Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Cell Commun Adhes ; 14(5): 239-49, 2007.
Article in English | MEDLINE | ID: mdl-18163233

ABSTRACT

Ischemic preconditioning increases the heart's tolerance to a subsequent longer ischemic period. The aim of this study was to investigate the effect of early and delayed preconditioning on gap junction communication, connexin abundance, and phosphorylation in cultured neonatal rat cardiac myocytes. Prolonged ischemia followed 5 minutes after preconditioning in the early protocol, whereas 20 hours separated preconditioning and prolonged ischemia in the delayed preconditioning protocol. Gap junctional intercellular communication (GJIC) was assessed by Lucifer yellow dye transfer. An initial reduction in communication in response to sublethal ischemia was observed. This may be one mechanism whereby neighboring cells are protected from damaging substances produced during the first phase of subsequent regional ischemia in early preconditioning protocols. With respect to delayed preconditioning, the transient decrease in GJIC disappeared prior to prolonged ischemia, indicating that other mechanisms are responsible for delayed protection. Both early and delayed preconditioning preserved intercellular coupling after prolonged ischemia and this correlated with presence of less connexin43 dephosphorylation assessed by immunoblot.


Subject(s)
Cytoprotection/physiology , Gap Junctions/metabolism , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cell Communication/physiology , Cells, Cultured , Connexin 43/metabolism , Connexins/metabolism , Isoquinolines , Myocardium/ultrastructure , Myocytes, Cardiac/ultrastructure , Phosphorylation , Rats , Rats, Wistar
2.
Cardiovasc Pathol ; 10(4): 169-77, 2001.
Article in English | MEDLINE | ID: mdl-11600334

ABSTRACT

Electrical activation of the heart requires cell-cell transfer of current via gap junctions, arrays of densely packed protein channels that permit intercellular passage of ions and small molecules. Because current transfer occurs only at gap junctions, the spatial distribution and biophysical properties of gap junction channels are important determinants of the conduction properties of cardiac muscle. Gap junction channels are composed of members of a multigene family of proteins called connexins. As a general rule, individual cells express multiple connexins, which creates the potential for considerable functional diversity in gap junction channels. Although gap junction channels are relatively nonselective in their permeability to ions and small molecules, cardiac myocytes actively adjust their level of coupling by multiple mechanisms including changes in connexin expression, regulation of connexin trafficking and turnover, and modulation of channel properties. In advanced stages of heart disease, connexin expression and intercellular coupling are diminished, and gap junction channels become redistributed. These changes have been strongly implicated in the pathogenesis of lethal ventricular arrhythmias. Ongoing studies in genetically engineered mice are revealing insights into the role of individual gap junction channel proteins in normal cardiac function and arrhythmogenesis.


Subject(s)
Arrhythmias, Cardiac/etiology , Gap Junctions/physiology , Heart Conduction System/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Cell Communication , Connexins/chemistry , Connexins/genetics , Connexins/metabolism , Down-Regulation , Gap Junctions/ultrastructure , Humans , Multigene Family , Myocardium/cytology , Myocardium/ultrastructure , Protein Conformation , Up-Regulation
3.
Cardiovasc Res ; 51(4): 681-90, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11530101

ABSTRACT

UNLABELLED: Analysis of mice with genetically altered expression of cardiac connexins can provide insights into the role of individual gap junction channel proteins in cell-to-cell communication, impulse propagation, and arrhythmias. However, conflicting results have been reported regarding conduction velocity slowing in mice heterozygous for a null mutation in the gene encoding connexin43 (Cx43). METHODS: High-resolution optical mapping was used to record action potentials from 256 sites, simultaneously, on the ventricular surface of Langendorff perfused hearts from 15 heterozygous (Cx43+/-) and 8 wildtype (Cx43+/+) mice (controls). A sensitive method for measuring epicardial conduction velocity was developed to minimize confounding influences of subepicardial breakthrough and virtual electrode effects. RESULTS: Epicardial conduction velocity was significantly slower (23 to 35%, P<0.01) in Cx43+/- mice compared to wildtype. There was no change in conduction patterns or anisotropic ratio (Cx43+/- 1.54+/-0.33; Cx43+/+ 1.57+/-0.17) suggesting that Cx43 expression was reduced uniformly throughout myocardium. The magnitude of reductions in conduction velocity and Cx43 protein expression (45%) were similar in mice in which the null allele occurred in a pure C57BL/6J genetic background versus a mixed (C57BL/6J X 129) background. Action potential duration did not differ between mice of different genotypes. CONCLUSIONS: A approximately 50% reduction of Cx43 expression causes significant conduction velocity slowing in the Cx43+/- mouse heart. The apparent lack of conduction velocity changes reported in previous studies may be related to technical factors rather than variations in genetic background. High-resolution optical mapping is a powerful tool for investigating molecular determinants of propagation and arrhythmias in genetically engineered mice.


Subject(s)
Connexin 43/genetics , Myocardium/metabolism , Action Potentials , Analysis of Variance , Animals , Connexin 43/metabolism , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Myocardium/chemistry , Perfusion , Purkinje Fibers/pathology , Video Recording
5.
J Clin Invest ; 107(7): 813-22, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285300

ABSTRACT

Inherited and acquired cardiomyopathies are associated with marked intracellular lipid accumulation in the heart. To test the hypothesis that mismatch between myocardial fatty acid uptake and utilization leads to the accumulation of cardiotoxic lipid species, and to establish a mouse model of metabolic cardiomyopathy, we generated transgenic mouse lines that overexpress long-chain acyl-CoA synthetase in the heart (MHC-ACS). This protein plays an important role in vectorial fatty acid transport across the plasma membrane. MHC-ACS mice demonstrate cardiac-restricted expression of the transgene and marked cardiac myocyte triglyceride accumulation. Lipid accumulation is associated with initial cardiac hypertrophy, followed by the development of left-ventricular dysfunction and premature death. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and cytochrome c release in transgenic hearts suggest that cardiac myocyte death occurs, in part, by lipid-induced programmed cell death. Taken together, our data demonstrate that fatty acid uptake/utilization mismatch in the heart leads to accumulation of lipid species toxic to cardiac myocytes. This novel mouse model will provide insight into the role of perturbations in myocardial lipid metabolism in the pathogenesis of inherited and acquired forms of heart failure.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Lipid Metabolism , Myocardium/metabolism , Repressor Proteins , Saccharomyces cerevisiae Proteins , Animals , Cardiomyopathy, Dilated/etiology , Cells, Cultured , Coenzyme A Ligases/biosynthesis , Coenzyme A Ligases/genetics , Disease Models, Animal , Female , Heart Failure/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/cytology
6.
Am J Physiol Heart Circ Physiol ; 280(3): H1232-40, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179068

ABSTRACT

This study reports the comparative quantitative, morphological, and electrophysiological properties of two pacemaker cell types, spider and spindle-shaped cells, isolated from the rabbit sinoatrial node. Isolated nodal cells were studied with perforated and ruptured patch whole cell recording techniques. The basic spontaneous cycle length of the spider cells was 381 +/- 12 ms, and the basic spontaneous cycle length of the spindle cells was 456 +/- 17 ms (n = 12, P < 0.05). The spider cells had a more positive maximum diastolic potential (-54 +/- 1 mV) compared with the spindle cells (-68 +/- 1mV, P < 0.05). The overshoot and action potential amplitudes were also smaller in the spider cells. The hyperpolarization-activated inward (I(f)) current density, measured from their tail currents, was 15 +/- 1.3 pA/pF for the spider cells and 9 +/- 0.7 pA/pF for the spindle cells (P < 0.01). I(f) current activation voltage was more positive in the spider cells than the spindle cells. Isoproterenol (1 microM) decreased the spontaneous cycle length of the spider cells by 28 +/- 3% and the spindle cells by 20 +/- 1.5% (P < 0.05). Acetylcholine (0.5 microM) hyperpolarized the membrane potential of the spider cells to -86 +/- 0.7 mV and the spindle cells to -76 +/- 0.8 mV (P < 0.05). In summary, there are at least two distinct pacemaker cell types in the sinus node with different electrophysiological characteristics.


Subject(s)
Cell Membrane/physiology , Sinoatrial Node/cytology , Sinoatrial Node/physiology , Acetylcholine/pharmacology , Animals , Biological Clocks/physiology , Cardiotonic Agents/pharmacology , Cell Size/physiology , Female , Isoproterenol/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Rabbits , Vasodilator Agents/pharmacology
7.
J Heart Lung Transplant ; 20(1): 93-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11166617

ABSTRACT

We tested the hypothesis that cardiac allograft dysfunction in acute cardiac rejection may be related, in part, to diminished expression of connexin43, a gap junction channel protein that facilitates intercellular communication and coordinates electrical and mechanical cardiac function. We measured connexin43 levels using quantitative confocal immunofluorescence microscopy of endocardial biopsies from heart transplant recipients with histologic evidence of either no rejection or acute cellular rejection. Expression of connexin43 diminished significantly during acute cellular rejection and returned to baseline levels following resolution of rejection. Reversible down-regulation of connexin43 may contribute to ventricular dysfunction in allograft rejection.


Subject(s)
Connexin 43/metabolism , Down-Regulation , Graft Rejection/metabolism , Heart Transplantation/adverse effects , Myocardium/metabolism , Acute Disease , Adult , Biomarkers , Endocardium/metabolism , Endocardium/pathology , Graft Rejection/pathology , Heart Transplantation/pathology , Humans , Microscopy, Fluorescence , Severity of Illness Index
9.
J Clin Invest ; 106(7): 847-56, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11018072

ABSTRACT

Cardiac mitochondrial function is altered in a variety of inherited and acquired cardiovascular diseases. Recent studies have identified the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) as a regulator of mitochondrial function in tissues specialized for thermogenesis, such as brown adipose. We sought to determine whether PGC-1 controlled mitochondrial biogenesis and energy-producing capacity in the heart, a tissue specialized for high-capacity ATP production. We found that PGC-1 gene expression is induced in the mouse heart after birth and in response to short-term fasting, conditions known to increase cardiac mitochondrial energy production. Forced expression of PGC-1 in cardiac myocytes in culture induced the expression of nuclear and mitochondrial genes involved in multiple mitochondrial energy-transduction/energy-production pathways, increased cellular mitochondrial number, and stimulated coupled respiration. Cardiac-specific overexpression of PGC-1 in transgenic mice resulted in uncontrolled mitochondrial proliferation in cardiac myocytes leading to loss of sarcomeric structure and a dilated cardiomyopathy. These results identify PGC-1 as a critical regulatory molecule in the control of cardiac mitochondrial number and function in response to energy demands.


Subject(s)
Energy Metabolism , Fasting/physiology , Mitochondria, Heart/physiology , Transcription Factors/biosynthesis , Animals , Animals, Newborn , Cardiomyopathy, Dilated/genetics , Cell Nucleus/genetics , Cell Respiration , Cells, Cultured , Female , Gene Expression Regulation , Male , Mice , Mice, Transgenic , Myocardium/cytology , Transcription Factors/genetics
10.
Circ Res ; 87(8): 656-62, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11029400

ABSTRACT

Electrical uncoupling at gap junctions during acute myocardial ischemia contributes to conduction abnormalities and reentrant arrhythmias. Increased levels of intracellular Ca(2+) and H(+) and accumulation of amphipathic lipid metabolites during ischemia promote uncoupling, but other mechanisms may play a role. We tested the hypothesis that uncoupling induced by acute ischemia is associated with changes in phosphorylation of the major cardiac gap junction protein, connexin43 (Cx43). Adult rat hearts perfused on a Langendorff apparatus were subjected to ischemia or ischemia/reperfusion. Changes in coupling were monitored by measuring whole-tissue resistance. Changes in the amount and distribution of phosphorylated and nonphosphorylated isoforms of Cx43 were measured by immunoblotting and confocal immunofluorescence microscopy using isoform-specific antibodies. In control hearts, virtually all Cx43 identified immunohistochemically at apparent intercellular junctions was phosphorylated. During ischemia, however, Cx43 underwent progressive dephosphorylation with a time course similar to that of electrical uncoupling. The total amount of Cx43 did not change, but progressive reduction in total Cx43 immunofluorescent signal and concomitant accumulation of nonphosphorylated Cx43 signal occurred at sites of intercellular junctions. Functional recovery during reperfusion was associated with increased levels of phosphorylated Cx43. These observations suggest that uncoupling induced by ischemia is associated with dephosphorylation of Cx43, accumulation of nonphosphorylated Cx43 within gap junctions, and translocation of Cx43 from gap junctions into intracellular pools.


Subject(s)
Connexin 43/metabolism , Heart Conduction System/metabolism , Heart Ventricles/metabolism , Intracellular Fluid/metabolism , Myocardial Ischemia/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Disease Models, Animal , Electrocardiography , Fluorescent Antibody Technique , Gap Junctions/metabolism , Immunoblotting , In Vitro Techniques , Male , Myocardial Reperfusion , Phosphorylation , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley
11.
Circ Res ; 87(6): 467-73, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10988238

ABSTRACT

The aim of the present study was to morphologically and electrically characterize synthetic strands of mouse ventricular myocytes. Linear strands of mouse ventricular myocytes with widths of 34.7+/-4.4 microm (W(1)), 57.9+/-2.5 microm (W(2)), and 86.4+/-3. 6 microm (W(3)) and a length of 10 mm were produced on glass coverslips with a photolithographic technique. Action potentials (APs) were measured from individual cells within the strands with cell-attached microelectrodes. Impulse propagation and AP upstrokes were measured with multisite optical mapping (RH237). Immunostaining was performed to assess cell-cell connections and myofibril arrangement with polyclonal antisera against connexin43 and N-cadherins and monoclonal antibodies against cardiac myosin. Light microscopy and myosin staining showed dense growth of well-developed elongated myocytes with lengths of 34.2+/-4.2 microm (W(1)), 36. 9+/-5.8 microm (W(2)), and 43.7+/-6.9 microm (W(3)), and length/width ratios of 3.9+/-0.2. Gap junctions were distributed around the cell borders (3 to 4 junctions/microm(2) cell area). Each cell was connected by gap junctions to 6.5+/-1.1 neighboring cells. AP duration shortened with time in culture (action potential duration at 50% repolarization: day 4, 103+/-34 ms; day 8, 16+/-3 ms; P:<0.01). Minimum diastolic potential and AP amplitude were 71+/-5 and 97.2+/-7.6 mV, respectively. Conduction velocity and the maximum dV/dt of the AP upstroke were 43.9+/-13.6 cm/s and 196+/-67 V/s, respectively. Thus, neonatal ventricular mouse myocytes can be grown in continuous synthetic strands. Gap junction distribution is similar to the neonatal pattern observed in the hearts of larger mammals. Conduction velocity is in the range observed in adult mice and in the higher range for mammalian species probably due to the higher dV/dt(max). This technique will permit the study of propagation, AP, and structure-function relations at cellular resolution in genetically modified mice.


Subject(s)
Myocardium/cytology , Action Potentials , Animals , Animals, Newborn , Cell Size , Cells, Cultured , Connexin 43/metabolism , Fluorescent Dyes , Heart Ventricles/cytology , Heart Ventricles/metabolism , Heart Ventricles/ultrastructure , Immunohistochemistry , Intercellular Junctions/metabolism , Intercellular Junctions/physiology , Mice , Microelectrodes , Microscopy, Confocal , Myocardium/metabolism , Myocardium/ultrastructure , Optics and Photonics , Pyridinium Compounds
12.
Circ Res ; 87(4): 316-22, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10948066

ABSTRACT

Mechanical stretch is thought to play an important role in remodeling atrial and ventricular myocardium and may produce substrates that promote arrhythmogenesis. In the present work, neonatal rat ventricular myocytes were cultured for 4 days as confluent monolayers on thin silicone membranes and then subjected to linear pulsatile stretch for up to 6 hours. Action potential upstrokes and propagation velocity (theta) were measured with multisite optical recording of transmembrane voltage of the cells stained with the voltage-sensitive dye RH237. Expression of the gap junction protein connexin43 (Cx43) and the fascia adherens junction protein N-cadherin was measured immunohistochemically in the same preparations. Pulsatile stretch caused dramatic upregulation of intercellular junction proteins after only 1 hour and a further increase after 6 hours (Cx43 signal increased from 0.73 to 1.86 and 2.02% cell area, and N-cadherin signal increased from 1.21 to 2.11 and 2.74% cell area after 1 and 6 hours, respectively). This was paralleled by an increase in theta from 27 to 35 cm/s after 1 hour and 37 cm/s after 6 hours. No significant change in the upstroke velocity of the action potential or cell size was observed. Increased theta and protein expression were not reversible after 24 hours of relaxation. Nonpulsatile (static) stretch produced qualitatively similar but significantly smaller changes than pulsatile stretch. Thus, pulsatile linear stretch in vitro causes marked upregulation of proteins that form electrical and mechanical junctions, as well as a concomitant increase in propagation velocity. These changes may contribute to arrhythmogenesis in myocardium exposed to acute stretch.


Subject(s)
Cell Communication/physiology , Muscle Fibers, Skeletal/metabolism , Myocardial Contraction/physiology , Myocardium/cytology , Ventricular Remodeling/physiology , Action Potentials/physiology , Animals , Cadherins/analysis , Cadherins/biosynthesis , Cell Size/physiology , Cells, Cultured , Connexin 43/analysis , Connexin 43/biosynthesis , Heart Conduction System/physiology , Heart Ventricles/cytology , Muscle Fibers, Skeletal/chemistry , Myocardium/chemistry , Rats , Stress, Mechanical
13.
Braz J Med Biol Res ; 33(4): 407-13, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775305

ABSTRACT

Effective pump function of the heart depends on the precise control of spatial and temporal patterns of electrical activation. Accordingly, the distribution and function of gap junction channels are important determinants of the conduction properties of myocardium and undoubtedly play other roles in intercellular communication crucial to normal cardiac function. Recent advances have begun to elucidate mechanisms by which the heart regulates intercellular electrical coupling at gap junctions in response to stress or injury. Although responses to increased load or injury are generally adaptive in nature, remodeling of intercellular junctions under conditions of severe stress creates anatomic substrates conducive to the development of lethal ventricular arrhythmias. Potential mechanisms controlling the level of intercellular communication in the heart include regulation of connexin turnover dynamics and phosphorylation.


Subject(s)
Cardiomegaly/metabolism , Cell Communication/physiology , Connexins/metabolism , Gap Junctions/metabolism , Heart Failure/metabolism , Acute Disease , Arrhythmias, Cardiac/metabolism , Chronic Disease , Electric Conductivity , Humans , Phosphorylation
14.
Circ Res ; 86(7): 723-8, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10764404

ABSTRACT

Electrical activation of the heart requires current transfer from one cell to another via gap junctions, arrays of densely packed intercellular channels. The extent to which cardiac myocytes are coupled is determined by multiple mechanisms, including tissue-specific patterns of expression of diverse gap junction channel proteins (connexins), and regulatory pathways that control connexin synthesis, intracellular trafficking, assembly into channels, and degradation. Many connexins, including those expressed in the heart, have been found to turn over rapidly. Recent studies in the intact adult heart suggest that connexin43, the principal cardiac connexin, is surprisingly short-lived (half-life approximately 1.3 hours). Both the proteasome and the lysosome participate in connexin43 degradation. Other ion channel proteins, such as those forming selected voltage-gated K(+) channels, may also exhibit rapid turnover kinetics. Regulation of connexin degradation may be an important mechanism for adjusting intercellular coupling in the heart under normal and pathophysiological conditions.


Subject(s)
Connexins/genetics , Connexins/metabolism , Heart/physiology , Adult , Animals , Cysteine Endopeptidases/metabolism , Gap Junctions/physiology , Humans , Lysosomes/physiology , Models, Cardiovascular , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex
15.
Am J Physiol Heart Circ Physiol ; 278(5): H1662-70, 2000 May.
Article in English | MEDLINE | ID: mdl-10775147

ABSTRACT

Gap junction number and size vary widely in cardiac tissues with disparate conduction properties. Little is known about how tissue-specific patterns of intercellular junctions are established and regulated. To elucidate the relationship between gap junction channel protein expression and the structure of gap junctions, we analyzed Cx43 +/- mice, which have a genetic deficiency in expression of the major ventricular gap junction protein, connexin43 (Cx43). Quantitative confocal immunofluorescence microscopy revealed that diminished Cx43 signal in Cx43 +/- mice was due almost entirely to a reduction in the number of individual gap junctions (226 +/- 52 vs. 150 +/- 32 individual gap junctions/field in Cx43 +/+ and +/- ventricles, respectively; P < 0.05). The mean size of an individual gap junction was the same in both groups. Immunofluorescence results were confirmed with electron microscopic morphometry. Thus when connexin expression is diminished, ventricular myocytes become interconnected by a reduced number of large, normally sized gap junctions, rather than a normal number of smaller junctions. Maintenance of large gap junctions may be an adaptive response supporting safe ventricular conduction.


Subject(s)
Connexin 43/biosynthesis , Gap Junctions/ultrastructure , Heart Ventricles/ultrastructure , Myocardium/ultrastructure , Animals , Cadherins/analysis , Connexin 43/analysis , Fascia/ultrastructure , Fluorescent Antibody Technique , Heart Ventricles/metabolism , Heterozygote , Image Processing, Computer-Assisted , Mice , Mice, Mutant Strains , Microscopy, Confocal , Microscopy, Electron , Myocardium/metabolism , Papillary Muscles/ultrastructure
17.
Circulation ; 101(5): 547-52, 2000 Feb 08.
Article in English | MEDLINE | ID: mdl-10662753

ABSTRACT

BACKGROUND: Myocardial ischemia causes profound changes in both active membrane currents and passive electrical properties. Because these complex changes develop and progress concomitantly, it has not been possible to elucidate the relative contributions of any one component to arrhythmogenesis induced by acute ischemia. Cx43+/- mice express 50% of the normal level of connexin43 (Cx43), the major ventricular electrical coupling protein, but are otherwise identical to wild-type (Cx43+/+) mice. Comparison of arrhythmogenesis in Cx43+/- and +/+ mice can provide insights into the role of changes in electrical coupling as an independent variable in the complex setting of acute ischemia. METHODS AND RESULTS: Acute ischemia was induced in isolated perfused mouse hearts by occlusion of the left anterior descending coronary artery. Spontaneous ventricular tachyarrhythmias (VT) occurred in more than twice as many Cx43+/- hearts than Cx43+/+ hearts. VT was induced in nearly 3 times as many Cx43+/- hearts. Multiple runs and prolonged runs of spontaneous VT were more frequent in Cx43+/- hearts. Onset of the first run of VT occurred significantly earlier in Cx43+/- hearts. Premature ventricular beats were also more frequent in Cx43+/- hearts. The size of the hypoperfused region was equivalent in both groups. CONCLUSIONS: Reduced expression of Cx43 accelerates the onset and increases the incidence, frequency, and duration of ventricular tachyarrhythmias after coronary artery occlusion. Thus diminished electrical coupling per se plays a critical role in arrhythmogenesis induced by acute ischemia.


Subject(s)
Arrhythmias, Cardiac/etiology , Connexin 43/metabolism , Myocardial Ischemia/complications , Animals , Arrhythmias, Cardiac/physiopathology , Blood Pressure , Connexin 43/deficiency , Connexin 43/genetics , Electrophysiology , Heart Ventricles/physiopathology , In Vitro Techniques , Mice , Myocardial Ischemia/physiopathology , Perfusion , Tachycardia/physiopathology
18.
J Cardiovasc Electrophysiol ; 10(10): 1390-401, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10515564

ABSTRACT

INTRODUCTION: Dynamic interplay between active and passive electrical properties of cardiac myocytes is based on interrelationships between various channels responsible for depolarizing and repolarizing ionic currents and intercellular conductances. Mice with targeted disruption of the connexin43 (Cx43) gene have hearts completely devoid of Cx43, the principal gap junctional protein expressed in mammalian hearts. METHODS AND RESULTS: To determine whether cardiac myocytes that develop in an abnormal environment of reduced intercellular coupling have altered active membrane properties, we studied whole cell action potentials, Na+ channel currents, and Na+ channel expression and distribution via immunoblotting and confocal immunofluorescence in neonatal ventricular myocytes isolated from Cx43 wild-type, heterozygous, and homozygous null hearts. Action potential morphology, peak Na+ current, activation and inactivation kinetics, and Na+ channel protein expression and distribution were not different among myocytes isolated from wild-type, heterozygous, or null hearts. Active membrane properties and Na+ channel activity were completely normal in Cx43-deficient myocytes isolated from hearts that have been shown to exhibit markedly reduced Cx43 expression, gap junction number, and epicardial conduction delay. CONCLUSION: Despite a genetic inability to produce Cx43 and a developmental history that culminates in marked gross cardiac morphologic abnormalities, premature death, and myocardial inexcitability ex vivo, cardiac Na+ channel distribution and function appear to be normal in Cx43 null hearts. Although intimate structural and functional interrelationships have been described between ion channels and gap junction channels, expression and function of Na+ channels is not affected by the absence of Cx43.


Subject(s)
Connexin 43/physiology , Heart/physiology , Sodium Channels/physiology , Animals , Cells, Cultured , Heterozygote , Homozygote , Mice , Mice, Knockout , Microscopy, Confocal , Myocardium/cytology , Sodium Channels/ultrastructure
20.
Can J Physiol Pharmacol ; 77(7): 510-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10535711

ABSTRACT

This study tests the hypothesis that moderate myocardial dysfunction is associated with altered myocardial anisotropic properties and structurally altered ventricular fibrillation (VF). Mongrel dogs were randomized to either a control group or a group that was rapidly paced at 250 beats/min until the left ventricular ejection fraction was < or = 40%. Changes in anisotropic properties and the electrical characteristics of VF associated with the development of moderate myocardial dysfunction were assessed by microminiature epicardial mapping studies. In vivo conduction, refractory periods, and repolarization times were prolonged in both longitudinal and transverse directions in myopathic animals versus controls. VF was different in myopathic versus control animals. There were significantly more conducted deflections during VF in normal hearts compared with myopathic hearts. Propagated deflection-to-deflection intervals during VF were significantly longer in myopathic hearts compared with controls (125.5 +/- 49.06 versus 103.4 +/- 32.9 ms, p = 0.009). There were no abnormalities in cell size, cell shape, or the number of intercellular gap junctions and there was no detectable change in the expression of the gap junction proteins Cx43 and Cx45. Moderate myocardial dysfunction is associated with significant electrophysiological abnormalities in the absence of changes in myocardial cell morphology or intercellular connections, suggesting a functional abnormality in cell-to-cell communication.


Subject(s)
Cardiomyopathies/physiopathology , Heart Conduction System/physiopathology , Ventricular Fibrillation/physiopathology , Animals , Connexin 43/analysis , Dogs , Electric Countershock , Electrocardiography , Gap Junctions/ultrastructure , Myocardium/ultrastructure , Ventricular Function, Left
SELECTION OF CITATIONS
SEARCH DETAIL
...