Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Cardiol ; 168(2): 1447-52, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23360823

ABSTRACT

BACKGROUND: The transcription factor TWIST1 has been described to regulate the microRNA (miR)-199/214 cluster. Genetic disruption of TWIST1 resulted in a cachectic phenotype and early death of the knock-out mice. This might be connected to the activity of the ubiquitin-proteasome-system (UPS), as miR-199a has been suggested to regulate the ubiquitin E2 ligases Ube2i and Ube2g1. METHODS: Cardiac tissue from explanted hearts of 42 patients with dilated cardiomyopathy and 20 healthy donor hearts were analysed for protein expression of TWIST1 and its inhibitors Id-1, MuRF-1 and MAFbx, the expression of miR-199a, -199b and -214, as well as the activity of the UPS by using specific fluorogenic substrates. RESULTS: TWIST1 was repressed in patients with dilated cardiomyopathy by 43% (p=0.003), while Id1 expression was unchanged. This was paralleled by a reduced expression of miR-199a by 38 ± 9% (p=0.053), miR-199b by 36 ± 13% (p=0.019) and miR-214 by 41 ± 11% (p=0.0158) compared to donor hearts. An increased peptidylglutamyl-peptide-hydrolysing activity (p<0.0001) was observed in the UPS, while the chymotrypsin-like and trypsin-like activities were unchanged. The protein levels of the rate limiting ubiquitin E3-ligases MuRF-1 and MAFbx were up-regulated (p=0.005 and p=0.0156, respectively). Mechanistically silencing of TWIST1 using siRNA in primary rat cardiomyocytes led to a down-regulation of the miR-199/214 cluster and to a subsequent up-regulation of Ube2i. CONCLUSION: The TWIST1/miR-199/214 axis is down-regulated in dilated cardiomyopathy, which is likely to play a role in the increased activity of the UPS. This may contribute to the loss of cardiac mass during dilatation of the heart.


Subject(s)
Cardiomyopathy, Dilated/metabolism , MicroRNAs/biosynthesis , Nuclear Proteins/physiology , Proteasome Endopeptidase Complex/metabolism , Twist-Related Protein 1/physiology , Ubiquitin/metabolism , Adult , Animals , Animals, Newborn , Cardiomyopathy, Dilated/pathology , Female , Humans , Male , Middle Aged , Rats
2.
J Mol Med (Berl) ; 86(1): 61-74, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17646949

ABSTRACT

The prevalence and clinical manifestation of several cardiovascular diseases vary considerably with sex and age. Thus, a better understanding of the molecular basis of these differences may represent a starting point for an improved gender-specific medicine. Despite the fact that sex-specific differences have been observed in the cardiovascular system of humans and animal models, systematic analyses of sexual dimorphisms at the transcriptional level in the healthy heart are missing. Therefore we performed gene expression profiling on mouse and human cardiac samples of both sexes and young as well as aged individuals and verified our results for a subset of genes using real-time polymerase chain reaction in independent left ventricular samples. To tackle the question whether sex differences are evolutionarily conserved, we also compared sexually dimorphic genes between both species. We found that genes located on sex chromosomes were the most abundant ones among the sexually dimorphic genes. Male-specific expression of Y-linked genes was observed in mouse hearts as well as in the human myocardium (e.g. Ddx3y, Eif2s3y and Jarid1d). Higher expression levels of X-linked genes were detected in female mice for Xist, Timp1 and Car5b and XIST, EIF2S3X and GPM6B in women. Furthermore, genes on autosomal chromosomes encoding cytochromes of the monoxygenase family (e.g. Cyp2b10), carbonic anhydrases (e.g. Car2 and Car3) and natriuretic peptides (e.g. Nppb) were identified with sex- and/or age-specific expression levels. This study underlines the relevance of sex and age as modifiers of cardiac gene expression.


Subject(s)
Gene Expression Regulation , Myocardium/metabolism , Sex Characteristics , Sex Chromosomes/genetics , Age Factors , Animals , Chromosomes, Human, X , Chromosomes, Human, Y , Female , Gene Expression Profiling , Genes, X-Linked , Genes, Y-Linked , Humans , Male , Mice , Myocardium/chemistry , Sex Factors
3.
Eur J Heart Fail ; 9(6-7): 568-73, 2007.
Article in English | MEDLINE | ID: mdl-17468044

ABSTRACT

BACKGROUND: The role of melusin, a necessary component in pressure-induced left-ventricular hypertrophy (LVH) in mice, has not yet been determined in human cardiac hypertrophy. We analyzed for the first time the expression and regional distribution of melusin in human LVH due to aortic stenosis (AS) and determined AKT phosphorylation as a potential downstream effector of melusin signalling. METHODS: Regional distribution of melusin was evaluated in four normal hearts. Melusin staining, gene expression and protein content were assessed in biopsies from normal and diseased hearts and melusin gene expression was correlated with LV functional changes. The pAKT/AKT ratio was determined in parallel and correlated with melusin protein content. RESULTS: In normal hearts, melusin was found in the myocytes with a uniform regional distribution. Melusin staining, mRNA and protein were significantly decreased in human AS hearts. The reduction in melusin mRNA was significantly correlated with LVEF, LVEDD and LVESD. pAKT/AKT ratio was significantly decreased in human AS and was correlated with melusin content. CONCLUSION: Reduction in melusin expression parallels the functional cardiac impairment in human AS. The simultaneous decrease of melusin and AKT phosphorylation suggests a connection between the loss of melusin and the decrease in systolic function.


Subject(s)
Aortic Valve Stenosis/genetics , Cytoskeletal Proteins/genetics , Muscle Proteins/genetics , RNA, Messenger/genetics , Aged , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/pathology , Biopsy , Female , Gene Expression/physiology , Humans , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Immunoenzyme Techniques , Male , Middle Aged , Myocardium/pathology , Proto-Oncogene Proteins c-akt/genetics , Reference Values , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
4.
FASEB J ; 20(7): 926-34, 2006 May.
Article in English | MEDLINE | ID: mdl-16675850

ABSTRACT

Clinical and animal studies suggest that estrogen receptors are involved in the development of myocardial hypertrophy and heart failure. In this study, we investigated whether human myocardial estrogen receptor alpha (ERalpha) expression, localization, and association with structural proteins was altered in end stage-failing hearts. We found a 1.8-fold increase in ERalpha mRNA and protein in end-stage human dilated cardiomyopathy (DCM, n=41), as compared with controls (n=25). ERalpha was visualized by confocal immunofluorescence microscopy and localized to the cytoplasm, sarcolemma, intercalated discs and nuclei of cardiomyocytes. Immunofluorescence studies demonstrated colocalization of ERalpha with beta-catenin at the intercalated disc in control hearts and immunoprecipitation studies confirmed complex formation of both proteins. Interestingly, the ERalpha/beta-catenin colocalization was lost at the intercalated disc in DCM hearts. Thus, the ERalpha/beta-catenin colocalization in the intercalated disc may be of functional relevance and a loss of this association may play a role in the progression of heart failure. The increase of total ERalpha expression may represent a compensatory process to contribute to the stability of cardiac intercalated discs.


Subject(s)
Estrogen Receptor alpha/metabolism , Heart Failure/metabolism , Up-Regulation , Adult , Cardiomyopathy, Dilated/metabolism , Female , Heart Ventricles/pathology , Humans , Male , Middle Aged , beta Catenin/metabolism
5.
J Mol Cell Cardiol ; 40(2): 234-46, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16442122

ABSTRACT

Mutations in the gene for fibrillin-1 cause Marfan syndrome (MFS), a common hereditary disorder of connective tissue. Recent findings suggest that proteolysis, increased matrix metalloproteinase activity, and fragmentation of fibrillin-rich microfibrils in tissues of persons with MFS contribute to the complex pathogenesis of this disorder. In this study we show that a fibrillin-1 fragment containing a EGFEPG sequence that conforms to a putative GxxPG elastin-binding protein (EBP) consensus sequence upregulates the expression and production of matrix metalloproteinase (MMP)-1 by up to ninefold in a cell culture system. A mutation of the GxxPG consensus sequence site abrogated the effects. This is the first demonstration of such an effect for ligands other than elastin fragments. Molecular dynamics analysis of oligopeptides with the wildtype and mutant sequence support our biochemical results by predicting significant alterations of structural characteristics such as the potential for forming a type VIII beta-turn that are thought to be important for binding to the EBP. These results suggest that fibrillin-1 fragments may regulate MMP-1 expression, and that the dysregulation of MMPs related to fragmentation of fibrillin might contribute to the development of MFS. Our Gene Ontology (GO) analysis of the human proteome shows that proteins with multiple GxxPG motifs are highly enriched for GO terms related to the extracellular matrix. Matrix proteins with multiple GxxPG sites include fibrillin-1, -2, and -3, elastin, fibronectin, laminin, and several tenascins and collagens. Some of these proteins have been associated with disorders involving alterations in MMP regulation, and the results of the present study suggest a potential mechanism for these observations.


Subject(s)
Matrix Metalloproteinase 1/biosynthesis , Microfilament Proteins/physiology , Peptide Fragments/physiology , Receptors, Cell Surface/physiology , Amino Acid Sequence , Computational Biology , Consensus Sequence , Databases, Protein , Enzyme Induction/physiology , Fibrillin-1 , Fibrillins , Humans , Matrix Metalloproteinase 1/genetics , Microfilament Proteins/genetics , Mutation , Peptide Fragments/genetics , Receptors, Cell Surface/genetics , Up-Regulation
6.
Eur J Heart Fail ; 8(3): 290-4, 2006 May.
Article in English | MEDLINE | ID: mdl-16307905

ABSTRACT

BACKGROUND: The peroxisome proliferator-activated receptor alpha (PPARalpha) is a central regulator of myocardial fatty acid (FA) metabolism implicated in the pathogenesis of heart failure. AIMS: To characterize PPARalpha regulation in human dilated cardiomyopathy (DCM), we studied the expression of cardiac PPARalpha, cardiac carnitine palmitoyl-transferase I (CPT-1), a major PPARalpha target gene, and of the cardiac glucose transporter GLUT-4 in patients with DCM. METHODS: Left ventricular biopsies were taken from patients with DCM (n=16) and control subjects (n=15), and mRNA expression was quantitated using real-time PCR (SYBR((R))Green) and protein expression was measured by Western immunoblotting. RESULTS: Left ventricular PPARalpha mRNA levels were significantly increased in the DCM group compared to the control group (136+/-25.4% vs. control, p<0.01). Consistently, DCM patients had a significantly higher cardiac CPT-1 mRNA expression (147+/-51% vs. control, p<0.05) compared to the control group. Cardiac GLUT-4 expression was similar in both groups. CONCLUSION: Elevated cardiac PPARalpha levels followed by an induction of cardiac CPT-1 expression may result in increased fatty acid metabolism for cardiac energy production in DCM, suggesting a specific cardiac metabolic program in human DCM compared to other types of cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Myocardium/metabolism , PPAR alpha/genetics , Adolescent , Adult , Carnitine O-Palmitoyltransferase/genetics , Female , Glucose Transporter Type 4/genetics , Humans , Male , Middle Aged , RNA, Messenger/analysis
7.
Biogerontology ; 6(3): 165-71, 2005.
Article in English | MEDLINE | ID: mdl-16041620

ABSTRACT

The hypoxia-inducible factor (HIF)-1 is a master transcriptional activator of oxygen-regulated genes involved in energy metabolism, angiogenesis, and erythropoiesis. HIF-1 is composed of the two subunits HIF-1alpha and HIF-1beta (also called ARNT). The destruction of HIF-1alpha in the presence of oxygen is initiated by prolyl-4-hydroxylation. In human cells three closely related prolyl hydroxylases (PHDs) have been identified. An age-dependent decrease in HIF-1alpha expression was reported previously in brain, liver and kidney, which may be associated with a reduced adaptation to hypoxia as found in aged animals and humans. We have determined the expression of HIF-1alpha and the PHDs in human atrial trabeculae under normoxic and hypoxic conditions, in samples of human left ventricles as well as in heart extracts from female mice of different age (5 up to 16 months). With increasing age we found a decreased expression of HIF-1alpha, which correlated to an increased PHD3 expression in mouse and human heart. PHD3 was the most prominent HIF modifying hydroxylase found in human heart samples. Additionally, we found a strong ischemia/hypoxia-inducibility of PHD3 compared to PHD1 and PHD2 in atrial trabeculae. These data may explain the previously reported reduction of HIF-1alpha and HIF-1 target genes such as the vascular endothelial growth factor in ageing tissue.


Subject(s)
Aging/metabolism , Dioxygenases/metabolism , Myocardium/metabolism , Adolescent , Adult , Age Factors , Aged , Animals , Dioxygenases/genetics , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor-Proline Dioxygenases , Mice , Mice, Inbred C57BL , Middle Aged , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , RNA, Messenger/analysis
8.
Hum Genet ; 116(1-2): 51-61, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15517394

ABSTRACT

The Marfan syndrome (MFS), a relatively common autosomal dominant disorder of connective tissue, is caused by mutations in the gene for fibrillin-1 (FBN1). Fibrillin-1 is the main component of the 10- to 12-nm microfibrils that together with elastin form elastic fibers found in tissues such as the aortic media. Recently, FBN1 mutations have been shown to increase the susceptibility of fibrillin-1 to proteolysis in vitro, and other findings suggest that up-regulation of matrix metalloproteinases (MMP), as well as fragmentation of microfibrils, could play a role in the pathogenesis of MFS. In the present work, we have investigated the influence of fibrillin-1 fragments on the expression of MMP-1, MMP-2, and MMP-3 in a cell culture system. Cultured human dermal fibroblasts were incubated with several different recombinant fibrillin-1 fragments. The expression level of MMP-1, MMP-2, and MMP-3, was determined by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), and the concentration of the corresponding proteins was estimated by quantitative Western blotting. Our results establish that treatment of cultured human dermal fibroblasts with recombinant fibrillin-1 fragments containing the arginine-glycine-aspartic acid (RGD) integrin-binding motif of fibrillin-1 induces up-regulation of MMP-1 and MMP-3. A similar effect was seen upon stimulation with a synthetic RGD peptide. The expression of MMP-2 was not influenced by treatment. Our results suggest the possibility that fibrillin fragments could themselves have pathogenic effects by leading to up-regulation of MMPs, which in turn may be involved in the progressive breakdown of microfibrils thought to play a role in MFS.


Subject(s)
Marfan Syndrome/genetics , Matrix Metalloproteinases/metabolism , Microfilament Proteins/genetics , Oligopeptides/genetics , Peptide Fragments/genetics , Enzyme Induction , Fibrillin-1 , Fibrillins , Humans , Marfan Syndrome/etiology , Marfan Syndrome/metabolism , Matrix Metalloproteinases/genetics , Microfilament Proteins/metabolism , Oligopeptides/metabolism , Peptide Fragments/metabolism , Sequence Analysis, Protein
9.
Regul Pept ; 124(1-3): 195-201, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15544859

ABSTRACT

Substance P and neurokinin A are regulatory peptides of the tachykinin family that influence many aspects of human airway function in health and diseases such as bronchial asthma or chronic obstructive pulmonary disease (COPD). Tachykinin-induced mucus secretion has been regarded as sensory nerve-dependent so far. We studied the distribution of tachykinin-mRNA and -peptide and its relation to NK-1 subtype-positive cells in human airway glands to assess if tachykinins may also be expressed in inflammatory cells. RT-PCR demonstrated the expression of tachykinin- and NK-1-mRNA in human airway tissues. In situ hybridisation resulted in preprotachykinin (PPT)-A mRNA-signal detection in inflammatory cells which were in close contact to myoepithelial cells of airway glands. NK-1 immunoreactivity was found in myoepithelial cells which were in direct contact to the PPT-A mRNA and tachykinin-positive cells. The present data directly demonstrate the presence of both PPT-A mRNA and tachykinin immunoreactivity in inflammatory airway cells which are in direct contact to NK-1 receptor positive glandular myoepithelium. Our findings indicate that besides neurally released tachykinins, also inflammatory cell-derived tachykinins may lead to glandular secretion via NK-1 receptor stimulation. This points to a major second source of these proinflammatory mediators in chronic inflammatory airway diseases such as COPD or asthma.


Subject(s)
Bronchitis, Chronic/metabolism , Bronchitis, Chronic/pathology , Mucus/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Tachykinins/metabolism , Bronchitis, Chronic/genetics , Humans , Immunohistochemistry , Pneumonia/genetics , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Respiratory System/metabolism , Respiratory System/pathology , Tachykinins/genetics
10.
J Mol Med (Berl) ; 82(12): 809-20, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15558223

ABSTRACT

Aortic stenosis (AS) results in myocyte and extracellular matrix remodeling in the human left ventricle (LV). The myocardial renin-angiotensin system is activated and collagens I and III and fibronectin accumulate. We determined the yet unknown regulation of enzymes that control collagen turnover, i.e., LV matrix metalloproteinases (MMP) and their tissue inhibitors (TIMPs) in human AS. We compared LV samples from AS patients undergoing elective aortic valve replacement (n=19) with nonused donor hearts with normal LV function (controls, n=12). MMP-2, MMP-9, MT1-MMP, and extracellular matrix metalloproteinase inducer (EMMPRIN), TIMP-1, TIMP-2, TIMP-3, and TIMP-4 mRNA were quantitated by real-time RCR. MMP-1, MMP-2, MMP-3, TIMP-3, TIMP-4, and EMMPRIN protein were measured by immunoblotting and MMP-9 and TIMP-1 protein by ELISA. Gelatinolytic MMP-2 and MMP-9 activity was measured by zymography. MMP-2 was increased in AS at mRNA, protein, and activity levels (131%, 193%, and 138% of controls). MMP-3 protein (308%) and EMMPRIN mRNA and protein were also upregulated (171% and 200%). In contrast, MMP-1 (37%) and MMP-9 mRNA, protein, and activity (26%, 21%, and 52%) were downregulated. MMP-9 activity was inversely correlated with LV size. TIMP-1 mRNA and protein were decreased (55% and 73%). In contrast, TIMP-2 mRNA (358%), TIMP-3 mRNA and protein (145% and 249%) were increased. TIMP-4 mRNA was not altered, but TIMP-4 protein was upregulated to 350%. Changes were similar in AS patients with normal and impaired LV ejection fraction. The dysregulation of myocardial MMPs and TIMPs in human AS starts at an early disease stage when LV function is still normal. In spite of upregulation of some MMPs the balance between MMP and TIMP is shifted towards MMP inhibition in human AS and may contribute to collagen accumulation.


Subject(s)
Aortic Valve Stenosis/enzymology , Heart Ventricles/enzymology , Matrix Metalloproteinases/metabolism , Myocardium/enzymology , Tissue Inhibitor of Metalloproteinases/metabolism , Aged , Female , Humans , Male , Matrix Metalloproteinases/genetics , Middle Aged , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinases/genetics
11.
Circulation ; 110(20): 3270-5, 2004 Nov 16.
Article in English | MEDLINE | ID: mdl-15533858

ABSTRACT

BACKGROUND: Estrogen receptor (ER)-mediated effects have been associated with the modulation of myocardial hypertrophy in animal models and in humans, but ER expression in the human heart and its relation to hypertrophy-mediated gene expression have not yet been analyzed. We therefore investigated sex- and disease-dependent alterations of myocardial ER expression in human aortic stenosis together with the expression of hypertrophy-related genes. METHODS AND RESULTS: ER-alpha and -beta, calcineurin A-beta, and brain natriuretic peptide (BNP) mRNA were quantified by real-time polymerase chain reaction in left ventricular biopsies from patients with aortic valve stenosis (n=14) and control hearts with normal systolic function (n=17). ER protein was quantified by immunoblotting and visualized by immunofluorescence confocal microscopy. ER-alpha mRNA and protein were increased 2.6-fold (P=0.003) and 1.7-fold (P=0.026), respectively, in patients with aortic valve stenosis. Left ventricular ER-beta mRNA was increased 2.6-fold in patients with aortic valve stenosis (P<0.0001). ER-alpha and -beta were found in the cytoplasm and nuclei of human hearts. A strong inverse correlation exists between ER-beta and calcineurin A-beta mRNA in patients with aortic valve stenosis (r=-0.83, P=0.002) but not between ER-alpha or -beta and BNP mRNA. CONCLUSIONS: ER-alpha and -beta in the human heart are upregulated by myocardial pressure load.


Subject(s)
Aortic Valve Stenosis/metabolism , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Up-Regulation/physiology , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/genetics , Calcineurin/biosynthesis , Calcineurin/genetics , Cell Nucleus/metabolism , Cytoplasm/metabolism , Diuretics/pharmacology , Diuretics/therapeutic use , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation , Heart Ventricles/metabolism , Hormone Replacement Therapy , Humans , Male , Microscopy, Fluorescence , Middle Aged , Natriuretic Peptide, Brain/biosynthesis , Natriuretic Peptide, Brain/genetics , Pressure , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
12.
Circ Res ; 95(11): 1058-66, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15514164

ABSTRACT

Collagen types I and III, coded by COL1A1/COL1A2 and COL3A1 genes, are the major fibrillar collagens produced by fibroblasts, including cardiac fibroblasts of the adult heart. Characteristic for different cardiomyopathies is a remodeling process associated with an upregulation of collagen synthesis, which leads to fibrosis. We report identification of three mRNA-binding proteins, heterogeneous nuclear ribonucleoprote (hnRNP) A1, E1, and K, as positive effectors of collagen synthesis acting at the post-transcriptional level by interaction with the 3'-untranslated regions (3'-UTRs) of COL1A1, 1A2, and 3A1 mRNAs. In vitro, binding experiments (electromobility shift assay and UV cross-linking) reveal significant differences in binding to CU- and AU-rich binding motifs. Reporter gene cell transfection experiments and RNA stability assays show that hnRNPs A1, E1, and K stimulate collagen expression by stabilizing mRNAs. Collagen synthesis is activated via the angiotensin II type 1 (AT1) receptor. We demonstrate that transforming growth factor-beta1, a major product of stimulated AT1 receptor, does not activate solely collagen synthesis but synergistically the synthesis of hnRNP A1, E1, and K as well. Thus, post-transcriptional control of collagen synthesis at the mRNA level may substantially be caused by alteration of the expression of RNA-binding proteins. The pathophysiological impact of this finding was demonstrated by screening the expression of hnRNP E1 and K in cardiovascular diseases. In the heart muscle of patients experiencing aortic stenosis, ischemic cardiomyopathy, or dilatative cardiomyopathy, a significant increase in the expression of hnRNP E1, A1, and K was found between 1.5- and 4.5-fold relative to controls.


Subject(s)
3' Untranslated Regions/metabolism , Aortic Valve Stenosis/metabolism , Cardiomyopathy, Dilated/metabolism , Collagen Type III/genetics , Collagen Type I/genetics , Collagen/genetics , Gene Expression Regulation/physiology , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Myocardial Ischemia/metabolism , Ribonucleoproteins/physiology , Thymus Hormones/physiology , Aortic Valve Stenosis/genetics , Base Sequence , Cardiomyopathy, Dilated/genetics , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Collagen/biosynthesis , Collagen Type I/biosynthesis , Collagen Type I, alpha 1 Chain , Collagen Type III/biosynthesis , DNA-Binding Proteins , Fibroblasts/drug effects , Fibroblasts/metabolism , Heart Failure/metabolism , Heart Failure/pathology , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Heterogeneous-Nuclear Ribonucleoprotein K , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Molecular Sequence Data , Myocardial Ischemia/genetics , Protein Binding , Protein Interaction Mapping , RNA, Messenger/biosynthesis , RNA-Binding Proteins , Receptor, Angiotensin, Type 1/physiology , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/metabolism , Thymus Hormones/metabolism , Transfection , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
13.
Eur J Cardiothorac Surg ; 26(1): 89-95, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15200985

ABSTRACT

OBJECTIVE: Coronary artery bypass grafting (CABG) in patients with endstage coronary disease (CAD) significantly improves symptoms and prolongs life expectancy. Left ventricular function is also improved in some patients, but not in others. Factors which influence functional recovery of hibernating myocardium after revascularization are at present under investigation. METHODS: From 3/2000 to 8/2002, we analyzed 41 patients with an ejection fraction (EF) of < or =30%, who underwent CABG, prospectively. All patients received low-dose dobutamine echocardiography (DE), dobutamine myocardial scintigraphy with SPECT, dobutamine magnetic resonance tomography (MRI), contrast-enhanced MRI and, when necessary, positron emission tomography (PET). Hibernating myocardium (area of interest) was identified with these diagnostic tools preoperatively and biopsy samples were taken intraoperatively. RESULTS: All patients received complete coronary revascularization. Early mortality was 2.4%. Three patients died during follow-up. Six months after the operation DE, MRI and SPECT were repeated. EF increased in 23 patients (group I) by at least >or =5%, and in 14 patients (group II) it did not improve. The wall motion score in the area of interest had increased during preoperative DE in group I significantly. The score did not change in group II. In addition the diastolic-systolic wall thickness increase in the area of interest rose >15% during DE in group I preoperatively; the increase was < or =15% in group II. MRI hyperenhancement of the left ventricle was significantly lower in group I compared to group II preoperatively. SPECT showed myocardial viability in the area of interest in all 37 patients. There were no significant differences between group I and II seen in SPECT. When the area of interest was located in the anterior wall the patients more frequently showed ventricular improvement postoperatively than patients with an area of interest located in the inferior, lateral or posterior wall. Light microscopy showed more severe myocardial cell hypertrophy (>19 microm) and less severe destruction of myocardial cell architecture in biopsies of group I compared to group II (myocardial cell hypertrophy < or =17 microm). Electron microscopy showed mitochondrial abnormalities in size and shape, lack of contractile material and large areas containing nonspecified cytoplasm, lipid droplets, and large glycogen-filled regions, but no significant differences between the two groups. Gene expresssion of the pro-apoptotic genes BAK and BAX was lowered compared to expression in 'normal' myocardium. The anti-apoptotic gene BCL-XL was significantly more expressed in the 'area of interest' of group II patients than in group I patients. CONCLUSIONS: We conclude that in patients with endstage CAD myocardial recovery after coronary revascularization can be predicted using DE and MRI preoperatively. Myocardial regions without any potential of functional recovery show less adaptation (less pronounced myocardial cell hypertrophy), a more severe degree of myocardial architecture destruction and a higher degree of anti-apoptotic gene expression. We recommend a myocardial biopsy when DE and MRI are not favorable in a patient with end stage coronary artery disease referred to us with the option of heart transplantation or coronary bypass.


Subject(s)
Coronary Artery Bypass , Myocardial Stunning/surgery , Adult , Aged , Aged, 80 and over , Cardiotonic Agents , Dobutamine , Female , Humans , Male , Middle Aged , Myocardial Stunning/diagnostic imaging , Myocardial Stunning/pathology , Myocardial Stunning/physiopathology , Preoperative Care/methods , Prognosis , Prospective Studies , Stroke Volume , Treatment Outcome , Ultrasonography , Ventricular Function, Left
14.
Chin Med J (Engl) ; 117(3): 401-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15043781

ABSTRACT

BACKGROUND: The purposes of this study were to confirm the changes in myocardial collagen level after left ventricular assist device (LVAD) support in dilated cardiomyopathy (DCM), find the relation between these changes and prognosis, and test a practical method to assess the level of myocardial collagen. METHODS: Left ventricular samples were collected from DCM patients with different prognosis (transplanted group n = 8, weaning group n = 10) at the time when the LVADs were implanted and again during cardiac transplantation (n = 8). The level of neutral salt soluble collagen (NSC) and acid soluble collagen (ASC) was measured by Sircol collagen assay, and that of total collagen and insoluble collagen (ISC) by quantification of hydroxyproline (Hyp). Serum samples were collected from a portion of these patients (transplanted group, n = 6; weaning group n = 7) at the time the LVADs were implanted, 1 month after implantation and on explantation. Circulating concentration of carboxy-terminal propeptide of type I procollagen (P I CP), amino-terminal propeptide of type I procollagen (P I NP), amino-terminal propeptide of type III procollagen (P III NP) and type I collagen telopeptide (I CTP) were measured by the equilibrium type radioimmunoassay. RESULTS: Before LVAD implantation the level of NSC and ISC in the weaning group was higher but ASC in the transplanted group was lower than in the controls (P < 0.05). After LVAD support, the level of total collagen was higher, but ASC was also lower in the transplanted group than in the controls (P < 0.05). In comparison of the pre- and post-LVAD subgroups of the transplanted and weaning groups, all collagen fraction levels before LVAD implantation were lower in the transplanted group than in the weaning group (P < 0.05); but this difference disappeared after LVAD support. Comparison of the pre- and post-LVAD subgroups of the transplanted group showed increased level of NSC and total collagen after LVAD support. The changes of serum peptide concentration showed that P III NP increased constantly in the transplanted group, but P I CP and P I NP increased in the weaning group after LVAD implantation. CONCLUSIONS: The changes in myocardial collagen level as a sign of myocardial interstitial remodeling in DCM are not involved with total collagen but involved with collagen fractions, and they are related to prognosis. The changes of myocardial collagen content and serum procollagen peptide after LVAD support can be regarded as an expression of the reverse of maladaptive myocardial interstitial remodeling.


Subject(s)
Cardiomyopathy, Dilated/physiopathology , Collagen/analysis , Heart-Assist Devices , Myocardium/chemistry , Adult , Cardiomyopathy, Dilated/therapy , Female , Heart Transplantation , Humans , Hydroxyproline/analysis , Male , Middle Aged , Procollagen/blood , Prognosis
15.
Circulation ; 107(19): 2467-74, 2003 May 20.
Article in English | MEDLINE | ID: mdl-12742993

ABSTRACT

BACKGROUND: We present the first genome-wide cDNA array analysis of human congenitally malformed hearts and attempted to partially elucidate these complex phenotypes. Most congenital heart defects, which account for the largest number of birth defects in humans, represent complex genetic disorders. As a consequence of the malformation, abnormal hemodynamic features occur and cause an adaptation process of the heart. METHODS AND RESULTS: The statistical analysis of our data suggests distinct gene expression profiles associated with tetralogy of Fallot, ventricular septal defect, and right ventricular hypertrophy. Applying correspondence analysis, we could associate specific gene functions to specific phenotypes. Furthermore, our study design allows the suggestion that alterations associated with primary genetic abnormalities can be distinguished from those associated with the adaptive response of the heart to the malformation (right ventricular pressure overload hypertrophy). We provide evidence for the molecular transition of the hypertrophic right ventricle to normal left ventricular characteristics. Furthermore, we present data on chamber-specific gene expression. CONCLUSIONS: Our findings propose that array analysis of malformed human hearts opens a new window to understand the complex genetic network of cardiac development and adaptation. For detailed access, see the online-only Data Supplement.


Subject(s)
Gene Expression Profiling , Genome , Heart Defects, Congenital/genetics , Oligonucleotide Array Sequence Analysis , Adaptation, Physiological , Cluster Analysis , Heart Septal Defects, Ventricular/genetics , Humans , Hypertrophy, Right Ventricular/genetics , Multivariate Analysis , Phenotype , Reference Values , Sample Size , Tetralogy of Fallot/genetics
16.
Cardiol Young ; 13(1): 36-43, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12691286

ABSTRACT

BACKGROUND: Cardiac remodelling is now recognised as an important aspect of cardiovascular disease progression and is, therefore, emerging as a therapeutic target in cardiac failure due to different etiologies. Little is known about the influence of different therapies for cardiac failure on the remodelling seen in infants with congenital cardiac disease. METHODS: During follow-up of a prospective and randomized trial, we investigated therapeutic effects on neurohormonal activation, ventricular function, and myocardial gene expression. We compared the data from 8 infants with severe congestive heart failure due to left-to-right shunts, who received digoxin and diuretics alone, to 9 infants who received additional treatment with propranolol. RESULTS: In these infants, beta-adrenergic blockade significantly reduced highly elevated levels of renin, from 284 +/- 319 microU/ml compared to 1061 +/- 769 microU/ml. Systolic ventricular function was normal in both groups, but diastolic ventricular function was improved in those receiving propranolol, indicated by significantly lower left atrial pressures, lower end-diastolic pressures, and less pronounced ventricular hypertrophy, the latter estimated by lower ratios of myocardial wall to ventricular cavity areas on average of 42%. Further hemodynamic parameters showed no significant differences between the groups, except for the lower heart rate in infants treated with propranolol. In those treated with digoxin and diuretics, there was a significant downregulation of beta2-receptor and angiotensin-2 receptor genes, and up-regulation of endothelin A receptor and connective tissue growth factor genes, that were partially prevented by additional treatment with propranolol. CONCLUSIONS: Beta-blockade is a new therapeutic approach for congestive heart failure in infants with congenital cardiac disease, producing with significant benefits on neurohormonal activation, diastolic ventricular function, and cardiac remodelling.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Digoxin/administration & dosage , Diuretics/administration & dosage , Heart Defects, Congenital/complications , Heart Failure/drug therapy , Propranolol/administration & dosage , Ventricular Remodeling/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , Heart Defects, Congenital/diagnosis , Heart Failure/etiology , Heart Failure/mortality , Heart Function Tests , Hemodynamics/drug effects , Humans , Infant , Male , Probability , Prognosis , Prospective Studies , Reference Values , Risk Assessment , Severity of Illness Index , Statistics, Nonparametric , Survival Rate , Treatment Outcome
17.
Hum Mutat ; 20(3): 153-61, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12203987

ABSTRACT

The Marfan syndrome (MFS) is a pleiotropic, autosomal dominant disorder of connective tissue with highly variable clinical manifestations including aortic dilatation and dissection, ectopia lentis, and a series of skeletal anomalies. Mutations in the gene for fibrillin-1 (FBN1) cause MFS, and at least 337 mainly unique mutations have been published to date. FBN1 mutations have been found not only in MFS but also in a range of connective tissue disorders collectively termed fibrillinopathies ranging from mild phenotypes, such as isolated ectopia lentis, to severe disorders including neonatal MFS, which generally leads to death within the first two years of life. The present article intends to provide an overview of mutations found in MFS and related disorders and to discuss potential genotype-phenotype correlations in MFS.


Subject(s)
Marfan Syndrome/genetics , Microfilament Proteins/genetics , Fibrillin-1 , Fibrillins , Genotype , Humans , Marfan Syndrome/pathology , Mutation , Phenotype
18.
Hum Mutat ; 20(3): 197-208, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12203992

ABSTRACT

Mutations in the gene for fibrillin-1 (FBN1) cause Marfan syndrome (MFS), an autosomal dominant heritable disorder of connective tissue with prominent manifestations in the skeletal, ocular, and cardiovascular system. FBN1 mutations have also been identified in a series of related disorders of connective tissue collectively termed type-1 fibrillinopathies. We have developed temperature-gradient gel electrophoresis (TGGE) assays for all 65 FBN1 exons, screened 126 individuals with MFS, other type-1 fibrillinopathies, and other potentially related disorders of connective tissue for FBN1 mutations, and identified a total of 53 mutations, of which 33 are described here for the first time. Several mutations were identified in individuals with fibrillinopathies other than classic Marfan syndrome, including aneurysm of the ascending aorta with only minor skeletal anomalies, and several individuals with only skeletal and ocular involvement. The mutation detection rate in this study was 42% overall, but was only 12% in individuals not fulfilling the diagnostic criteria for MFS, suggesting that clinical overdiagnosis is one reason for the low detection rate observed for FBN1 mutation analysis.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Exons/genetics , Marfan Syndrome/genetics , Microfilament Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Female , Fibrillin-1 , Fibrillins , Humans , Male , Marfan Syndrome/pathology , Mutation , Polymerase Chain Reaction
19.
Ann Thorac Surg ; 73(2): 610-3, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11845882

ABSTRACT

BACKGROUND: Neurohormonal activation in children with heart failure due to congenital heart disease leads to downregulation of myocardial beta-receptors that may influence the postoperative course after cardiothoracic surgery. METHODS: Myocardial biopsies of 26 children (aged 14+/-4 months) were obtained from the right atrium during cardiac surgery. Patients were allocated to either of two groups based on the duration of their intensive care unit stay: group 1 comprised those who stayed less than 7 days (n = 17), whereas group 2 comprised those who stayed more than 7 days, plus 3 infants who died during the early postoperative course (n = 9). For beta1- and beta2-mRNA quantitation, real-time polymerase chain reaction with fluorescence-labeled products was used. RESULTS: Values for myocardial beta1-receptor gene expression were twice as high in group 1 children compared with group 2 (beta1-receptor 0.12+/-0.07 versus 0.06+/-0.03, p = 0.0016; beta2-receptor 0.12+/-0.07 versus 0.06+/-0.03, p = 0.0071). Beta-receptor gene expression in 16 children who received standard treatment for heart failure averaged lower than in the 10 children who received additional propranolol. CONCLUSIONS: Beta-receptor downregulation due to congestive heart failure has an impact on the postoperative course in children with congenital disease and depends on heart failure therapy.


Subject(s)
Heart Defects, Congenital/surgery , Heart Failure/surgery , Postoperative Complications/physiopathology , Receptors, Adrenergic, beta/physiology , Biopsy , Child, Preschool , Digoxin/administration & dosage , Diuretics/administration & dosage , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Therapy, Combination , Female , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Heart Failure/pathology , Heart Failure/physiopathology , Hospital Mortality , Humans , Infant , Length of Stay , Male , Myocardium/pathology , Postoperative Complications/drug therapy , Postoperative Complications/pathology , Propranolol/administration & dosage , Prospective Studies , Receptors, Adrenergic, beta/drug effects , Risk Factors
20.
Circulation ; 105(3): 286-9, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11804980

ABSTRACT

BACKGROUND: The regulation of the cardiac neutral endopeptidase (EC 24.10, NEP) that degrades bradykinin and natriuretic peptides has been investigated in human cardiac hypertrophy and heart failure. Methods and Results- NEP mRNA was quantitated by real-time polymerase chain reaction (PCR) in left ventricular biopsies from patients with aortic valve stenosis (AS, n=19) and heart failure due to dilated cardiomyopathy (DCM, n=14), and control subjects with normal systolic function (CON, n=14). Left ventricular NEP mRNA content was increased 3-fold in AS (P<0.005) and 4.1-fold in DCM (P<0.002). The increase in NEP mRNA was related to the increase in end diastolic pressure in AS and DCM. In a second series, myocardial NEP enzymatic activity was determined. It increased 3.6-fold in AS (P<0.02) and 4-fold in DCM (P<0.002). NEP was localized in the myocardium by immunofluorescence microscopy and in situ PCR to myocytes and nonmyocyte areas and cells. CONCLUSIONS: Elevated cardiac NEP activity in pressure loaded and failing human hearts may increase the local degradation of bradykinin and natriuretic peptides.


Subject(s)
Aortic Valve Stenosis/enzymology , Heart Failure/enzymology , Neprilysin/metabolism , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/genetics , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/genetics , Enzyme Activation , Fluorescent Antibody Technique , Heart Failure/drug therapy , Heart Failure/genetics , Heart Ventricles/enzymology , Humans , Microscopy, Fluorescence , Myocardium/enzymology , Neprilysin/genetics , RNA, Messenger/biosynthesis , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...