Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Histochem ; 56(2): e15, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22688296

ABSTRACT

Sodium nitroprusside (SNP) is used clinically as a rapid-acting vasodilator and in experimental models as donor of nitric oxide (NO). High concentrations of NO have been reported to induce cardiotoxic effects including apoptosis by the formation of reactive oxygen species. We have therefore investigated effects of SNP on the myofibrillar cytoskeleton, contractility and cell death in long-term cultured adult rat cardiomyocytes at different time points after treatment. Our results show, that SNP treatment at first results in a gradual increase of cytoskeleton degradation marked by the loss of actin labeling and fragmentation of sarcomeric structure, followed by the appearance of TUNEL-positive nuclei. Already lower doses of SNP decreased contractility of cardiomyocytes paced at 2 Hz without changes of intracellular calcium concentration. Ultrastructural analysis of the cultured cells demonstrated mitochondrial changes and disintegration of sarcomeric alignment. These adverse effects of SNP in cardiomyocytes were reminiscent of anthracycline-induced cardiotoxicity, which also involves a dysregulation of NO with the consequence of myofibrillar degradation and ultimately cell death. An inhibition of the pathways leading to the generation of reactive NO products, or their neutralization, may be of significant therapeutic benefit for both SNP and anthracycline-induced cardiotoxicity.


Subject(s)
Anthracyclines/adverse effects , Cardiotoxins/adverse effects , Cytoskeleton , Myocytes, Cardiac , Nitroprusside/adverse effects , Animals , Anthracyclines/pharmacology , Cardiotoxins/pharmacology , Cell Death/drug effects , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/pathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitroprusside/pharmacology , Rats , Rats, Wistar
2.
Physiol Res ; 59(1): 35-42, 2010.
Article in English | MEDLINE | ID: mdl-19249908

ABSTRACT

Left ventricular hypertrophy (LVH) is due to pressure overload or mechanical stretch and is thought to be associated with remodeling of gap-junctions. We investigated whether the expression of connexin 43 (Cx43) is altered in humans in response to different degrees of LVH. The expression of Cx43 was analyzed by quantitative polymerase chain reaction, Western blot analysis and immunohistochemistry on left ventricular biopsies from patients undergoing aortic or mitral valve replacement. Three groups were analyzed: patients with aortic stenosis with severe LVH (n=9) versus only mild LVH (n=7), and patients with LVH caused by mitral regurgitation (n=5). Cx43 mRNA expression and protein expression were similar in the three groups studied. Furthermore, immunohistochemistry revealed no change in Cx43 distribution. We can conclude that when compared with mild LVH or with LVH due to volume overload, severe LVH due to chronic pressure overload is not accompanied by detectable changes of Cx43 expression or spatial distribution.


Subject(s)
Aortic Valve Stenosis/complications , Connexin 43/analysis , Hypertrophy, Left Ventricular/mortality , Mitral Valve Insufficiency/complications , Myocardium/chemistry , Aged , Aged, 80 and over , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/physiopathology , Biopsy , Blood Pressure , Blotting, Western , Connexin 43/genetics , Female , Gene Expression Regulation , Humans , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Immunohistochemistry , Male , Middle Aged , Mitral Valve Insufficiency/metabolism , Mitral Valve Insufficiency/pathology , Mitral Valve Insufficiency/physiopathology , Myocardium/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Ventricular Function, Left
3.
J Cell Biol ; 153(4): 763-72, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11352937

ABSTRACT

In this study, we investigated cardiomyocyte cytoarchitecture in a mouse model for dilated cardiomyopathy (DCM), the muscle LIM protein (MLP) knockout mouse and substantiated several observations in a second DCM model, the tropomodulin-overexpressing transgenic (TOT) mouse. Freshly isolated cardiomyocytes from both strains are characterized by a more irregular shape compared with wild-type cells. Alterations are observed at the intercalated disks, the specialized areas of mechanical coupling between cardiomyocytes, whereas the subcellular organization of contractile proteins in the sarcomeres of MLP knockout mice appears unchanged. Distinct parts of the intercalated disks are affected differently. Components from the adherens junctions are upregulated, desmosomal proteins are unchanged, and gap junction proteins are downregulated. In addition, the expression of N-RAP, a LIM domain- containing protein located at the intercalated disks, is upregulated in MLP knockout as well as in TOT mice. Detailed analysis of intercalated disk composition during postnatal development reveals that an upregulation of N-RAP expression might serve as an early marker for the development of DCM. Altered expression levels of cytoskeletal proteins (either the lack of MLP or an increased expression of tropomodulin) apparently lead to impaired function of the myofibrillar apparatus and to physiological stress that ultimately results in DCM and is accompanied by an altered appearance and composition of the intercalated disks.


Subject(s)
Microfilament Proteins , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gap Junctions/metabolism , Gene Expression/physiology , LIM Domain Proteins , Mice , Mice, Knockout , Microscopy, Electron , Muscle Fibers, Skeletal/pathology , Sarcomeres/metabolism , Tropomodulin
4.
J Cell Sci ; 113 ( Pt 22): 4121-35, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11058098

ABSTRACT

The cytoplasmic domain of the transmembrane protein M-cadherin is involved in anchoring cytoskeletal elements to the plasma membrane at cell-cell contact sites. Several members of the armadillo repeat protein family mediate this linkage. We show here that ARVCF, a member of the p120 (ctn) subfamily, is a ligand for the cytoplasmic domain of M-cadherin, and characterize the regions involved in this interaction in detail. Complex formation in an in vivo environment was demonstrated in (1) yeast two-hybrid screens, using a cDNA library from differentiating skeletal muscle and part of the cytoplasmic M-cadherin tail as a bait, and (2) mammalian cells, using a novel experimental system, the MOM recruitment assay. Immunoprecipitation and in vitro binding assays confirmed this interaction. Ectopically expressed EGFP-ARVCF-C11, an N-terminal truncated fragment, targets to junctional structures in epithelial MCF7 cells and cardiomyocytes, where it colocalizes with the respective cadherins, beta-catenin and p120 (ctn). Hence, the N terminus of ARVCF is not required for junctional localization. In contrast, deletion of the four N-terminal armadillo repeats abolishes this ability in cardiomyocytes. Detailed mutational analysis revealed the armadillo repeat region of ARVCF as sufficient and necessary for interaction with the 55 membrane-proximal amino acids of the M-cadherin tail.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Intercellular Junctions/physiology , Muscles/physiology , Muscles/ultrastructure , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Armadillo Domain Proteins , Binding Sites , Cadherins/chemistry , Cell Membrane/physiology , Cell Membrane/ultrastructure , Cells, Cultured , Heart Ventricles , Humans , Intercellular Junctions/ultrastructure , Ligands , Mice , Molecular Sequence Data , Muscles/cytology , Myocardium/ultrastructure , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
5.
J Mol Cell Cardiol ; 32(4): 539-55, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10756112

ABSTRACT

We investigated dynamic events during the formation of intercalated disc-like structures of adult rat cardiomyocytes (ARC) in long-term culture. Given the complexity of ARC cytoIarchitecture after de- and re-differentiation, and the non-uniform morphological development of individual cells, green fluorescent protein (GFP) technology was used to track N-cadherin in living cells. Sorting and functionality of the GFP fusion protein was tested in ARC. Isolated ARC were micro-injected with the expression construct at the onset of spreading in culture, and the fluorescence signals were tracked during contact formation and in fully redifferentiated living cells. The first contact sites were found to be established by cellular protrusions, which were marked by an ultrastructure similar to microspikes and probably have a role as exploratory units in the spreading phase. Subsequently, initial contact sites served as anchorage for the most prominent stress fibre-like structures. The fusion protein appeared before connexin-43 at newly established cell-cell contacts. Membrane invaginations at the sarcolemma facing the substratum of cultured ARC may be responsible for the appearance of a striped pattern of N-cadherin and other adherens junction proteins away from intercalated disc-like structures. The stripes were immobile in redifferentiated cells, while the distinct small fluorescent particles in the cell body were found to move directionally at speeds around 10 micro m/min. These results contribute to the understanding of the mechanisms of cell-cell contact formation of adult cardiomyocytes, which is a prerequisite for any future implantation technology.


Subject(s)
Cadherins/metabolism , Cell Communication/physiology , Luminescent Proteins/metabolism , Animals , CHO Cells , Cadherins/genetics , Cells, Cultured , Cricetinae , Green Fluorescent Proteins , Intercellular Junctions/ultrastructure , Luminescent Proteins/genetics , Myocardium/cytology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
Heart Fail Rev ; 5(3): 251-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-16228908

ABSTRACT

N-Cadherin belongs to a superfamily of calcium-dependent transmembrane adhesion proteins. It mediates adhesion in the intercalated discs at the termini of cardiomyocytes thereby serving as anchor for myofibrils at cell-cell contacts. A large body of data on the molecular structure and function of N-cadherin exists, however, little is known concerning spatial and temporal interactions between the different junctional structures during formation of the intercalated disc and its maturation in postnatal development. The progression of compensated left ventricular hypertrophy to congestive left heart failure is accompanied by intercalated disc remodeling and has been demonstrated in animal models and in patients. The long-term culture of adult rat cardiomyocytes allows to investigate the development of de novo intercalated disc-like structures. In order to analyze the dynamics of the cytoskeletal redifferentiation in living cells, we used the expression of chimeric proteins tagged with the green fluorescent protein reporter. This technique is becoming a routine method in basic research and complements video time-lapse and confocal microscopy. Cultured cardiomyocytes have been used for a variety of studies in cell biology and pharmacology. Their ability to form an electrically coupled beating tissue-like network in culture possibly allows reimplantation of such cells into injured myocardium, where they eventually will form new contacts with the healthy muscle tissue. Several groups have already shown that cardiomyocytes can be grafted successfully into sites of myocardial infarcts or cryoinjuries. Autologous adult cardiomyocyte implantation, might indeed contribute to cardiac repair after infarction, thanks to advances in tissue engineering.


Subject(s)
Cadherins/physiology , Cadherins/ultrastructure , Intercellular Junctions/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/ultrastructure , Adherens Junctions , Animals , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/physiopathology , Cell Communication/physiology , Cells, Cultured , Chick Embryo , Disease Models, Animal , Mice , Microscopy, Electron , Rats , Reference Values , Sensitivity and Specificity
7.
FASEB J ; 13 Suppl: S83-9, 1999.
Article in English | MEDLINE | ID: mdl-10352149

ABSTRACT

Primary adult rat cardiomyocytes (ARC)in culture are shown to be a model system for cardiac cell hypertrophy in vitro. ARC undergo a process of morphological transformation and grow only by increase in cell size, however, without loss of the cardiac phenotype. The isolated cells spread and establish new cell-cell contacts, eventually forming a two-dimensional heart tissue-like synchronously beating cell sheet. The reformation of specific cell contacts (intercalated disks) is shown also between ventricular and atrial cardiomyocytes by using antibodies against the gap junction protein connexin-43 and after microinjection into ARC of N-cadherin cDNA fused to reporter green fluorescent protein (GFP) cDNA. The expressed fusion protein allowed the study of live cell cultures and of the dynamics of the adherens junction protein N-cadherin during the formation of new cell-cell contacts. The possible use of the formed ARC cell-sheet cells under microgravity conditions as a test system for the reformation of the cytoskeleton of heart muscle cells is proposed.


Subject(s)
Cell Communication , Myocardium/cytology , Animals , Cadherins/genetics , Cell Membrane/metabolism , Cells, Cultured , DNA, Complementary , Female , Green Fluorescent Proteins , Luminescent Proteins/genetics , Microscopy, Electron , Models, Biological , Myocardium/metabolism , Myocardium/ultrastructure , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics
8.
J Mol Cell Cardiol ; 29(8): 2027-39, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9281436

ABSTRACT

In the present study, we compare expression, storage and secretion of the atrial natriuretic factor (ANF) in atrial and ventricular adult rat cardiomyocytes (aARC and vARC) in long-term culture. The influence of insulin-like growth factor-I (IGF-I) and of basic fibroblast growth factor (bFGF) on ANF production and secretion, as well as on the expression of a structural component, alpha-smooth muscle actin (alpha-sm actin), was studied in the two cell types. Antibodies against alpha-ANF were used for immunocytochemical localization of ANF. aARC contained more ANF-granules than vARC, and they were distributed throughout the cell bodies. Quantitative determination of ANF storage and secretion was done by radioimmunoassay (RIA; 125I), and it was demonstrated that aARC stored and secreted ANF 18- and 16-times more, respectively, when compared to vARC. Immuno-electron microscopy confirmed that ANF storing secretory granules were present in both types of cardiomyocytes. Expression of ANF and alpha-sm actin in aARC and vARC responded differently to treatment with either IGF-I or bFGF. In aARC, neither IGF-I nor bFGF had an influence on expression of ANF. In vARC, expression of ANF was downregulated by IGF-I and upregulated by bFGF with regard to both immunoreactivity and message. In contrast to vARC, expression of alpha-sm actin was not affected by IGF-I in aARC, whereas bFGF produced a strong upregulation similar to that found in vARC. Mitogen-activated protein kinases (MAPK) 42 and 44, though, were equally activated by bFGF and IGF-I in both aARC and vARC.


Subject(s)
Actins/biosynthesis , Atrial Natriuretic Factor/biosynthesis , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Heart Atria/drug effects , Heart Ventricles/drug effects , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinases , Actins/genetics , Animals , Atrial Natriuretic Factor/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Female , Heart Atria/cytology , Heart Ventricles/cytology , Hypertrophy , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Organ Specificity , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...