocyte chemoattractants involved in monocyte infiltration. However, the role of LDL and ox-LDL in the early monocyte adhesion to glomerular endothelial cells (ECs) and associated intracellular signaling mechanisms are not clearly understood. METHODS: In this study, we examined the effect of LDL and ox-LDL on intracellular signaling mechanisms associated with monocyte adhesion to glomerular ECs and intercellular adhesion molecule-1 (ICAM-1) expression. RESULTS: Ox-LDL, but not LDL, stimulated EC ICAM-1 expression and monocyte adhesion. Ox-LDL elevated protein tyrosine kinase (PTK) activity, and the preincubation of ECs with specific PTK inhibitors blocked ox-LDL-induced ICAM-1 message and monocyte adhesion. Whereas experimental maneuvers that inhibit either protein kinase C activation (by PKC depletion or with inhibitors) or Gi-protein-mediated pathways (pertussis toxin sensitive) had no effect on ox-LDL-induced monocyte adhesion and ICAM-1 expression. cAMP-elevating compounds did not induce ICAM-1 or monocyte adhesion. CONCLUSIONS: The data indicate that ox-LDL, by stimulating monocyte adhesion to the glomerular endothelium, may regulate monocyte infiltration within the glomerulus, supporting an early pathobiological role for atherogenic lipoproteins in glomerular injury. The results suggest that the activation of specific PTK and associated signaling may, at least in part, play a critical role in ox-LDL-mediated endothelial-monocyte interaction-related events. The data suggest that the interventions aimed at modifying associated intracellular signaling events within the glomerulus may provide potential therapeutic modalities in monocyte/macrophage-mediated glomerular disease.


Subject(s)
Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Lipoproteins, LDL/pharmacology , Monocytes/cytology , Monocytes/drug effects , Animals , Cell Adhesion/drug effects , Cells, Cultured , Endothelium/cytology , Endothelium/drug effects , Endothelium/metabolism , GTP-Binding Proteins/metabolism , Gene Expression/drug effects , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kidney Glomerulus/metabolism , Mice , Monocytes/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
12.
Arterioscler Thromb Vasc Biol ; 19(4): 1051-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195935

ABSTRACT

The mechanism by which the potent drug niacin decreases apoB-containing atherogenic lipoproteins and prevents coronary disease is unclear. Utilizing human hepatoblastoma (HepG2) cells as an in vitro model, we have examined the effect of niacin on intracellular degradation of apoB and the regulatory mechanisms involved in apoB processing. Niacin significantly increased apoB degradation in a dose- and time-dependent manner. Treatment of HepG2 cells with calpain inhibitor I [N-acetyl-leucyl-leucyl-norleucinal (ALLN), an inhibitor of certain protease-mediated apoB degradation], did not alter niacin-induced apoB degradation. Niacin decreased inhibition of oleate-mediated apoB degradation. Niacin dose-dependently inhibited the synthesis of both fatty acids and triacylglycerol (TG) by 20% to 40% as determined by the incorporation of 14C-acetate and 3H-glycerol into fatty acids and TG, respectively. Incubation of HepG2 cells with niacin significantly inhibited (by 12% to 15%) fatty acid esterification to produce TG as assessed by the incorporation of 3H-oleic acid into TG. 14C-acetate incorporation into cholesterol and phospholipids was unchanged. The activity of microsomal triglyceride transfer protein (MTP), a carrier protein for lipids, was not altered by pretreatment of cells with niacin. ApoB mRNA expression and 125I-LDL protein uptake were also unchanged. These data indicate that niacin accelerates hepatic intracellular post-translational degradation of apoB by selectively reducing triglyceride synthesis (through inhibiting both fatty acid synthesis and fatty acid esterification to produce TG) without affecting ALLN-inhibitable protease- or MTP-mediated intracellular apoB processing, resulting in decreased apoB secretion and hence lower circulating levels of the atherogenic lipoproteins.


Subject(s)
Apolipoproteins B/metabolism , Hepatoblastoma/metabolism , Intracellular Fluid/metabolism , Niacin/pharmacology , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis , Apolipoproteins B/biosynthesis , Apolipoproteins B/genetics , Hepatoblastoma/pathology , Humans , Intracellular Fluid/drug effects , Tumor Cells, Cultured
13.
Biochim Biophys Acta ; 1449(2): 137-49, 1999 Mar 08.
Article in English | MEDLINE | ID: mdl-10082972

ABSTRACT

Intermediary metabolites of cholesterol synthetic pathway are involved in cell proliferation. Lovastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase, blocks mevalonate synthesis, and has been shown to inhibit mesangial cell proliferation associated with diverse glomerular diseases. Since inhibition of farnesylation and plasma membrane anchorage of the Ras proteins is one suggested mechanism by which lovastatin prevents cellular proliferation, we investigated the effect of lovastatin and key mevalonate metabolites on the activation of mitogen-activated protein kinase (MAP kinase) and Ras in murine glomerular mesangial cells. The preincubation of mesangial cells with lovastatin inhibited the activation of MAP kinase stimulated by either FBS, PDGF, or EGF. Mevalonic acid and farnesyl-pyrophosphate, but not cholesterol or LDL, significantly prevented lovastatin-induced inhibition of agonist-stimulated MAP kinase. Lovastatin inhibited agonist-induced activation of Ras, and mevalonic acid and farnesylpyrophosphate antagonized this effect. Parallel to the MAP kinase and Ras data, lovastatin suppressed cell growth stimulated by serum, and mevalonic acid and farnesylpyrophosphate prevented lovastatin-mediated inhibition of cellular growth. These results suggest that lovastatin, by inhibiting the synthesis of farnesol, a key isoprenoid metabolite of mevalonate, modulates Ras-mediated cell signaling events associated with mesangial cell proliferation.


Subject(s)
Cholesterol/biosynthesis , Extracellular Signal-Regulated MAP Kinases , Glomerular Mesangium/enzymology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , MAP Kinase Kinase Kinases , Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Line , Cells, Cultured , Enzyme Activation/drug effects , Epidermal Growth Factor/antagonists & inhibitors , Glomerular Mesangium/drug effects , Mevalonic Acid/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases , Platelet-Derived Growth Factor/antagonists & inhibitors , Polyisoprenyl Phosphates/pharmacology , Protein Kinase Inhibitors , Sesquiterpenes , Signal Transduction
14.
Am J Nephrol ; 18(4): 344-50, 1998.
Article in English | MEDLINE | ID: mdl-9653842

ABSTRACT

The proliferation of intrinsic glomerular cells and the accumulation of extracellular matrix proteins are principal histopathological features seen in glomerular injury. Because of the marked similarity between the cellular and molecular events that occur in both atherosclerosis and glomerulosclerosis and the commonly accepted hypothesis that lipoproteins are implicated in the pathogenesis of glomerulosclerosis, we examined the effect of three atherogenic lipoproteins, low-density lipoprotein (LDL), oxidized (ox)-LDL, and minimally modified (mm)-LDL on the synthesis and secretion of extracellular matrix (ECM) proteins by mesangial cells. The incubation of SV-40 transformed murine mesangial cells with LDL (25-100 microg/ml) increased the synthesis and secretion of both fibronectin and laminin in a dose-dependent manner. Similarly, oxidized forms of LDL (25-100 micro/ml) increased fibronectin and laminin synthesis and secretion dose dependently. However, both oxidatively modified forms of LDL had a greater effect on increasing ECM protein synthesis than their native counterpart. Northern blot analysis showed a dose-dependent increase in mRNA transcripts for fibronectin and laminin in response to the incubation of mesangial cells with LDL, ox-LDL, and mm-LDL. Similar to the ECM protein expression data, the oxidatively modified forms of LDL had more pronounced effects on the gene expression of both fibronectin and laminin. These data show that both LDL and, perhaps more importantly, its oxidatively modified forms stimulate mesangial cells to upregulate both the gene expression and synthesis and secretion of ECM proteins, supporting a role for atherogenic lipoproteins in the pathobiology of glomerular injury.


Subject(s)
Fibronectins/biosynthesis , Glomerular Mesangium/metabolism , Laminin/biosynthesis , Lipoproteins, LDL/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Gene Expression , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Humans , Mice , Oxidation-Reduction , RNA, Messenger/genetics
15.
Arterioscler Thromb Vasc Biol ; 18(6): 999-1006, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9633943

ABSTRACT

Estrogen therapy increases plasma HDL levels, which may reduce cardiovascular risk in postmenopausal women. The mechanism of action of estrogen in influencing various steps in hepatic HDL and apolipoprotein (apo) A-I synthesis and secretion are not fully understood. In this study, we have used the human hepatoblastoma cell line (Hep G2) as an in vitro model system to delineate the effect of estradiol on multiple regulatory steps involved in hepatic HDL metabolism. Incubation of Hep G2 cells with estradiol resulted in the following statistically significant findings: (1) increased accumulation of apoA-I in the medium without affecting uptake/removal of radiolabeled HDL-protein; (2) accelerated incorporation of [3H]leucine into apoA-I; (3) selective increase in [3H]leucine incorporation into lipoprotein (LP) A-I but not LP A-I+A-II HDL particles (HDL particles without and with apoA-II, respectively); (4) increased ability of apoA-I-containing particles to efflux cholesterol from fibroblasts; (5) stimulated steady state apoA-I but not apoA-II mRNA expression; and (6) increased newly transcribed apoA-I mRNA message without effect on apoA-I mRNA half-life. The data indicate that estradiol stimulates newly transcribed hepatic apoA-I mRNA, resulting in a selective increase in LP A-I, a subfraction of HDL that is associated with decreased atherosclerotic cardiovascular disease, especially in premenopausal women.


Subject(s)
Apolipoprotein A-II/genetics , Apolipoprotein A-I/genetics , Estradiol/pharmacology , Lipoproteins, HDL , Pregnancy Proteins , Transcription, Genetic/drug effects , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-II/biosynthesis , Carrier Proteins/metabolism , Humans , RNA, Messenger , Racemases and Epimerases , Tumor Cells, Cultured
16.
J Lab Clin Med ; 131(5): 456-65, 1998 May.
Article in English | MEDLINE | ID: mdl-9605111

ABSTRACT

Mesangial cell proliferation and extracellular matrix accumulation are fundamental in the pathogenesis of glomerulosclerosis. Platelet-derived growth factor (PDGF) is a major cytokine involved in mesangial cell proliferation, and its increased expression is seen in glomerular injury. Atherogenic lipoproteins stimulate mesangial cell proliferation and induce glomerular injury in experimental animals. We examined the effect of low-density lipoprotein (LDL) and its more atherogenic oxidized forms, minimally modified LDL (mm-LDL) and oxidized LDL (ox-LDL) on mesangial cell PDGF mRNA expression. Incubation with 2.5 to 25 microg/ml LDL or mm-LDL for 1 to 4 hours stimulated mesangial cell PDGF mRNA expression (mm-LDL 2 to 3 times greater than LDL); ox-LDL had no effect. Similarly, both LDL and mm-LDL induced mesangial cell DNA synthesis (mm-LDL 1.5 to 2 times greater). In further studies evaluating key associated intracellular signal transduction mechanisms, the protein tyrosine kinase (PTK) inhibitors herbimycin and genistein markedly decreased basal and lipoprotein-induced PDGF mRNA expression. Both pertussis toxin and isoproterenol, cyclic AMP-generating substances, stimulated PDGF mRNA expression. Preincubation with H-8 or H-89, cyclic AMP-dependent protein kinase A (PKA) inhibitors, blocked the lipoprotein-induced PDGF message, whereas preincubation with calphostin C, a protein kinase C inhibitor, did not alter LDL- or mm-LDL-mediated PDGF mRNA expression. These data suggest that the accumulation of atherogenic lipoproteins and their endogenous oxidized forms within the glomerulus may regulate mesangial cell PDGF expression and related cellular responses. These events appear to be modulated by signal transduction pathways involving PTK and PKA.


Subject(s)
Arteriosclerosis/etiology , Cyclic AMP-Dependent Protein Kinases/physiology , Glomerular Mesangium/metabolism , Lipoproteins/physiology , Platelet-Derived Growth Factor/metabolism , Protein-Tyrosine Kinases/physiology , Animals , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA/biosynthesis , Enzyme Inhibitors/pharmacology , Glomerular Mesangium/cytology , Humans , Lipoproteins, LDL/pharmacology , Mice , Platelet-Derived Growth Factor/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/metabolism
17.
J Am Soc Nephrol ; 9(3): 488-96, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9513913

ABSTRACT

Previously, it has been shown that atherogenic lipoproteins, through the activation of glomerular cells, stimulate pathobiological processes involved in monocyte infiltration into the mesangium. This study examined the role of LDL and its oxidatively modified variants (mildly oxidatively modified LDL [mm-LDL] and oxidatively modified LDL [ox-LDL]) on the activation of mesangial cell p42 mitogen-activated protein kinase (MAP kinase), a key intracellular signaling mechanism associated with cell proliferation. The incubation of mesangial cells with either LDL, mm-LDL, or ox-LDL induced the activation of MAP kinase dose dependently. The activation of MAP kinase by these lipoproteins in mesangial cells occurred biphasically: initially at 15 min of incubation period and at later time points of 8 to 24 h. No activation of MAP kinase was noted between 30 min (except in LDL) and 6 h. The induction of MAP kinase by both mm-LDL and ox-LDL was greater by 1.5- to 2-fold when compared with LDL. Similarly, these atherogenic lipoproteins stimulated mesangial cell proliferation. Lysophosphatidylcholine, a component of both oxidatively modified variants of LDL, markedly stimulated mesangial cell MAP kinase activity at early incubation times (5 to 30 min) but not at later time points (3 to 24 h), suggesting that lysophosphatidylcholine may, at least in part but not solely, act as an active component of ox-LDL-mediated effects. These data define putative key signal transduction events associated with lipoprotein-mediated induction of mesangial cell proliferation.


Subject(s)
Lipoproteins, LDL/pharmacology , Mitogen-Activated Protein Kinase 1/drug effects , Animals , Arteriosclerosis/chemically induced , Cell Division/drug effects , Cell Line , Dose-Response Relationship, Drug , Glomerular Mesangium/cytology , Glomerular Mesangium/enzymology , Lipoproteins, LDL/administration & dosage , Lysophosphatidylcholines/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Oxidation-Reduction , Platelet-Derived Growth Factor/pharmacology , Precipitin Tests , Time Factors
18.
Am J Nephrol ; 18(1): 83-8, 1998.
Article in English | MEDLINE | ID: mdl-9481446

ABSTRACT

Tubulointerstitial changes, characterized by the accumulation of extracellular matrix proteins (ECM) and fibrosis, are often associated with primary glomerular injury. Furthermore, these changes may be better prognostic indicators for decline in renal function than the anatomical changes seen within the glomerulus itself. Although hyperlipidemia and the increased renal accumulation of atherogenic lipoproteins are commonly seen in both human and experimental models of renal disease, the possible role that atherogenic lipoproteins may play in the cellular and molecular events associated with the development of tubulointerstitial injury remains unclear. Since atherogenic lipoproteins have been shown to be mediators of renal injury, we examined the effects of native LDL and oxidatively-modified LDL (ox-LDL, a more atherogenic form of LDL) on fibronectin protein synthesis and gene expression in proximal tubular epithelial cells (TEC). Human LDL was freshly isolated and ox-LDL prepared by incubation of LDL with 100 microM CuS04. Incubation of TEC with LDL or ox-LDL (25-50 micrograms/ml) for 24 h increased the steady-state mRNA expression of fibronectin by 16-135% over control as measured by Northern blot analysis and the effect was greater with ox-LDL than native LDL. Additional studies were done to examine whether the increased fibronectin message in response to lipoprotein activation was translated into TEC protein synthesis. The activation of TEC by LDL or ox-LDL stimulated the synthesis and secretion of fibronectin (52-150%, over control) as measured by Western blot analysis. The data show that LDL and ox-LDL stimulate TEC fibronectin gene message and protein synthesis supporting a pathobiological role for these atherogenic lipoproteins in tubulointerstitial fibrosis.


Subject(s)
Fibronectins/biosynthesis , Gene Expression , Kidney Tubules, Proximal/metabolism , Lipoproteins, LDL/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cell Line, Transformed , Epithelial Cells/metabolism , Fibronectins/genetics , Humans , Kidney Cortex , Kidney Tubules, Proximal/cytology , Mice , Nephritis, Interstitial , Oxidation-Reduction , RNA, Messenger/analysis
19.
Histol Histopathol ; 13(1): 169-79, 1998 01.
Article in English | MEDLINE | ID: mdl-9476647

ABSTRACT

The association between hyperlipidemia and renal disease was noted by Virchow as early as the 19th century. Subsequently, similar histopathological lipid depositions were confirmed-in diverse human and experimental renal disease. Although, no studies have been established in man to suggest a causal relationship between lipids and the pathogenesis of renal disease, compelling evidence accumulated in experimental animals suggests a direct role of lipids in the initiation and progression of glomerular disease. These studies showed that cholesterol-feeding to various experimental animals induced the development of glomerular injury. Furthermore, the treatment of hyperlipidemic animals with lipid lowering drugs prevented the development of glomerulosclerosis. In this article, we will review recent advances made in understanding various aspects of lipid-mediated renal injury including biochemical mechanisms of hyperlipidemia, a possible direct role of hyperlipidemia in the pathogenesis of renal disease, pathobiological accumulation of lipids and lipoproteins, biochemical and histological similarities between systemic atherosclerosis and glomerulosclerosis, and cellular processes involved in the development of glomerular disease. Furthermore, we will define cellular and molecular hypotheses that provide putative mechanisms by which hyperlipidemia and atherogenic lipoproteins induce series of cytoregulatory peptide-mediated events involved in the development of glomerular disease.


Subject(s)
Hyperlipidemias/complications , Hyperlipidemias/pathology , Kidney Diseases/complications , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Animals , Cholesterol, Dietary , Humans , Kidney Tubules/pathology
20.
Arterioscler Thromb Vasc Biol ; 17(10): 2020-8, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9351367

ABSTRACT

Niacin (nicotinic acid) is the most potent clinically used agent for increasing plasma HDL and apolipoprotein (apo) A-I. The mechanism by which niacin increases apoA-I is not clearly understood. We have examined the effect of niacin on the hepatic production and removal of apoA-I using Hep G2 cells as an in vitro model. Incubation of Hep G2 cells with niacin resulted in increased accumulation of apoA-I in the medium in a dose-dependent manner. Incorporation of [3H]leucine and [35S]methionine into apoA-I and apoA-I mRNA expression was unchanged by niacin, suggesting that it did not affect apoA-I de novo synthesis. Uptake of radiolabeled HDL protein and HDL apoA-I by Hep G2 cells was significantly reduced to as much as 82.9 +/- 2.2% (P = .04) and 84.2 +/- 2.8% (P = .02), respectively, of the baseline with increasing concentrations of niacin (0 to 3.0 mmol/L). Specific 125I-HDL protein uptake measured with a 50-fold excess of unlabeled HDL was reduced to as much as 78.3 +/- 4.8% (P = .005) in niacin-treated cells. The uptake of labeled cholesterol esters in HDL was unaffected by niacin. Niacin also effected a similar decrease in HDL protein uptake, but not cholesterol esters, from apoA-I-containing HDL particles isolated by immunoaffinity. The conditioned medium obtained from Hep G2 cells incubated with niacin significantly (P = .002) increased cholesterol efflux from cultured human fibroblasts. These data indicate a novel mechanism whereby niacin selectively decreases hepatic removal of HDL apoA-I but not cholesterol esters, thereby increasing the capacity of retained apoA-I to augment reverse cholesterol transport.


Subject(s)
Apolipoprotein A-I/metabolism , Cholesterol Esters/metabolism , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Niacin/pharmacology , Biological Transport , Cells, Cultured , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...