Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Microcirculation ; 17(1): 69-78, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20141602

ABSTRACT

BACKGROUND: This study was designed to explore the effect of transient inducible nitric oxide synthase (iNOS) overexpression via cationic liposome-mediated gene transfer on cardiac function, fibrosis, and microvascular perfusion in a porcine model of chronic ischemia. METHODS AND RESULTS: Chronic myocardial ischemia was induced using a minimally invasive model in 23 landrace pigs. Upon demonstration of heart failure, 10 animals were treated with liposome-mediated iNOS-gene-transfer by local intramyocardial injection and 13 animals received a sham procedure to serve as control. The efficacy of this iNOS-gene-transfer was demonstrated for up to 7 days by reverse transcriptase-polymerase chain reaction in preliminary studies. Four weeks after iNOS transfer, magnetic resonance imaging showed no effect of iNOS overexpression on cardiac contractility at rest and during dobutamine stress (resting ejection fraction: control 27%, iNOS 26%; P = ns). Late enhancement, infarct size, and the amount of fibrosis were similar between groups. Although perfusion and perfusion reserve in response to adenosine and dobutamine were not significantly modified by iNOS-transfer, both vessel number and diameter were significantly increased in the ischemic area in the iNOS-treated group versus control (point score: control 15.3, iNOS 34.7; P < 0.05). CONCLUSIONS: Our findings demonstrate that transient iNOS overexpression does not aggravate cardiac dysfunction or postischemic fibrosis, while potentially contributing to neovascularization in the chronically ischemic heart.


Subject(s)
Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Animals , Arterioles/pathology , Arterioles/physiopathology , Female , Fibrosis , Gene Expression , Gene Transfer Techniques , Humans , Liposomes , Magnetic Resonance Imaging , Male , Myocardial Ischemia/genetics , Neovascularization, Pathologic , Nitric Oxide/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sus scrofa , Ventricular Function, Left
2.
Mol Immunol ; 45(9): 2678-89, 2008 May.
Article in English | MEDLINE | ID: mdl-18262272

ABSTRACT

Cytokines interleukin-1beta (IL-1beta) and interleukin-6 (IL-6) are involved in acute phase response (APR). C-reactive protein (CRP), the prototype acute phase protein, may represent an important component in the pathogenesis of arteriosclerosis and may also be a target for drug development. Inhibition of CRP synthesis is one potential strategy. Understanding CRP synthesis, however, is a prerequirement for the development of CRP-inhibitors. From studies in hepatoma cell lines, IL-1beta and IL-6 were considered as equal inductors of APR and CRP. We investigated IL-1beta- and IL-6-effects on primary human hepatocytes (PHH) and Hep3B-cells. Kupffer cell contamination in PHH preparations was <3%. In PHH, several APP like CRP, haptoglobin (HP), lipopolysaccharide-binding protein (LBP) or hepcidin (HAMP) were regulated similarly by IL-1beta and IL-6, though signal transduction pathways of these cytokines are different. In Hep3B-cells, APP were regulated exclusively by IL-6. IL-1beta induced IL-6-synthesis in PHH but not in Hep3B-cells. C/EBPbeta-overexpression in Hep3B-cells reconstituted IL-1beta-mediated IL-6/CRP inducibility. In PHH and in C/EBPbeta-overexpressing Hep3B-cells, neutralizing anti-IL-6-antibodies blocked IL-1beta-mediated APR. Inhibition of protein synthesis and NFkappaB-signalling blocked IL-1beta- but not IL-6-mediated CRP-expression in PHH, whereas Janus-Kinase-1-inhibition blocked IL-1beta- and IL-6-mediated APR. IL-1beta induces APR in PHH via an NFkappaB- and C/EBPbeta-dependent autocrine IL-6-loop. These findings partly reconcile the understanding of APR and may help to design a transcriptional suppressor of CRP for the treatment of cardiovascular disease.


Subject(s)
Acute-Phase Reaction , C-Reactive Protein/biosynthesis , Hepatocytes/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , Acute-Phase Proteins/biosynthesis , Acute-Phase Proteins/genetics , Acute-Phase Reaction/immunology , C-Reactive Protein/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Profiling , Hepatocytes/cytology , Hepatocytes/immunology , Humans , Interleukin-1beta/immunology , Interleukin-6/immunology , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction
3.
J Mol Cell Cardiol ; 38(2): 375-85, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15698844

ABSTRACT

Akt is a serine/threonine kinase that mediates a variety of cellular responses to external stimuli. Among the three members of mammalian Akt (Akt1, Akt2 and Akt3), Akt3 is unique in that it has an alternatively spliced variant that lacks the carboxy-terminal regulatory phosphorylation site. However, little is known regarding in vivo functions of Akt3 and its spliced variant. In this study we investigated the potential functions of the Akt3 spliced variant by overexpressing its activated form in the heart. Cardiac-specific Akt3 transgenic (TG) mice exhibited marked cardiac hypertrophy. Contractile function of TG hearts was preserved at 4 and 12 weeks, but was impaired at 20 weeks of age. When treated with cardiotoxic drug doxorubicin (Dox), TG mice at 4 weeks of age exhibited improved survival and preserved contractile function. However, these cardioprotective effects were not evident when Dox was injected at 12 weeks of age, and TG mice exhibited even higher mortality rate than wild-type animals when Dox was injected at 20 weeks of age. Endogenous Akt1 and Akt2 protein and phosphorylation levels were downregulated in Akt3 TG hearts, suggesting the existence of negative feedback regulation of Akt signaling at the level of Akt protein amount. Taken together, the Akt3 spliced variant is functional in vivo, promotes cardiac growth and mediates cardioprotective effects. However, continuous overexpression of Akt3 results in contractile dysfunction and increased susceptibility to cardiac injury. Thus, sustained activation of Akt signaling results in progression from adaptive to maladaptive hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/pathology , Myocardium/enzymology , Myocardium/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/genetics , Disease Progression , Doxorubicin/pharmacology , Gene Expression , Humans , Mice , Mice, Transgenic , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction , Survival Rate , Up-Regulation
4.
Arterioscler Thromb Vasc Biol ; 25(1): 186-92, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15539624

ABSTRACT

OBJECTIVE: C-reactive protein (CRP) is the prototype acute phase protein and a cardiovascular risk factor. Interleukin-1beta (IL-1beta) and IL-6 stimulate CRP synthesis in hepatocytes. We searched for additional pathways regulating CRP expression. METHODS AND RESULTS: Primary human hepatocytes (PHHs) were treated with IL-1beta, IL-6, and protein kinase C (PKC) activator phorbol 12,13-dibutyrate (PDBu). CRP was analyzed by quantitative RT-PCR and ELISA. PDBu significantly induced CRP transcription by 21.0+/-9.24-fold and protein release by 2.9+/-0.5-fold. Transcriptional regulation was studied in detail in hepatoma G2 (HepG2) cells stably transfected with the 1-kb CRP promoter (HepG2-ABEK14 cells). In these cells, PDBu significantly induced CRP transcription by 5.39+/-0.66-fold. Competitive inhibition with bisindolylmaleimide derivative LY333531 abolished PDBu-mediated promoter activation. Competitive inhibition with IkappaB kinase inhibitor I229 also inhibited PDBu effects. Importantly, IL-8 significantly induced CRP release in PHHs by 58.675+/-19.1-fold, which was blockable by LY333531. CONCLUSIONS: This study describes a novel PKC-dependent transcriptional regulation of CRP gene expression, which, in analogy to the classical IL-1beta and IL-6 pathways, is operational in hepatocytes only. It also identifies IL-8 as a potential physiological PKC activator. HepG2-ABEK14 cells may be useful for high throughput screening to identify inhibitors of CRP synthesis for the prevention of cardiovascular disease.


Subject(s)
C-Reactive Protein/genetics , Gene Expression Regulation/physiology , Hepatocytes/chemistry , Hepatocytes/metabolism , Protein Kinase C/metabolism , Transcription, Genetic/physiology , Aorta/cytology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Interleukin-8/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase C beta , Transcription, Genetic/drug effects , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...