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










Publication year range
2.
Oxid Med Cell Longev ; 2018: 3939714, 2018.
Article in English | MEDLINE | ID: mdl-30402205

ABSTRACT

Diabetes-associated cardiac fibrosis is a severe cardiovascular complication. Momordicine I, a bioactive triterpenoid isolated from bitter melon, has been demonstrated to have antidiabetic properties. This study investigated the effects of momordicine I on high-glucose-induced cardiac fibroblast activation. Rat cardiac fibroblasts were cultured in a high-glucose (25 mM) medium in the absence or presence of momordicine I, and the changes in collagen synthesis, transforming growth factor-ß1 (TGF-ß1) production, and related signaling molecules were assessed. Increased oxidative stress plays a critical role in the development of high-glucose-induced cardiac fibrosis; we further explored momordicine I's antioxidant activity and its effect on fibroblasts. Our data revealed that a high-glucose condition promoted fibroblast proliferation and collagen synthesis and these effects were abolished by momordicine I (0.3 and 1 µM) pretreatment. Furthermore, the inhibitory effect of momordicine I on high-glucose-induced fibroblast activation may be associated with its activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the inhibition of reactive oxygen species formation, TGF-ß1 production, and Smad2/3 phosphorylation. The addition of brusatol (a selective inhibitor of Nrf2) or Nrf2 siRNA significantly abolished the inhibitory effect of momordicine I on fibroblast activation. Our findings revealed that the antifibrotic effect of momordicine I was mediated, at least partially, by the inhibition of the TGF-ß1/Smad pathway, fibroblast proliferation, and collagen synthesis through Nrf2 activation. Thus, this work provides crucial insights into the molecular pathways for the clinical application of momordicine I for treating diabetes-associated cardiac fibrosis.


Subject(s)
Collagen/biosynthesis , Fibroblasts/metabolism , Fibroblasts/pathology , Glucose/toxicity , Myocardium/pathology , Sterols/pharmacology , Animals , Antioxidants/pharmacology , Cell Proliferation/drug effects , Fibroblasts/drug effects , Models, Biological , NF-E2-Related Factor 2/metabolism , Phosphorylation/drug effects , Quassins/pharmacology , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Sterols/chemistry , Transforming Growth Factor beta1/metabolism
3.
Nanotechnology ; 28(8): 085705, 2017 Feb 24.
Article in English | MEDLINE | ID: mdl-28102176

ABSTRACT

Molecularly imprinted polymer nanoparticles incorporating magnetic nanoparticles (MNPs) have been investigated for their selective adsorption properties. Here we describe the synthesis and characterization of magnetic cytosine-imprinted chitosan nanoparticles (CIPs) for gene delivery. In particular, CIPs carrying the mammalian expression plasmid of enhanced green fluorescent protein were prepared by the co-precipitation of MNPs, chitosan and a template nucleobase (cytosine). The results show that the selective reabsorption of cytosine to magnetic CIPs was at least double that of non-imprinted polymers and other nucleobases (such as adenine and thymine). The gene carrier CIPs were used for the transfection of human embryonic kidney 293 cells showing dramatic increase their efficiency with that of conventional chitosan nanoparticles. Furthermore, the gene carrier magnetic CIPs also exhibit low toxicity compared to that of commercially available cationic lipids.

4.
Pharmacology ; 98(1-2): 42-50, 2016.
Article in English | MEDLINE | ID: mdl-27050899

ABSTRACT

BACKGROUND: Nicorandil, a mitochondrial adenosine triphosphate-sensitive potassium (mitoKATP) channel opener, exerts protective effects on the cardiovascular system. This study examined the effect of nicorandil on cyclic strain-induced interleukin-8 (IL-8) expression in human umbilical vein endothelial cells (HUVECs). METHODS: Cultured HUVECs were exposed to cyclic strain in the presence or absence of nicorandil (1-10 µmol/l); we then analyzed IL-8 expression. We also assessed the effects of nicorandil on heme oxygenase-1 (HO-1) expression and cyclic strain-modulated IL-8 expression after HO-1 silencing in HUVECs. SUMMARY: HUVECs exposed to cyclic strain showed increased IL-8 messenger RNA expression and protein secretion. Nicorandil (1-10 µmol/l) inhibited cyclic strain-induced IL-8 expression, whereas 5-hydroxydecanoate (100 µmol/l), a selective inhibitor of the mitoKATP channel, completely reversed the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression. We demonstrated that nicorandil increased HO-1 expression in HUVECs. In addition, cobalt protoporphyrin (10 µmol/l), an inducer of HO-1 expression, mimicked the effects of nicorandil and inhibited IL-8 expression under cyclic strain, whereas zinc protoporphyrin IX (10 µmol/l), an inhibitor of HO-1 expression, antagonized the effect of nicorandil. HO-1 silencing significantly abrogated the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression, suggesting that HO-1 plays a role in the mechanism of action of nicorandil. KEY MESSAGES: This study is the first to report that nicorandil inhibits cyclic strain-induced IL-8 expression through the induction of HO-1 expression in HUVECs. This finding provides valuable new insight into the molecular pathways contributing to the vasoprotective effects of nicorandil.


Subject(s)
Cardiovascular Agents/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-8/metabolism , Nicorandil/pharmacology , Stress, Mechanical , Biomechanical Phenomena , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-8/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics
5.
Bioorg Med Chem Lett ; 24(6): 1562-4, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24529868

ABSTRACT

Chemical investigations on the acetone extract of the Formosan soft coral Sinularia gyrosa have obtained a novel C-4 norcembranoid possessing an unprecedented tricyclo[9.3.0.0(3,8)]tetradecane skeleton, namely sinugyrosanolide A. The NMR spectroscopic data of the novel norcembranoid were completely assigned by using a combination of 2D NMR experiments including (1)H-(1)H COSY, HSQC, HMBC, and NOESY. The cytotoxicities, anti-HCMV (human cytomegalovirus) endonuclease activities and antibacterial activities were evaluated in vitro. It showed moderate cytotoxicity against P-388 (mouse lymphocytic leukemia) cancer cell line with an EC50 of 11.8µM.


Subject(s)
Anthozoa/chemistry , Anti-Bacterial Agents/chemistry , Diterpenes/chemistry , Alkanes/chemistry , Animals , Anthozoa/metabolism , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Carbon/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cytomegalovirus/enzymology , Diterpenes/isolation & purification , Diterpenes/pharmacology , Endonucleases/antagonists & inhibitors , Endonucleases/metabolism , Humans , Magnetic Resonance Spectroscopy , Mice , Molecular Conformation , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
6.
Bioorg Med Chem Lett ; 24(2): 473-5, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24370010

ABSTRACT

Two new kelsoane-type sesquiterpenes, namely kelsoenethiol (1) and dikelsoenyl ether (2), were obtained from the Formosan soft coral Nephthea erecta. Their structures were elucidated through extensive spectroscopic analyses, ESI orbitrap mass and quantum chemical calculations (QCC). The cytotoxicity against A-459 (human lung carcinoma), P-388 (mouse lymphocytic leukemia), and HT-29 (human colon adenocarcinoma) cancer cell lines of 1 and 2 was evaluated in vitro. Compound 1 showed cytotoxicity against P-388 and HT-29 cells with ED50s of 1.3 and 1.8 µg/mL, respectively.


Subject(s)
Anthozoa , Sesquiterpenes/chemistry , Animals , Drug Screening Assays, Antitumor/methods , HT29 Cells , Humans , Leukemia P388 , Mice , Sesquiterpenes/isolation & purification
7.
Article in English | MEDLINE | ID: mdl-19962446

ABSTRACT

Angiogenesis is a highly regulated physiological process in animals. Angiopoietin-1 (Angpt1) induces the signaling pathways related to vessel maturation in late phase of angiogenesis, which recruits pericyte supplements to make compact interaction with vessel tubes. There are only few data showing Angpt1 functions in fish. By using degenerate primers, partial sequence (812 bp) of Angpt1 was cloned from Anguilla japonica, and deduced amino acids showed 80% similarity to those of zebrafish. Physiological functions of cloned eel Angpt1 were studied by in vitro and in vivo manipulations with gas glands (rete mirabile) taken as the tested target tissues. RT-PCR and immunofluorescent staining techniques were performed to examine the expression patterns of Angpt1 as well as VEGF-Flk. Experimental data showed that, in vitro, bFGF, PPAR beta agonist, and estradiol affected Angpt1 expression; while cobalt ions, a VEGF expression-inducer, did not affect Angpt1 expression. In vivo, expression levels of Angpt1 increased with body growth. Furthermore, Angpt1 expressions increased significantly in the late stage of gas glands in the stimulated eel. Successive expression patterns on VEGF-Flk, and Angpt1 on different development stages of gas glands were observed. Our results suggest that the original function of angiopoietin-1 on angiogenesis is conserved during evolution.


Subject(s)
Angiopoietin-1/genetics , Anguilla/embryology , Anguilla/genetics , Animal Structures/embryology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Vascular Endothelial Growth Factor Receptor-2/genetics , Angiopoietin-1/metabolism , Animal Structures/cytology , Animal Structures/drug effects , Animal Structures/metabolism , Animals , Body Size , Catfishes , Cells, Cultured , Cobalt/pharmacology , DNA, Complementary/genetics , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gonadal Steroid Hormones/pharmacology , Insulin/pharmacology , Male , Peroxisome Proliferator-Activated Receptors/agonists , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Sequence Alignment , Tissue Extracts , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Clin Exp Pharmacol Physiol ; 36(5-6): 559-66, 2009 May.
Article in English | MEDLINE | ID: mdl-19673940

ABSTRACT

1. Mechanical stretch leads to cardiac hypertrophy and may ultimately cause heart failure. However, the effect of mechanical stretch on gene induction in cardiomyocytes remains to be determined. 2. In the present study, we compared transcript profiles of mechanically stretched neonatal rat cardiomyocytes with those of unstretched cells using cDNA microarrays. The microarrays contained probes for 480 known genes, including those involved in signal transduction, cell cycle regulation, the cytoskeleton and cell motility. Eighteen genes, including the eNOS gene, were identified as having significantly differential expression in response to mechanical stretch in cardiomyocytes. 3. Northern and western blot analysis further quantified the expression of the eNOS gene. Mechanical stretch increased constitutive NOS activity and nitric oxide (NO) production. The NO donor s-nitroso-N-acetylpenicillamine (SNAP) inhibited mechanical stretch-stimulated protein synthesis, as measured by [3H]-leucine uptake. In addition, cardiomyocytes were infected with adenoviral vectors encoding cDNA for eNOS (Ad-eNOS) and a phosphoglycerate kinase (PGK) empty vector (Ad-PGK). In contrast with Ad-PGK-infected cells, in cardiomyocytes infected with Ad-eNOS, there was increased calcium-dependent NOS activity and nitrite production. Cardiomyocytes infected with Ad-eNOS exhibited diminished mechanical stretch-stimulated protein synthesis. In contrast, in eNOS-knockdown cells, the increased eNOS protein levels and NOS activity induced by mechanical stretch were abolished, but protein synthesis was enhanced. 4. The results of the present study indicate that eNOS gene expression is induced by mechanical stretch, leading to increased constitutive NOS activity and NO production, which may be a negative regulator in cardiomyocyte hypertrophy.


Subject(s)
Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type III/genetics , Stress, Mechanical , Animals , Animals, Newborn , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Hypertrophy/genetics , Muscle Tonus/drug effects , Muscle Tonus/genetics , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , RNA, Small Interfering/pharmacology , Rats , Transfection
9.
Free Radic Biol Med ; 40(8): 1313-22, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16631521

ABSTRACT

Alpha-lipoic acid (ALA) is a natural antioxidant that scavenges reactive oxygen species (ROS) and regenerates or recycles endogenous antioxidants. ALA has recently been reported to protect against oxidative injury in various disease processes. The aim of this study was to investigate whether the antioxidant effect of ALA is mediated by the induction of heme oxygenase (HO)-1 in rat aortic smooth muscle cells (A10 cells). ALA significantly induced HO-1 expression accompanied by an increase in HO activity in A10 cells. Pretreatment with ALA increased the resistance of A10 cells to hydrogen-peroxide-induced oxidant stress. This protection of ALA was abrogated in the presence of the HO inhibitor zinc protoporphyrin IX. ALA significantly increased ROS, and this effect was blocked by N-acetyl-cysteine, which also inhibited ALA-induced activation of p44/42 mitogen-activated protein kinase (MAPK) and AP-1, HO-1 expression, and HO activity. These results suggest that ALA induces HO-1 expression through the production of ROS and subsequent activation of the p44/42 MAPK pathway and AP-1 in vascular smooth muscle cells. This study demonstrated that ALA increases the expression of HO-1, a critical cytoprotective molecule, and identified a novel pleiotropic effect of ALA on cardiovascular protection.


Subject(s)
Heme Oxygenase-1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Thioctic Acid/pharmacology , Animals , Cells, Cultured , DNA/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Rats , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
10.
Mol Pharmacol ; 69(4): 1347-55, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16391241

ABSTRACT

Endothelin-1 (ET-1) is implicated in fibroblast proliferation, which results in cardiac fibrosis. Both reactive oxygen species (ROS) generation and epidermal growth factor receptor (EGFR) transactivation play critical roles in ET-1 signal transduction. In this study, we used rat cardiac fibroblasts treated with ET-1 to investigate the connection between ROS generation and EGFR transactivation. ET-1 treatment was found to stimulate the phosphorylation of EGFR and ROS generation, which were abolished by ETA receptor antagonist N-(N-(N-((hexahydro-1H-azepin-1-yl)carbonyl)-L-leucyl)-D-tryptophyl)-D-tryptophan (BQ485). NADPH oxidase inhibitor diphenyleneiodonium chloride (DPI), ROS scavenger N-acetyl cysteine (NAC), and p47phox small interfering RNA knockdown all inhibited the EGFR transactivation induced by ET-1. In contrast, EGFR inhibitor 4-(3'-chloroanilino)-6,7-dimethoxyquinazoline (AG-1478) cannot inhibit intracellular ROS generation induced by ET-1. Src homology 2-containing tyrosine phosphatase (SHP-2) was shown to be associated with EGFR during ET-1 treatment by EGFR coimmunoprecipitation. ROS have been reported to transiently oxidize the catalytic cysteine of phosphotyrosine phosphatases to inhibit their activity. We examined the effect of ROS on SHP-2 in cardiac fibroblasts using a modified malachite green phosphatase assay. SHP-2 was transiently oxidized during ET-1 treatment, and this transient oxidization could be repressed by DPI or NAC treatment. In SHP-2 knockdown cells, ET-1-induced phosphorylation of EGFR was dramatically elevated and is not influenced by NAC and DPI. However, this elevation was suppressed by GM6001 [a matrix metalloproteinase (MMP) inhibitor] and heparin binding (HB)-epidermal growth factor (EGF) neutralizing antibody. Our data suggest that ET-1-ETA-mediated ROS generation can transiently inhibit SHP-2 activity to facilitate the MMP-dependent and HB-EGF-stimulated EGFR transactivation and mitogenic signal transduction in rat cardiac fibroblasts.


Subject(s)
Endothelin-1/metabolism , ErbB Receptors/genetics , Protein Tyrosine Phosphatases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transcriptional Activation , Animals , Immunoprecipitation , Oxidation-Reduction , Rats , src Homology Domains
11.
Mol Pharmacol ; 68(4): 1183-92, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16049167

ABSTRACT

Cardiac hypertrophy is a compensatory mechanism in response to a variety of cardiovascular diseases. Recently, reactive oxygen species and nitric oxide (NO) have been demonstrated to be involved in the pathogenesis of atherosclerosis; however, the role of these free radicals in the development of cardiac hypertrophy remains unclear. In this study, we investigate NO modulation of cellular signaling in endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy in culture. ET-1 treatment of cardiomyocytes increased constitutive NO synthase activity and induced NO production via the stimulation of ET-receptor subtype ET(B). Using Northern blot analysis and chloramphenicol acetyltransferase assay, we found that NO suppressed the ET-1-induced increase in c-fos mRNA level and promoter activity. In contrast, ET-1 stimulation of c-fos expression was augmented by depletion of endogenous NO generation with the addition of NO scavenger PTIO into cardiomyocytes. Cells cotransfected with the dominant negative and positive mutants of signaling molecules revealed that the Ras/Raf/extracellular-signal regulated kinase (ERK) signaling pathway is involved in ET-induced c-fos gene expression. Furthermore, NO directly inhibited ET-1-induced ERK phosphorylation and activation in a cGMP-dependent manner, indicating that NO modulates ET-1-induced c-fos expression via its inhibitory effect on ERK signaling pathway. The ET-1-stimulated activator protein-1 (AP-1) DNA binding activity and AP-1-mediated reporter activity were attenuated by NO. In addition, NO also significantly inhibited ET-1-stimulated promoter activity of hypertrophic marker gene beta-myosin heavy chain and the enhanced protein synthesis. Taken together, our findings provide the molecular basis of NO as a negative regulator in ET-1-induced cardiac hypertrophy.


Subject(s)
Cyclic GMP/physiology , Endothelin-1/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Ventricles/metabolism , Nitric Oxide/physiology , Animals , Cells, Cultured , Electrophoretic Mobility Shift Assay , Endothelin-1/physiology , Gene Expression Regulation/physiology , Genes, fos , Heart Ventricles/enzymology , Heart Ventricles/pathology , Nitric Oxide Synthase/metabolism , Phosphorylation , Rats , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/physiology
12.
J Biomed Sci ; 12(1): 123-33, 2005.
Article in English | MEDLINE | ID: mdl-15864745

ABSTRACT

Endothelin-1 (ET-1) has been found to increase cardiac beta-myosin heavy chain (beta-MyHC) gene expression and induce hypertrophy in cardiomyocytes. ET-1 has been demonstrated to increase intracellular reactive oxygen species (ROS) in cardiomyocytes. The exact molecular mechanism by which ROS regulate ET-1-induced beta-MyHC gene expression and hypertrophy in cardiomyocytes, however, has not yet been fully described. We aim to elucidate the molecular regulatory mechanism of ROS on ET-1-induced beta-MyHC gene expression and hypertrophic signaling in neonatal rat cardiomyocytes. Following stimulation with ET-1, cultured neonatal rat cardiomyocytes were examined for 3H-leucine incorporation and beta-MyHC promoter activities. The effects of antioxidant pretreatment on ET-1-induced cardiac hypertrophy and mitogen-activated protein kinase (MAPKs) phosphorylation were studied to elucidate the redox-sensitive pathway in cardiomyocyte hypertrophy and beta-MyHC gene expression. ET-1 increased 3H-leucine incorporation and beta-MyHC promoter activities, which were blocked by the specific ET(A) receptor antagonist BQ-485. Antioxidants significantly reduced ET-1-induced 3H-leucine incorporation, beta-MyHC gene promoter activities and MAPK (extracellular signal-regulated kinase, p38, and c-Jun NH2 -terminal kinase) phosphorylation. Both PD98059 and SB203580 inhibited ET-1-increased 3H-leucine incorporation and beta-MyHC promoter activities. Co-transfection of the dominant negative mutant of Ras, Raf, and MEK1 decreased the ET-1-induced beta-MyHC promoter activities, suggesting that the Ras-Raf-MAPK pathway is required for ET-1 action. Truncation analysis of the beta-MyHC gene promoter showed that the activator protein-2 (AP-2)/specificity protein-1 (SP-1) binding site(s) were(was) important cis-element(s) in ET-1-induced beta-MyHC gene expression. Moreover, ET-1-induced AP-2 and SP-1 binding activities were also inhibited by antioxidant. These data demonstrate the involvement of ROS in ET-1-induced hypertrophic responses and beta-MyHC expression. ROS mediate ET-1-induced activation of MAPK pathways, which culminates in hypertrophic responses and beta-MyHC expression.


Subject(s)
Endothelin-1/metabolism , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Myosin Heavy Chains/genetics , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Cardiac Myosins , Cells, Cultured , Enzyme Inhibitors/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Hypertrophy , Myosin Heavy Chains/metabolism , Oxidation-Reduction , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/metabolism
13.
J Biomed Sci ; 11(1): 11-8, 2004.
Article in English | MEDLINE | ID: mdl-14730205

ABSTRACT

The myocardial protective effects of trilinolein, isolated from the traditional Chinese herb Sanchi (Panax notoginseng), are thought to be related to its antioxidant activity. However, the intracellular mechanism underlying the protective effect of trilinolein in the heart remains unclear. In the present study, we investigated the effect of trilinolein on norepinephrine (NE)-induced protein synthesis in cardiomyocytes. Cultured neonatal rat cardiomyocytes were stimulated with NE, then protein content, [(3)H]-leucine incorporation, and beta-myosin heavy chain (beta-MyHC) promoter activity were examined. The effect of trilinolein on NE-induced intracellular reactive oxygen species (ROS) generation was measured with a redox- sensitive fluorescent dye (2',7'-dichlorofluorescin diacetate) and extracellular signal-regulated kinase (ERK) phosphorylation by Western blotting. Trilinolein inhibited NE-increased protein synthesis, beta-MyHC promoter activity, and intracellular ROS. Both trilinolein and the antioxidant, N-acetyl-cysteine, decreased NE- and H(2)O(2)-induced protein synthesis, beta-MyHC promoter activity, and ERK phosphorylation. These data indicate that trilinolein inhibits NE-induced protein synthesis via attenuation of ROS generation in cardiomyocytes.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , Norepinephrine/pharmacology , Promoter Regions, Genetic/drug effects , Reactive Oxygen Species/metabolism , Triglycerides/pharmacology , Ventricular Myosins/genetics , Acetylcysteine/metabolism , Animals , Animals, Newborn , Cells, Cultured , Fluorescent Dyes/metabolism , Free Radical Scavengers/metabolism , Gene Expression Regulation/drug effects , Hydrogen Peroxide/metabolism , Molecular Structure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Oxidants/metabolism , Oxidation-Reduction , Phosphorylation , Rats , Rats, Sprague-Dawley , Triglycerides/chemistry , Ventricular Myosins/metabolism
14.
Eur J Pharmacol ; 484(1): 1-8, 2004 Jan 19.
Article in English | MEDLINE | ID: mdl-14729376

ABSTRACT

The myocardial protective effects of trilinolein, isolated from the Chinese herb Sanchi (Panax notoginseng), may be related to its antioxidant effects. In the present study, we investigated the effects of trilinolein on angiotensin II-induced cardiomyocyte hypertrophy. Cultured neonatal rat cardiomyocytes were stimulated with angiotensin II, [3H]leucine incorporation and the beta-myosin heavy chain promoter activity were examined. We also examined the effects of trilinolein on angiotensin II-induced intracellular reactive oxygen species generation. Trilinolein significantly inhibited angiotensin II-increased protein synthesis, beta-myosin heavy chain promoter activity, and intracellular reactive oxygen species generation. Antioxidant N-acetylcysteine also decreased angiotensin II-increased protein synthesis and beta-myosin heavy chain promoter activity. Furthermore, trilinolein and N-acetylcysteine decreased angiotensin II- or hydrogen peroxide (H2O2)-activated mitogen-activated protein kinases (MAPKs) phosphorylation, and activator protein-1 (AP-1)- [or nuclear factor-kappaB (NF-kappaB)]-reporter activities. These data indicate that trilinolein inhibits angiotensin II-induced cardiomyocyte hypertrophy and beta-myosin heavy chain promoter activity via attenuation of reactive oxygen species generation.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Cardiomegaly/prevention & control , Myocytes, Cardiac/drug effects , Triglycerides/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Triglycerides/therapeutic use
15.
Naunyn Schmiedebergs Arch Pharmacol ; 369(2): 239-44, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14663554

ABSTRACT

Resveratrol is proposed to account in part for the protective effect of red wine on the cardiovascular system. Angiotensin II (Ang II) is a potent hypertrophic stimulus in cardiomyocytes. In this study, we determined the effect of resveratrol on Ang II-induced cardiomyocyte hypertrophy. Cultured neonatal rat cardiomyocytes were stimulated with Ang II, and [3H]leucine incorporation and beta-myosin heavy chain (beta-MyHC) promoter activity were examined. Intracellular reactive oxygen species (ROS) were measured by a redox-sensitive fluorescent dye, 2' 7'-dichlorofluorescin diacetate, and the extracellular signal-regulated kinase (ERK) phosphorylation was examined by Western blotting. Resveratrol inhibited Ang II-increased intracellular ROS levels. Furthermore, resveratrol, as well as the antioxidant N-acetyl-cysteine, decreased Ang II- or H2O2-increased protein synthesis, beta-MyHC promoter activity, and ERK phosphorylation. In summary, we demonstrate for the first time that resveratrol inhibits Ang II-induced cardiomyocyte hypertrophy via attenuation of ROS generation.


Subject(s)
Angiotensin II/metabolism , Myocytes, Cardiac/drug effects , Stilbenes/pharmacology , Acetylcysteine/pharmacology , Angiotensin II/pharmacology , Animals , Animals, Newborn , Antioxidants/pharmacology , Cells, Cultured , Hypertrophy/prevention & control , Leucine/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Phosphorylation , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resveratrol , Transfection , Ventricular Myosins/genetics
16.
J Am Coll Cardiol ; 42(10): 1845-54, 2003 Nov 19.
Article in English | MEDLINE | ID: mdl-14642698

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effects of angiotensin II (Ang II) on fibroblast proliferation and endothelin-1 (ET-1) gene induction, focusing especially on reactive oxygen species (ROS)-mediated signaling in cardiac fibroblasts. BACKGROUND: Angiotensin II increases ET-1 expression, which plays an important role in Ang II-induced fibroblast proliferation. Angiotensin II also stimulates ROS generation in cardiac fibroblasts. However, whether ROS are involved in Ang II-induced proliferation and ET-1 expression remains unknown. METHODS: Cultured neonatal rat cardiac fibroblasts were stimulated with Ang II, and then [(3)H]thymidine incorporation and the ET-1 gene expression were examined. We also examined the effects of antioxidants on Ang II-induced proliferation and mitogen-activated protein kinase (MAPK) phosphorylation to elucidate the redox-sensitive pathway in fibroblast proliferation and ET-1 gene expression. RESULTS: Both AT(1) receptor antagonist (losartan) and ET(A) receptor antagonist (BQ485) inhibited Ang II-increased DNA synthesis. Endothelin-1 gene was induced with Ang II as revealed by Northern blotting and promoter activity assay. Angiotensin II increased intracellular ROS levels, which were inhibited with losartan and antioxidants. Antioxidants further suppressed Ang II-induced ET-1 gene expression, DNA synthesis, and MAPK phosphorylation. PD98059, but not SB203580, fully inhibited Ang II-induced ET-1 expression. Truncation and mutational analysis of the ET-1 gene promoter showed that AP-1 binding site was an important cis-element in Ang II-induced ET-1 gene expression. CONCLUSIONS: Our data suggest that ROS are involved in Ang II-induced proliferation and ET-1 gene expression. Our findings imply that the combination of AT(I) and ET(A) receptor antagonists plus antioxidants may be beneficial in preventing the formation of excessive cardiac fibrosis.


Subject(s)
Angiotensin II/metabolism , Endothelin-1/genetics , Fibroblasts/physiology , Gene Expression/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Angiotensin Receptor Antagonists , Animals , Antihypertensive Agents/pharmacology , Azepines/pharmacology , Cell Culture Techniques , Cell Division/physiology , Endothelin Receptor Antagonists , Endothelin-1/metabolism , Heart/physiology , Losartan/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Oligopeptides/pharmacology , Oxidation-Reduction , Rats , Receptors, Angiotensin/metabolism , Receptors, Endothelin/metabolism , Up-Regulation
17.
Mol Pharmacol ; 63(5): 1002-11, 2003 May.
Article in English | MEDLINE | ID: mdl-12695528

ABSTRACT

Endothelin-1 (ET-1) has been implicated in fibroblast proliferation. However, the mechanism involving ET-1 is not clear. The present study was performed to examine the role of endogenous ET-1 in ET-1-stimulated fibroblast proliferation and to investigate the regulatory mechanism of ET-1-induced ET-1 gene expression in cardiac fibroblasts. Both ET(A) receptor antagonist [(hexahydro-1H-azepinyl)carbonyl-Leu-D-Trp-D-OH (BQ485)] and endothelin-converting enzyme inhibitor (phosphoramidon) inhibited the increased DNA synthesis caused by ET-1. ET-1 gene was induced by ET-1, as revealed with Northern blotting and ET-1 promoter activity assay. ET-1 increased intracellular reactive oxygen species (ROS), which were significantly inhibited by BQ485 and antioxidants. Antioxidants suppressed ET-1 gene expression and DNA synthesis stimulated by ET-1. ET-1 activated mitogen-activated protein kinases (MAPK), including extracellular signal-regulated kinase (ERK), p38 MAPK, and c-Jun N-terminal kinase, which were significantly inhibited by antioxidants. Only ERK inhibitor U0126 could inhibit ET-1-induced transcription of the ET-1 gene. Cotransfection of dominant-negative mutant of Ras, Raf, and MEK1 decreased the ET-1-induced increase in ET-1 transcription, suggesting that the Ras-Raf-ERK pathway is required for ET-1 action. Truncation and mutational analysis of the ET-1 gene promoter showed that the activator protein-1 (AP-1) binding site was an important cis-element in ET-1-induced ET-1 gene expression. Antioxidants attenuated the ET-1-stimulated AP-1 binding activity. Our data suggest that ROS were involved in ET-1-induced fibroblast proliferation and mediated ET-1-induced activation of ERK pathways, which culminated in ET-1 gene expression.


Subject(s)
Endothelin-1/genetics , Fibroblasts/physiology , Gene Expression/physiology , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Cell Division , DNA/biosynthesis , Humans , Myocardium/cytology , Oxidation-Reduction , Rats , Receptor, Endothelin A , Receptors, Endothelin/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...