Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Lipids ; 55(2): 193-198, 2020 03.
Article in English | MEDLINE | ID: mdl-32065674

ABSTRACT

HepG2 cells were incubated with a 16.5:1.7:1 ratio of cholesterol:sitosterol:campesterol (CSC), a ratio of the major sterols observed in the plasma of phytosterolemia patients, or with cholesterol alone in combination with [14 C]acetate for 24 h and the radioactivity incorporated into lipids determined. Cells incubated with CSC exhibited a 40% reduction in cholesterol esterification (p < 0.05) compared to cells incubated with cholesterol alone. In addition, a 17.5-fold reduction (p < 0.05) in total cholesterol (cholesterol plus cholesteryl ester) synthesis from [14 C]acetate was observed in cells incubated with CSC compared to cholesterol alone. Low-density lipoprotein receptor (LDLR) mRNA abundance was lower in cells incubated with CSC compared to cells incubated with cholesterol alone. Our results suggest that incubation of HepG2 cells with a ratio of sterols that mimic the plasma concentration seen in phytosterolemia patients reduces cholesterol esterification, total cholesterol synthesis, and inhibits LDLR mRNA abundance. We suggest that future cell and animal-based work on phytostosterolemia might employ this methodology to serve as a novel paradigm of the disease.


Subject(s)
Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Hypercholesterolemia/genetics , Intestinal Diseases/genetics , Lipid Metabolism, Inborn Errors/genetics , Phytosterols/adverse effects , Receptors, LDL/genetics , Sitosterols/pharmacology , Carbon Radioisotopes/pharmacology , Cholesterol Esters/metabolism , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Hypercholesterolemia/metabolism , Intestinal Diseases/metabolism , Lipid Metabolism, Inborn Errors/metabolism , Models, Biological , Phytosterols/genetics , Phytosterols/metabolism , Phytosterols/pharmacology
2.
Clin Invest Med ; 41: 26-28, 2018 11 03.
Article in English | MEDLINE | ID: mdl-30394277

ABSTRACT

Patrick obtained his undergraduate degree at McGill University, and received his graduate training in Medicine and Biochemistry at the University of North Dakota. His postdoctoral work was conducted at the University of British Columbia. He joined the Faculty of Medicine, University of Manitoba as an Assistant Professor of Biochemistry in 1979. He was the recipient of a New Investigator Award (1979-85) from the Heart and Stroke Foundation of Canada, followed by a Scientist Award (1985-90) from the MRC/CIHR. These career awards allowed him to devote the majority of his time to research. He was promoted to Associate Professor in 1983 and Full Professor in 1986. He was cross-appointed as Professor of Pathology and participated in the teaching of General Pathology to residents. He was the founding Director of the Centre for Re-search and Treatment of Atherosclerosis at the University of Manitoba and the Winni-peg Health Sciences Centre. Administratively, he was appointed as the Professor and Head of Biochemistry and Medical Genetics (1992-1999), and an Associate Dean in the Faculty of Medicine in 1999 until his retirement in 2010.Professionally, Patrick served as President of the Canadian Biochemical Society, Chair of the Medical Research Council/Canadian Institute of Health Research New Investigator Awards Committee, Chair of the Alberta Heritage Foundation Senior Investigator Awards Committee, Vice-President of the Heart and Stroke Foundation of Manitoba, and Secretary/Treasurer of the Canadian Society for Clinical Investigation. He continues to serve on Editorial Boards of scientific journals, including the Journal of Clinical and Investigative Medicine. He was invested into the Order of Manitoba in 2011.


Subject(s)
Biomedical Research , Canada , Humans , Lipid Metabolism , Lipids/chemistry , Research Personnel
3.
PLoS One ; 7(11): e48628, 2012.
Article in English | MEDLINE | ID: mdl-23152787

ABSTRACT

Cardiolipin (CL) is a mitochondrial membrane phospholipid which plays a key role in apoptosis and supports mitochondrial respiratory chain complexes involved in the generation of ATP. In order to facilitate its role CL must be remodeled with appropriate fatty acids. We previously identified a human monolysocardiolipin acyltransferase activity which remodels CL via acylation of monolysocardiolipin (MLCL) to CL and was identical to the alpha subunit of trifunctional protein (αTFP) lacking the first 227 amino acids. Full length αTFP is an enzyme that plays a prominent role in mitochondrial ß-oxidation, and in this study we assessed the role, if any, which this metabolic enzyme plays in the remodeling of CL. Purified human recombinant αTFP exhibited acyl-CoA acyltransferase activity in the acylation of MLCL to CL with linoleoyl-CoA, oleoyl-CoA and palmitoyl-CoA as substrates. Expression of αTFP increased radioactive linoleate or oleate or palmitate incorporation into CL in HeLa cells. Expression of αTFP in Barth Syndrome lymphoblasts, which exhibit reduced tetralinoleoyl-CL, elevated linoleoyl-CoA acylation of MLCL to CL in vitro, increased mitochondrial respiratory Complex proteins and increased linoleate-containing species of CL. Knock down of αTFP in Barth Syndrome lymphoblasts resulted in greater accumulation of MLCL than those with normal αTFP levels. The results clearly indicate that the human αTFP exhibits MLCL acyltransferase activity for the resynthesis of CL from MLCL and directly links an enzyme of mitochondrial ß-oxidation to CL remodeling.


Subject(s)
Cardiolipins/metabolism , Multienzyme Complexes/metabolism , Acyl Coenzyme A/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Sequence , Animals , Cell Line , Enzyme Activation , Fatty Acids/metabolism , Gene Expression , Gene Expression Regulation/drug effects , Humans , Lysophospholipids/metabolism , Male , Mitochondrial Trifunctional Protein , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Oxidation-Reduction , Palmitoyl Coenzyme A/metabolism , Protein Binding , RNA Interference , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Substrate Specificity , Thyroxine/pharmacology
4.
Lipids ; 46(9): 795-804, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21710238

ABSTRACT

ATP binding cassette A1 (ABCA1) transports cholesterol, phospholipids and lipophilic molecules to and across cellular membranes. We examined if ABCA1 expression altered cellular de novo glycerolipid biosynthesis in growing Baby hamster kidney (BHK) cells. Mock BHK cells or cells expressing a mifepristone-inducible ABCA1 (ABCA1) were incubated plus or minus mifepristone and then with [(3)H]serine or [(3)H]inositol or [(3)H]ethanolamine or [methyl-(3)H]choline or [(3)H]glycerol or [(14)C]oleate and radioactivity incorporated into glycerolipids determined. Mifepristone did not affect [1,3-(3)H]glycerol or [(14)C]oleate or [(3)H]ethanolamine or [methyl-(3)H]choline uptake in BHK cells. In contrast, [(3)H]glycerol and [(14)C]oleate incorporated into phosphatidylserine (PtdSer) were elevated 2.4-fold (p < 0.05) and 54% (p < 0.05), respectively, upon ABCA1 induction confirming increased PtdSer biosynthesis from these precursors. However, mifepristone inhibited [(3)H]serine uptake and incorporation into PtdSer indicating that PtdSer synthesis from serine in BHK cells is dependent on serine uptake. Mifepristone stimulated [(3)H]inositol uptake in mock and ABCA1 cells but not its incorporation into phosphatidylinositol indicating that its synthesis from inositol is independent of inositol uptake in BHK cells. [(3)H]glycerol and [(14)C]oleate incorporated into triacylglycerol were reduced and into diacylglycerol elevated only in mifepristone-induced ABCA1 expressing cells due to a decrease in diacylglycerol acyltransferase-1 (DGAT-1) activity. The presence of trichostatin A, a class I and II histone deacetylase inhibitor, reversed the ABCA1-mediated reduction in DGAT-1 activity but did not affect DGAT-1 mRNA expression. Thus, mifepristone has diverse effects on de novo glycerolipid synthesis. We suggest that caution should be exercised when using mifepristone-inducible systems for studies of glycerolipid metabolism in cells expressing glucocorticoid responsive receptors.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Gene Expression/drug effects , Glycerides/biosynthesis , Mifepristone/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line , Cricetinae , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays , Glycerol/metabolism , Glycerophospholipids/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inositol/metabolism , Oleic Acid/metabolism , Phosphatidylserines/biosynthesis , Serine/metabolism
5.
Can J Physiol Pharmacol ; 88(11): 1084-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21076496

ABSTRACT

Ischemia-reperfusion (IR)-induced cell apoptosis involves the activation of c-Jun NH2-terminal kinase (JNK). The activation of JNK requires the presence of scaffold proteins called JNK-interacting proteins (JIP), which bind several members of a signaling cascade for proper signaling specificity. In this study, the expression of scaffold proteins JIP1 and JIP3 and their roles in the regulation of JNK activity were investigated in simulated IR in a cell model (H9c2). JIP1 protein expression was significantly decreased, whereas JIP3 protein expression was increased in IR H9c2 cells. Adenovirus-induced overexpression of JIP1 reduced IR-induced JNK activity and apoptosis. Conversely, overexpression of JIP3 increased JNK activity and apoptosis following IR. Depletion of endogenous JIP1 by siRNA treatment increased the IR-induced JNK activity, whereas siRNA-mediated depletion of endogenous JIP3 inhibited JNK activity. These results suggest that JIP1 and JIP3 play important roles in the activation of JNK during simulated IR challenge in H9c2 cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Myocardial Ischemia/enzymology , Myocardial Reperfusion , Nerve Tissue Proteins/physiology , Adaptor Proteins, Signal Transducing/analysis , Animals , Apoptosis , Cell Line , Enzyme Activation , Nerve Tissue Proteins/analysis , RNA Interference , Rats
6.
Biochem Cell Biol ; 86(5): 405-15, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18923542

ABSTRACT

Protein-protein interaction plays a key role in the regulation of biological processes. The human potassium (HERG) channel is encoded by the ether-à-go-go-related gene (herg), and its activity may be regulated by association with other cellular proteins. To identify cellular proteins that might play a role in the regulation of the HERG channel, we screened a human heart cDNA library with the N terminus of HERG using a yeast 2-hybrid system, and identified caveolin-1 as a potential HERG partner. The interaction between these 2 proteins was confirmed by coimmunoprecipitation assay, and their overlapping subcellular localization was demonstrated by fluorescence immunocytochemistry. The physiologic implication of the protein-protein interaction was studied in whole-cell patch-clamp electrophysiology experiments. A significant increase in HERG current amplitude and a faster deactivation of tail current were observed in HEK293/HERG cells in a membrane lipid rafts disruption model and caveolin-1 knocked down cells by RNA interference. Alternatively, when caveolin-1 was overexpressed, the HERG current amplitude was significantly reduced and the tail current was deactivated more slowly. Taken together, these data indicate that HERG channels interact with caveolin-1 and are negatively regulated by this interaction. The finding from this study clearly demonstrates the regulatory role of caveolin-1 on HERG channels, and may help to understand biochemical events leading to arrhythmogenesis in the long QT syndrome in cardiac patients.


Subject(s)
Caveolin 1/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Heart/physiology , Animals , Caveolin 1/genetics , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Humans , Long QT Syndrome/metabolism , Membrane Microdomains , Patch-Clamp Techniques , RNA Interference , Rats , Rats, Wistar , Two-Hybrid System Techniques
7.
FEBS J ; 275(18): 4531-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18680509

ABSTRACT

Protein-protein interactions are critical for protein trafficking, localization and the regulation of ion channels. The human ether-a-go-go-related gene (herg) encodes the alpha-subunit of the potassium channel underlying the rapid component of the cardiac delayed rectifier current. To identify the cellular proteins involved in the regulation of the HERG channel, a human heart cDNA library was screened using a yeast two-hybrid system, with the N-terminus of HERG as bait. The four and a half LIM domain protein 2 (FHL2) was identified as a potential HERG partner. The interaction between these two proteins was confirmed by co-immunoprecipitation and glutathione transferase pull-down assays and immunocytochemical analysis. The physiological implication of HERG-FHL2 interaction, assessed by whole-cell, patch-clamp electrophysiology experiments, showed a significant increase in the HERG current amplitude and a faster deactivation of the tail current in human embryonic kidney 293 cells co-expressing HERG and FHL2. These data indicate that FHL2 interacts with and regulates the HERG channel. Our findings may aid in the further understanding of the molecular basis of HERG channel diversity and arrhythmogenesis in the long-QT syndrome.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels/analysis , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Humans , Immunoprecipitation , LIM-Homeodomain Proteins , Muscle Proteins/analysis , Muscle Proteins/genetics , Patch-Clamp Techniques , Rats , Recombinant Fusion Proteins/metabolism , Transcription Factors/analysis , Transcription Factors/genetics , Two-Hybrid System Techniques
8.
Mol Cell Biochem ; 311(1-2): 37-44, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18165926

ABSTRACT

Soy consumption is associated with a lower risk of atherosclerotic disease in the oriental population. Genistein is a soy isoflavone bearing estrogenic properties. Previous experiments in our laboratory demonstrated the potentiation of endothelium-independent relaxation of coronary artery by both estrogen and genistein. The potentiating effects of both estrogen and genistein were mediated through the cAMP-signaling pathway. We hypothesize that genistein could enhance protein kinase A (PKA) activity in porcine coronary artery smooth muscle, thereby offering a mechanism for the potentiation of vascular relaxation by genistein. In our study, a high concentration of genistein (10(-4.5) M) significantly increased PKA activity in porcine coronary artery rings. While genistein at 10(-5.5) M and forskolin at 10(-7) M had no effect on PKA activity, the combination of the two compounds at the prescribed concentrations caused a significant increase in PKA activity. The increase in PKA activity by genistein was abolished by SQ 22536 (adenylate cyclase blocker), but not by NF 449 (Gs protein blocker) or ICI 182780 (estrogen receptor antagonist). Our results suggest that the action of genistein is mediated via adenylate cyclase, but does not appear to involve Gs protein or ICI 182780-sensitive estrogen receptor.


Subject(s)
Coronary Vessels/metabolism , Genistein/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinase Inhibitors/metabolism , Adenine/analogs & derivatives , Adenine/metabolism , Adenine/pharmacology , Animals , Benzenesulfonates/metabolism , Benzenesulfonates/pharmacology , Colforsin/metabolism , Coronary Vessels/anatomy & histology , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Genistein/pharmacology , Humans , Muscle, Smooth, Vascular/drug effects , Protein Kinase Inhibitors/pharmacology , Swine
9.
Can J Physiol Pharmacol ; 85(11): 1053-62, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18066107

ABSTRACT

The activation of c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in the induction of cell apoptosis. We previously reported that magnesium tanshinoate B (MTB), a compound purified from a Chinese herb danshen (Salvia miltiorrhiza), could inhibit ischemia/reperfusion-induced myocyte apoptosis in the heart. The objective of the present study was to investigate whether MTB can prevent oxidized lipoprotein-induced apoptosis in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were incubated with copper-oxidized very low density lipoprotein (Cu-OxVLDL) or copper-oxidized low density lipoprotein (Cu-OxLDL). Treatment of cells with Cu-OxVLDL or Cu-OxLDL resulted in a 3-fold increase in the JNK activity. The amount of cytochrome c released and the activity of caspase-3 in cells treated with Cu-OxVLDL or Cu-OxLDL were significantly elevated, indicating the occurrence of apoptosis. The presence of MTB was able to abolish the JNK activation, cytochrome c release, and caspase-3 activation induced by Cu-OxVLDL or Cu-OxLDL, resulting in a marked reduction in apoptosis in endothelial cells. The data from this study indicate that oxidized lipoproteins induce apoptosis in endothelial cells. We postulate that the inhibition of the JNK signaling pathway by MTB is a key mechanism that protects these cells from oxidized lipoprotein-induced apoptosis.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Lipoproteins, LDL/toxicity , Magnesium/pharmacology , Phenanthrolines/pharmacology , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/metabolism , Endothelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism
10.
J Lipid Res ; 47(6): 1140-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16547353

ABSTRACT

Cardiolipin (CL) is a phospholipid localized to the mitochondria, and its biosynthesis is essential for mitochondrial structure and function. We report here the identification and characterization of a cDNA encoding the first mammalian cardiolipin synthase (CLS1) in humans and mice. This cDNA exhibits sequence homology with members of a CLS gene family that share similar domain structure and chemical properties. Expression of the human CLS (hCLS1) cDNA in reticulocyte lysates or insect cells led to a marked increase in CLS activity. The enzyme is specific for CL synthesis, because no significant increase in phosphatidylglycerol phosphate synthase activity was observed. In addition, CL pool size was increased in hCLS1-overexpressing cells compared with controls. Furthermore, the hCLS1 gene was highly expressed in tissues such as heart, skeletal muscle, and liver, which have been shown to have high CLS activities. These results demonstrate that hCLS1 encodes an enzyme that synthesizes CL.


Subject(s)
Cloning, Molecular/methods , DNA, Complementary/genetics , Membrane Proteins/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Amino Acid Sequence , Animals , Cell Line , Chromosome Mapping , DNA, Complementary/chemistry , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Rats , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Spodoptera , Substrate Specificity , Transferases (Other Substituted Phosphate Groups)/metabolism
11.
Mol Cell Biochem ; 275(1-2): 165-71, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16335796

ABSTRACT

Nitric oxide (NO) is a principal mediator in many physiological and pathological processes. Overproduction of NO via the inducible nitric oxide synthase (iNOS) has cytotoxic effect through the formation of peroxynitrite with superoxide anion. The iNOS is mainly expressed in macrophages and is able to produce large amount of NO. The expression of iNOS is mainly regulated at the transcriptional level. The iNOS-mediated NO production plays a role in the development of atherosclerosis. Ganoderma lucidum (G. lucidum, Linzhi or Reishi) is a traditional herbal medicine which is commonly used as health supplement. Several studies have demonstrated its effectiveness against cancer, immunological disorders and cardiovascular diseases. The objective of the present study was to investigate the effect of G. lucidum on iNOS-mediated NO production in macrophages. Human monocytic cell (THP-1) derived macrophages were incubated with lipopolysaccharide (LPS) for 24 h. Such treatment significantly stimulated NO production (253% versus the control). Such a stimulatory effect was resulted from increased iNOS mRNA expression (270% versus the control) and iNOS activity (169.5% versus the control) in macrophages. The superoxide anion level was also elevated (150% versus the control) in LPS-treated macrophages. Treatment of macrophages with G. lucidum extract (100 microg/ml) completely abolished LPS-induced iNOS mRNA expression and NO production. Such an inhibitory effect of G. lucidum was mediated via its antioxidant action against LPS-induced superoxide anion generation in macrophages. These results suggest that G. lucidum may exert a therapeutic effect against atherosclerosis via ameliorating iNOS-mediated NO overproduction in macrophages.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression/drug effects , Macrophages/drug effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Reishi/chemistry , Antioxidants/pharmacology , Blotting, Western , Cell Culture Techniques , Cell Line , Humans , Macrophages/enzymology , Nitrates/analysis , Nitrates/metabolism , Nitric Oxide Synthase Type II/genetics , Nitrites/analysis , Nitrites/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Superoxides/analysis , Superoxides/metabolism , Tetradecanoylphorbol Acetate/pharmacology
12.
Can J Physiol Pharmacol ; 83(4): 321-34, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15877107

ABSTRACT

Traditional Chinese medicine (TCM) has many beneficial effects and has been practiced for several thousand years. It is known to treat the cause of a disease rather than to alleviate its symptoms. Based on a belief that TCM is natural, safe, and of lower cost, consumers worldwide are spending more out-of-pocket money on this form of therapy. This increased spending, and reports of adverse reactions, has drawn the attention of many regulatory agencies. Scientists have called for more evidence-based and scientific research on the risks and benefits of TCM. In Canada, the Natural Health Product Regulations came into effect January 2004. TCM herbal product manufacturers will need to provide products of reputable quality to the market. Many will apply modern technology and good science to support their products. The issues facing producers, scientists, and consumers alike are quality control and assessment, standardization of bioactive components, mechanisms of actions, and integration of the evolved modern Chinese medicine into the healthcare system. Solid science, better regulation of the final product, and better education of consumers are necessary to extract the best of TCM to complement existing conventional medicine to deliver the best healthcare.


Subject(s)
Medicine, Chinese Traditional/trends , Animals , Chromatography , History, Ancient , Humans , Medicine, Chinese Traditional/history , Plants, Medicinal/chemistry , Quality Control
13.
Am J Physiol Endocrinol Metab ; 288(5): E1002-10, 2005 May.
Article in English | MEDLINE | ID: mdl-15644462

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for cardiovascular disorders. Elevated plasma homocysteine (Hcy) concentration is associated with other cardiovascular risk factors. We previously reported that Hcy stimulated cholesterol biosynthesis in HepG2 cells. In the present study, we investigated the underlying mechanisms of Hcy-induced hepatic cholesterol biosynthesis in an animal model. Hyperhomocysteinemia was induced in Sprague-Dawley rats by feeding a high-methionine diet for 4 wk. The mRNA expression and the enzyme activity of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase were significantly increased in livers of hyperhomocysteinemic rats. There were marked hepatic lipid accumulation and an elevation of plasma cholesterol concentration in hyperhomocysteinemic rats. Three transcription factors, namely, sterol regulatory element-binding protein-2 (SREBP-2), cAMP response element-binding protein (CREB), and nuclear factor Y (NF-Y) were activated in livers of hyperhomocysteinemic rats. Upon Hcy treatment of hepatocytes, there was a significant increase in HMG-CoA reductase mRNA expression in these cells. The activation of SREBP-2, CREB, and NF-Y preceded the increase in HMG-CoA reductase expression in Hcy-treated cells. Pretreatment of hepatocytes with inhibitors for transcription factors not only blocked the activation of SREBP-2, CREB, and NF-Y but also attenuated Hcy-induced HMG-CoA reductase mRNA expression. These results suggested that hyperhomocysteinemia-induced activation of SREBP-2, CREB, and NF-Y was responsible for increased cholesterol biosynthesis by transcriptionally regulating HMG-CoA reductase expression in the liver leading to hepatic lipid accumulation and subsequently hypercholesterolemia. In conclusion, the stimulatory effect of Hcy on hepatic cholesterol biosynthesis may represent an important mechanism for hepatic lipid accumulation and cardiovascular disorder associated with hyperhomocysteinemia.


Subject(s)
Acyl Coenzyme A/metabolism , Cholesterol/biosynthesis , Homocysteine/blood , Hyperhomocysteinemia/metabolism , Lipid Metabolism , Liver/metabolism , Transcription Factors/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
14.
Biochem J ; 385(Pt 2): 469-77, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15367102

ABSTRACT

AGPAT (1-acyl-sn-glycerol 3-phosphate acyltransferase) exists in at least five isoforms in humans, termed as AGPAT1, AGPAT2, AGPAT3, AGPAT4 and AGPAT5. Although they catalyse the same biochemical reaction, their relative function, tissue expression and regulation are poorly understood. Linkage studies in humans have revealed that AGPAT2 contributes to glycerolipid synthesis and plays an important role in regulating lipid metabolism. We report the molecular cloning, tissue distribution, and enzyme characterization of mAGPATs (murine AGPATs) and regulation of cardiac mAGPATs by PPARalpha (peroxisome-proliferator-activated receptor alpha). mAGPATs demonstrated differential tissue expression profiles: mAGPAT1 and mAGPAT3 were ubiquitously expressed in most tissues, whereas mAGPAT2, mAGPAT4 and mAGPAT5 were expressed in a tissue-specific manner. mAGPAT2 expressed in in vitro transcription and translation reactions and in transfected COS-1 cells exhibited specificity for 1-acyl-sn-glycerol 3-phosphate. When amino acid sequences of five mAGPATs were compared, three highly conserved motifs were identified, including one novel motif/pattern KX2LX6GX12R. Cardiac mAGPAT activities were 25% lower (P<0.05) in PPARalpha null mice compared with wild-type. In addition, cardiac mAGPAT activities were 50% lower (P<0.05) in PPARalpha null mice fed clofibrate compared with clofibrate fed wild-type animals. This modulation of AGPAT activity was accompanied by significant enhancement/reduction of the mRNA levels of mAGPAT3/mAGPAT2 respectively. Finally, mRNA expression of cardiac mAGPAT3 appeared to be regulated by PPARalpha activation. We conclude that cardiac mAGPAT activity may be regulated by both the composition of mAGPAT isoforms and the levels of each isoform.


Subject(s)
Acyltransferases/chemistry , Acyltransferases/genetics , Acyltransferases/physiology , Cloning, Molecular/methods , Myocardium/chemistry , Myocardium/metabolism , PPAR alpha/physiology , 1-Acylglycerol-3-Phosphate O-Acyltransferase , Acyltransferases/biosynthesis , Amino Acid Sequence , Animals , COS Cells/chemistry , COS Cells/metabolism , Carbon Radioisotopes/metabolism , Chlorocebus aethiops , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Molecular Sequence Data , Myocardium/enzymology , PPAR alpha/deficiency , Phosphatidic Acids/chemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Sequence Alignment/methods
15.
Mol Cell Biochem ; 262(1-2): 101-10, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15532714

ABSTRACT

Prostaglandins (PGs) are potent eicosanoid lipid mediators that have been implicated in numerous homeostatic functions and inflammation. Estrogens have been shown to regulate the expression of genes in lipid metabolism in many cellular systems. In this study, the activation of macrophages and the modulation of PG release by estrogens were examined. The effects of 17-alpha and 17-beta estradiols, phytoestrogen Genistein and several selective estrogen receptor modulators on the release of PGE2 were investigated in human U937-derived macrophages. 17-Beta estradiol caused an enhancement of PGE2 production in a time- and dose-dependent manner. Treatment of macrophages with 17-beta estradiol elicited an increased arachidonic acid (AA) release and an up-regulation of both cyclooxygenesis-1 and cyclooxygenesis-2 enzymes at both the transcript and protein levels. In addition, immunostaining of nuclear estrogen receptor alpha and the observation of ICI182 780 blockade of PGE2 production indicated that 17-beta estradiol-induced PGE2 release was mainly through nuclear estrogen receptor alpha.


Subject(s)
Dinoprostone/biosynthesis , Estradiol/pharmacology , Macrophages/drug effects , Arachidonic Acid/metabolism , Cell Differentiation , Cell Line , Cyclooxygenase 1 , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Estrogen Receptor alpha , Humans , Ligands , Macrophages/metabolism , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/analysis , RNA/analysis , Up-Regulation
16.
Biochem Cell Biol ; 82(5): 569-76, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15499385

ABSTRACT

Tafazzins, a group of proteins that are defective in patients with Barth syndrome, are produced by alternate splicing of the gene G4.5 or TAZ. RT-PCR and transcription-coupled in vitro translation analysis were undertaken to determine the expression of alternatively spliced TAZ mRNA in mouse tissues and human cell lines. Only two tafazzin transcripts, both lacking exon 5, were expressed in murine tissues, whereas four tafazzin transcripts, all lacking exon 5, were observed in human umbilical vein vascular endothelial cells and U937 human monoblasts indicating a species-specific difference in the expression of TAZ mRNAs in mouse and humans. Only TAZ lacking exon 5 was expressed in murine heart. Differentiation of U937 human monoblasts into macrophages did not alter expression of the tafazzin transcripts indicating that TAZ expression is independent of monocyte differentiation. Cloning and in vitro expression of both murine and human tafazzin cDNA revealed two prominent protein bands that corresponded to the expected sizes of alternative translation. A novel fifth motif, identified as critical for the glycerolphosphate acyltransferase family, was observed in human tafazzin. The presence of a mutation in this region in Barth syndrome patients indicates that this motif is essential for tafazzin function.


Subject(s)
Gene Expression Regulation/physiology , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Macrophages/metabolism , Proteins/metabolism , Transcription Factors/metabolism , Acyltransferases , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Regulation/genetics , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Humans , Macrophages/cytology , Mice , Molecular Sequence Data , Mutation/genetics , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Distribution , Transcription Factors/genetics
17.
Biochem Cell Biol ; 82(1): 212-24, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15052339

ABSTRACT

Atherosclerosis is the leading cause of death in North America and within the next two decades will be the leading cause worldwide. Atherosclerosis is characterized by vascular obstruction from the deposits of plaque, resulting in reduced blood flow. Plaque rupture and the consequent thrombosis may lead to sudden blockage of the arteries and cause heart attack. High serum lipid levels, especially the elevated level of low-density lipoprotein (LDL), have been shown to be strongly related to the development of atherosclerosis. It is generally accepted that atherosclerotic lesions are initiated via an enhancement of LDL uptake by monocytes and macrophages. In the liver, uptake of plasma LDL is mediated via specific LDL receptors, but a scavenger receptor system is employed by macrophages. Plasma LDL must be modified prior to uptake by macrophages. Analysis of the lipid content in the oxidatively modified LDL from hyper lipidemic patients revealed that the level of lysophosphatidylcholine was greatly elevated, and the high level of the lysolipid was shown to impair the endothelium-dependent relaxation of the blood vessels. In a separate study, we showed that a high level of homocysteine caused the increase in cholesterol production and apolipoprotein B-100 secretion in hepatic cells. Statins have been used effectively to control the production of cholesterol in the liver, and recently, ezetimibe has been shown to supplement the efficacy of statins by inhibiting cholesterol absorption. The factor of elevated levels of triglyceride-rich lipoproteins in association with depressed high-density lipoproteins, usually in the context of insulin resistance, is an important contributor to atherosclerosis and can be effectively treated with fibric acid derivatives. In hyperhomocysteinemia, folic acid supplements may have a role in the control of cholesterol by reducing the plasma homocysteine level.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Lipids/blood , Arteriosclerosis/etiology , Chemokines/genetics , Chemokines/metabolism , Cholesterol/metabolism , Endothelium, Vascular/metabolism , Homocysteine/blood , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hyperlipidemias/therapy , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/physiology , Risk Factors
18.
J Lipid Res ; 45(2): 244-52, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14594999

ABSTRACT

The role of peroxisome proliferator-activated receptor alpha (PPARalpha)-stimulated phospholipase A2 (PLA2) in cardiac mitochondrial cardiolipin (CL) biosynthesis was examined in both in vivo and in vitro models. Treatment of rat heart H9c2 cells with clofibrate increased the expression and activity of 14 kDa PLA2 but did not affect the pool size of CL. Clofibrate treatment stimulated de novo CL biosynthesis via an increase in phosphatidylglycerolphosphate (PGP) synthase activity, accounting for the unaltered CL content. Cardiac PLA2, PGP synthase, and CDP-1,2-diacyl-sn-glycerol synthase (CDS-2) activities and CDS-2 mRNA levels were elevated in mice fed clofibrate for 14 days compared with controls. In PPARalpha-null mice, clofibrate feeding did not alter cardiac PLA2, PGP synthase activities, or CDS-2 activity and mRNA level, confirming that these enzymes are regulated by PPARalpha activation. In contrast to mouse heart, clofibrate treatment did not affect the activity or mRNA levels of CDS-2 in H9c2 cells, indicating that CDS-2 is regulated differently in rat heart H9c2 cells in vitro and in mouse heart in vivo. These results clearly indicate that cardiac CL de novo biosynthesis is stimulated by PPARalpha activation in responsive rodent models and that CDS-2 is an example of an enzyme that exhibits alternative regulation in vivo and in cultured cell lines. This study is the first to demonstrate that CL de novo biosynthesis is regulated by PPARalpha activation.


Subject(s)
Cardiolipins/biosynthesis , Diacylglycerol Cholinephosphotransferase/genetics , Myocardium/metabolism , Phospholipases A/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , Anticholesteremic Agents/pharmacology , Cell Line , Clofibrate/pharmacology , Diacylglycerol Cholinephosphotransferase/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/metabolism , Myocardium/cytology , Phospholipases A2 , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/metabolism , Transferases (Other Substituted Phosphate Groups)
19.
Life Sci ; 73(10): 1253-63, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12850241

ABSTRACT

Hyperlipidemia, a condition normally observed in cholestatic liver disease, is also a risk factor for the development of atherosclerosis. The relationship between the elevation of lipoproteins in cholestatic liver diseases and atherosclerosis formation has not been elucidated. In this study, we propose that the impairment of endothelium-dependent relaxation (EDR) of blood vessels in cholestatic liver diseases may lead to the development of atherosclerosis. Using bile duct ligation (BDL) in rats as a model, we examined the liver function, serum lipid profile, EDR and morphologic change of the aorta from both sham operated and BDL rats. Significant increases in liver and spleen weights, serum alanine transaminase (ALT) and aspartate transaminase (AST) activities and the bilirubin level were observed in BDL rats. Upon bile duct ligation, the total and low-density lipoprotein cholesterol levels were increased but the high-density lipoprotein cholesterol and triglyceride levels were reduced. Less contractility and lowered response to acetylcholine-induced relaxation were found in aorta segments. In addition, the acetylcholine-induced relaxation was blocked by both L-NAME and 15 mM KCl. Our results suggest that both nitric oxide and endothelium-derived hyperpolarizing factor are important elements for the impairment of the EDR in BDL rats. In addition, a mild atrophy of the media of the aorta was detected in BDL rats. We conclude that the alterations of lipid profile and the mild atrophy of the media may lead to the impairment of EDR in the aorta in BDL rats, and these factors may potentiate the development of atherosclerosis.


Subject(s)
Aorta, Thoracic/physiopathology , Endothelium, Vascular/physiopathology , Lipids/blood , Liver Cirrhosis, Experimental/blood , Vasodilation , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Bile Ducts/surgery , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Antagonism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Ligation , Liver Cirrhosis, Experimental/complications , Male , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Organ Size , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , Vasodilation/physiology
20.
Mol Cell Biochem ; 246(1-2): 31-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12841340

ABSTRACT

Cytosolic phospholipases A2 (cPLA2) and cyclooxygenases-1 and -2 (COX-1 and -2) play a pivotal role in the metabolism of arachidonic acid (AA) and in eicosanoid production. The coordinate regulation and expression of these enzymes is not well defined. In this study, the effect of phorbol 12-myristate 13-acetate (PMA), tumor necrosis factor alpha (TNFalpha), lipopolysaccharide (LPS) and macrophage-colony stimulating factor (M-CSF) on AA release and prostaglandin E2 (PGE2) production and the expression of cPLA2 and COX-1 and -2 were investigated in U937 human pre-monocytic cells and fully differentiated macrophages. Treatment of U937 cells with PMA or macrophages with LPS increased AA release and PGE2 production. Incubation of U937 cells or macrophages for 8 h with all stimuli elevated cPLA2 expression. In contrast, cPLA2 expression was reduced upon further incubation of U937 cells or macrophages for 24 h with all stimuli indicating a bi-phasic expression pattern of this enzyme. PMA induced COX-1 expression in U937 cells whereas LPS induced COX-2 expression in macrophages. Although TNFalpha and M-CSF induced a significant amount of AA release in both cell models, they failed to induce a comparable production of PGE2 since they were unable to induce the coordinate expression of the downstream key enzymes, COX-1 or COX-2. The results suggest that the enhancement of AA release in both U937 cells and macrophages may be caused by both increased cPLA2 activity and elevated cPLA2 protein expression. In addition, PMA stimulates PGE2 production via up-regulation of COX-1, and likely COX-2, expression in U937 cells whereas LPS stimulates PGE2 production via induction of COX-2 expression in macrophages.


Subject(s)
Isoenzymes/metabolism , Macrophages/drug effects , Macrophages/enzymology , Monocytes/drug effects , Monocytes/enzymology , Phospholipases A/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Arachidonic Acid/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Cytosol/enzymology , Dinoprostone/biosynthesis , Humans , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Membrane Proteins , Phospholipases A2 , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...