Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Cell Death Differ ; 15(9): 1417-28, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18497758

ABSTRACT

Different cardiac stem/progenitor cells have been recently identified in the post-natal heart. We describe here the identification, clonal expansion and characterization of self-renewing progenitors that differ from those previously described for high spontaneous cardiac differentiation. Unique coexpression of endothelial and pericyte markers identify these cells as cardiac mesoangioblasts and allow prospective isolation and clonal expansion from the juvenile mouse ventricle. Cardiac mesoangioblasts express many cardiac transcription factors and spontaneously differentiate into beating cardiomyocytes that assemble mature sarcomeres and express typical cardiac ion channels. Cells similarly isolated from the atrium do not spontaneously differentiate. When injected into the ventricle after coronary artery ligation, cardiac mesoangioblasts efficiently generate new myocardium in the peripheral area of the necrotic zone, as they do when grafted in the embryonic chick heart. These data identify cardiac mesoangioblasts as committed progenitors, downstream of earlier stem/progenitor cells and suitable for the cell therapy of a subset of juvenile cardiac diseases.


Subject(s)
Heart Ventricles/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Endothelium, Vascular/cytology , Heart Ventricles/growth & development , Humans , Mice , Myocardium/cytology , Patch-Clamp Techniques , Rats , Stem Cells/metabolism , Stem Cells/physiology
2.
Science ; 243(4891): 669-71, 1989 Feb 03.
Article in English | MEDLINE | ID: mdl-2916119

ABSTRACT

Slowing of cardiac pacemaking induced by cholinergic input is thought to arise from the opening of potassium channels caused by muscarinic receptor stimulation. In mammalian sinoatrial node cells, however, muscarinic stimulation also inhibits the hyperpolarization-activated current (If), which is involved in the generation of pacemaker activity and its acceleration by catecholamines. Acetylcholine at nanomolar concentrations inhibits If and slows spontaneous rate, whereas 20 times higher concentrations are required to activate the acetylcholine-dependent potassium current (IK,ACh). Thus, modulation of If, rather than IK,ACh, is the mechanism underlying the muscarinic control of cardiac pacing at low (nanomolar) acetylcholine concentrations.


Subject(s)
Acetylcholine/pharmacology , Heart Rate/drug effects , Receptors, Muscarinic/physiology , Sinoatrial Node/physiology , Acetylcholine/administration & dosage , Action Potentials/drug effects , Animals , Barium/pharmacology , Dose-Response Relationship, Drug , Electric Conductivity , Potassium Channels/physiology , Rabbits , Receptors, Muscarinic/drug effects
3.
Science ; 287(5458): 1641-4, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10698737

ABSTRACT

The large chlorella virus PBCV-1, which contains double-stranded DNA (dsDNA), encodes a 94-codon open reading frame (ORF) that contains a motif resembling the signature sequence of the pore domain of potassium channel proteins. Phylogenetic analyses of the encoded protein, Kcv, indicate a previously unidentified type of potassium channel. The messenger RNA encoded by the ORF leads to functional expression of a potassium-selective conductance in Xenopus laevis oocytes. The channel blockers amantadine and barium, but not cesium, inhibit this conductance, in addition to virus plaque formation. Thus, PBCV-1 encodes the first known viral protein that functions as a potassium-selective channel and is essential in the virus life cycle.


Subject(s)
Phycodnaviridae/genetics , Phycodnaviridae/physiology , Potassium Channels/chemistry , Potassium Channels/physiology , Viral Proteins , Amantadine/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Animals , Barium/pharmacology , Cesium/pharmacology , Chlorella/virology , Isoelectric Point , Molecular Sequence Data , Molecular Weight , Oocytes , Patch-Clamp Techniques , Phycodnaviridae/chemistry , Phycodnaviridae/drug effects , Potassium/metabolism , Potassium Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sodium/metabolism , Viral Plaque Assay , Virus Replication/drug effects , Xenopus laevis
4.
Prog Biophys Mol Biol ; 120(1-3): 50-66, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26718599

ABSTRACT

We used the Dynamic Clamp technique for i) comparative validation of conflicting computational models of the hyperpolarization-activated funny current, If, and ii) quantification of the role of If in mediating autonomic modulation of heart rate. Experimental protocols based on the injection of a real-time recalculated synthetic If current in sinoatrial rabbit cells were developed. Preliminary results of experiments mimicking the autonomic modulation of If demonstrated the need for a customization procedure to compensate for cellular heterogeneity. For this reason, we used a cell-specific approach, scaling the maximal conductance of the injected current based on the cell's spontaneous firing rate. The pacemaking rate, which was significantly reduced after application of Ivabradine, was restored by the injection of synthetic current based on the Severi-DiFrancesco formulation, while the injection of synthetic current based on the Maltsev-Lakatta formulation did not produce any significant variation. A positive virtual shift of the If activation curve, mimicking the Isoprenaline effects, led to a significant increase in pacemaking rate (+17.3 ± 6.7%, p < 0.01), although of lower magnitude than that induced by real Isoprenaline (+45.0 ± 26.1%). Similarly, a negative virtual shift of the activation curve significantly lowered the pacemaking rate (-11.8 ± 1.9%, p < 0.001), as did the application of real Acetylcholine (-20.5 ± 5.1%). The Dynamic Clamp approach, applied to the If study in cardiomyocytes for the first time and rate-adapted to manage intercellular variability, indicated that: i) the quantitative description of the If current in the Severi-DiFrancesco model accurately reproduces the effects of the real current on rabbit sinoatrial cell pacemaking rate and ii) a significant portion (50-60%) of the physiological autonomic rate modulation is due to the shift of the If activation curve.


Subject(s)
Cytological Techniques , Electrophysiological Phenomena , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Sinoatrial Node/physiology , Acetylcholine/pharmacology , Action Potentials/drug effects , Animals , Benzazepines/pharmacology , Electrophysiological Phenomena/drug effects , Heart Rate/drug effects , Ivabradine , Models, Cardiovascular , Rabbits , Single-Cell Analysis , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism
5.
Biochim Biophys Acta ; 1446(3): 419-25, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10524219

ABSTRACT

Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, underlying 'pacemaker' currents (I(f)/Ih), are involved in pacemaker activity of cardiac sinoatrial node myocytes and central neurons. Several cDNAs deriving from four different genes were recently identified which code for channels characterized by six transmembrane domains and a cyclic nucleotide binding domain. We report here the identification of the human HCN2 gene and show that its functional expression in a human kidney cell line generates a current with properties similar to the native pacemaker f-channel of the heart. The hHCN2 gene maps to the telomeric region of chromosome 19, band p13.3. This is the first identification of a genetic locus coding for an HCN channel.


Subject(s)
Heart Rate/genetics , Heart/physiology , Ion Channels/genetics , Muscle Proteins , Amino Acid Sequence , Base Sequence , Cell Line , Chromosomes, Human, Pair 19 , DNA, Complementary/chemistry , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Situ Hybridization, Fluorescence , Kidney/metabolism , Molecular Sequence Data , Potassium Channels , Sequence Alignment
6.
J Gen Physiol ; 117(6): 519-32, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11382803

ABSTRACT

Hyperpolarization-activated (pacemaker) channels are dually gated by negative voltage and intracellular cAMP. Kinetics of native cardiac f-channels are not compatible with HH gating, and require closed/open multistate models. We verified that members of the HCN channel family (mHCN1, hHCN2, hHCN4) also have properties not complying with HH gating, such as sigmoidal activation and deactivation, activation deviating from fixed power of an exponential, removal of activation "delay" by preconditioning hyperpolarization. Previous work on native channels has indicated that the shifting action of cAMP on the open probability (Po) curve can be accounted for by an allosteric model, whereby cAMP binds more favorably to open than closed channels. We therefore asked whether not only cAMP-dependent, but also voltage-dependent gating of hyperpolarization-activated channels could be explained by an allosteric model. We hypothesized that HCN channels are tetramers and that each subunit comprises a voltage sensor moving between "reluctant" and "willing" states, whereas voltage sensors are independently gated by voltage, channel closed/open transitions occur allosterically. These hypotheses led to a multistate scheme comprising five open and five closed channel states. We estimated model rate constants by fitting first activation delay curves and single exponential time constant curves, and then individual activation/deactivation traces. By simply using different sets of rate constants, the model accounts for qualitative and quantitative aspects of voltage gating of all three HCN isoforms investigated, and allows an interpretation of the different kinetic properties of different isoforms. For example, faster kinetics of HCN1 relative to HCN2/HCN4 are attributable to higher HCN1 voltage sensors' rates and looser voltage-independent interactions between subunits in closed/open transitions. It also accounts for experimental evidence that reduction of sensors' positive charge leads to negative voltage shifts of Po curve, with little change of curve slope. HCN voltage gating thus involves two processes: voltage sensor gating and allosteric opening/closing.


Subject(s)
Cyclic AMP/physiology , Ion Channel Gating/physiology , Ion Channels/physiology , Models, Biological , Nerve Tissue Proteins , Animals , Cloning, Molecular , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Kinetics , Mice , Potassium Channels
7.
Trends Cardiovasc Med ; 1(6): 250-5, 1991 Sep.
Article in English | MEDLINE | ID: mdl-21239304

ABSTRACT

Several currents contribute to the electrical activity of mammalian pacemaker cells. Of these, the hyperpolarization-activated current, i(f), is involved in the generation of the diastolic depolarization phase, and therefore has a main role in controlling the most peculiar feature of these cells: their ability to beat spontaneously and to drive the heartbeat. More than this, i(f) represents the key mechanism by which sympathetic and parasympathetic stimuli regulate, via the diastolic depolarization phase, the pacing frequency of sinoatrial node cells and thus the heart rate. This is achieved through regulation of adenylyl-cyclase activity and of intracellular cAMP, which is the second messenger in i(f) modulation. A fine regulation of i(f) is thus the basis by which epinephrine and acetylcholine exert their fine control on cardiac rhythm.

8.
Proc Biol Sci ; 250(1329): 199-207, 1992 Dec 22.
Article in English | MEDLINE | ID: mdl-1283636

ABSTRACT

Experiments and computations were done to clarify the role of the various inward currents in generating and modulating pacemaker frequency. Ionic currents in rabbit single isolated sino-atrial (SA) node cells were measured using the nystatin-permeabilized patch-clamp technique. The results were used to refine the Noble-DiFrancesco-Denyer model of spontaneous pacemaker activity of the SA node. This model was then used to show that the pacemaker frequency is relatively insensitive to the magnitude of the sodium-dependent inward background current ib, Na. This is because reducing ib, Na hyperpolarizes the cell and so activates more hyperpolarizing-activated current, i(f), whereas the converse occurs when ib, Na is increased. The result is that i(f) and ib, Na replace one another and so stabilize nodal pacemaker frequency.


Subject(s)
Ion Channels/physiology , Models, Cardiovascular , Sinoatrial Node/physiology , Animals , In Vitro Techniques , Mathematics , Membrane Potentials , Rabbits , Sodium Channels/physiology
9.
Br J Pharmacol ; 130(6): 1315-20, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10903971

ABSTRACT

1. The effect of the antiarrhythmic drug dronedarone on the Acetylcholine-activated K(+) current (I(K(ACh))) was investigated in single cells isolated from sinoatrial node (SAN) tissue of rabbit hearts. 2. Externally perfused dronedarone (0.001 - 1 microM) caused a potent, voltage independent block of I(K(ACh)). Fitting of the dose response curve of I(K(ACh)) block yielded an IC(50) value of 63 nM, a value over one order of magnitude lower than those reported for dronedarone block of other cardiac currents. 3. I(K(ACh)) block was not due to an inhibitory action of dronedarone on the muscarinic M2 receptor activation, since the drug was effective on I(K(ACh)) constitutively activated by intracellular perfusion with GTP-gammaS. 4. External cell perfusion with dronedarone inhibited the activity of I(K(ACh)) channels recorded from cell-attached patches by reducing the channel open probability (from 0.56 to 0.11) without modification of the single-channel conductance. 5. These data suggest that dronedarone blocks I(K(ACh)) channels either by disrupting the G-protein-mediated activation or by a direct inhibitory interaction with the channel protein.


Subject(s)
Acetylcholine/pharmacology , Amiodarone/analogs & derivatives , Membrane Potentials/drug effects , Sinoatrial Node/drug effects , Amiodarone/pharmacology , Animals , Dose-Response Relationship, Drug , Dronedarone , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/physiology , Rabbits , Receptor, Muscarinic M2 , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Sinoatrial Node/cytology , Sinoatrial Node/physiology
10.
Neuroreport ; 5(14): 1813-6, 1994 Sep 08.
Article in English | MEDLINE | ID: mdl-7827339

ABSTRACT

Long-term depression of field excitatory postsynaptic potentials (EPSP) in the CA1 region of hippocampal slices was evoked by delivering a 15 min train of pulses at 1 Hz to the Schaffer-commissural-CA1 pathway, and prevented by adding an N-methyl-D-aspartate (NMDA) receptor antagonist (AP-5, 50 microM) to the perfusing medium. Superfusion of the slices with Cs (2 mM) during the 1 Hz stimulation period could both inhibit the maintenance phase of the depression itself and elicit spontaneous rhythmic activity. Cs had no effect on the postsynaptic response to the GABA-B agonist, baclofen. As a major effect of Cs is a block of the hyperpolarization-activated current (Ih), these results suggest the possible involvement of Ih in the maintenance of long-term depression.


Subject(s)
Cesium/pharmacology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Depression, Chemical , Electric Stimulation , Hippocampus/cytology , In Vitro Techniques , Male , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
11.
Acta Cardiol ; 50(6): 413-27, 1995.
Article in English | MEDLINE | ID: mdl-8932564

ABSTRACT

After more than 15 years since the "new" interpretation of the Purkinje fibre's pacemaker current was proposed, much progress has been made in the understanding of the basic functional principles of cardiac pacemaking. We now know that, in both the SA node and Purkinje fibres, the diastolic depolarization is generated by the interplay of several ionic components, the key process being represented by the turning-on of the hyperpolarization-activated i(f) current towards the end of the action potential repolarization phase. The properties of i(f) are well suited not only to generate, but also to mediate the control of cardiac rate by autonomic transmitters. This control is exerted through modulation of adenylate-cyclase and of cAMP, and allows a fine and rapid adjustment of heart rate to the changing needs of our normal day-life. Still, several problems remain to be clarified : for example, it is not clear how the degree of involvement of i(f) and other components changes in different areas of the nodal region, and whether this process is under control of the autonomic nervous system; more importantly, it is still unknown if the pacemaking mechanisms are similar in the newborn and in the adult, or if developmental changes in the way pacemaker activity is generated and modulated exist.


Subject(s)
Electrocardiography , Heart Conduction System/physiology , Pacemaker, Artificial , Autonomic Nervous System/physiology , Computer Simulation , Heart/innervation , Humans , Models, Cardiovascular , Potassium Channels/physiology , Purkinje Fibers/physiology
13.
Prog Biophys Mol Biol ; 107(1): 4-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745496

ABSTRACT

Cardiac experimental electrophysiology is in need of a well-defined Minimum Information Standard for recording, annotating, and reporting experimental data. As a step towards establishing this, we present a draft standard, called Minimum Information about a Cardiac Electrophysiology Experiment (MICEE). The ultimate goal is to develop a useful tool for cardiac electrophysiologists which facilitates and improves dissemination of the minimum information necessary for reproduction of cardiac electrophysiology research, allowing for easier comparison and utilisation of findings by others. It is hoped that this will enhance the integration of individual results into experimental, computational, and conceptual models. In its present form, this draft is intended for assessment and development by the research community. We invite the reader to join this effort, and, if deemed productive, implement the Minimum Information about a Cardiac Electrophysiology Experiment standard in their own work.


Subject(s)
Electrophysiological Phenomena , Heart/physiology , Information Dissemination/methods , Models, Biological , Research Design/standards , Animals , Humans , Reference Standards , Reproducibility of Results
18.
19.
J Physiol ; 572(Pt 2): 335-46, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16484306

ABSTRACT

Ivabradine is a 'heart rate-reducing' agent able to slow heart rate, without complicating side-effects. Its action results from a selective and specific block of pacemaker f-channels of the cardiac sinoatrial node (SAN). Investigation has shown that block by ivabradine requires open f-channels, is use dependent, and is affected by the direction of current flow. The constitutive elements of native pacemaker channels are the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, of which four isoforms (HCN1-4) are known; in rabbit SAN tissue HCN4 is expressed strongly, and HCN1 weakly. In this study we have investigated the blocking action of ivabradine on mouse (m) HCN1 and human (h) HCN4 channels heterologously expressed in HEK 293 cells. Ivabradine blocked both channels in a dose-dependent way with half-block concentrations of 0.94 microm for mHCN1 and 2.0 microm for hHCN4. Properties of block changed substantially for the two channels. Block of hHCN4 required open channels, was strengthened by depolarization and was relieved by hyperpolarization. Block of mHCN1 did not occur, nor was it relieved, when channels were in the open state during hyperpolarization; block required channels to be either closed, or in a transitional state between open and closed configurations. The dependence of block upon current flow was limited for hHCN4, and not significant for mHCN1 channels. In summary our results indicate that ivabradine is an 'open-channel' blocker of hHCN4, and a 'closed-channel' blocker of mHCN1 channels. The mode of action of ivabradine on the two channels is discussed by implementing a simplified version of a previously developed model of f-channel kinetics.


Subject(s)
Benzazepines/pharmacology , Biological Clocks/drug effects , Ion Channels/drug effects , Muscle Proteins/drug effects , Nerve Tissue Proteins/drug effects , Action Potentials/physiology , Cell Line , Cyclic Nucleotide-Gated Cation Channels , Dose-Response Relationship, Drug , Electrophysiology , Heart Rate/physiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Ivabradine , Kidney/cytology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Patch-Clamp Techniques , Potassium Channels , Protein Isoforms , Sinoatrial Node/drug effects , Sinoatrial Node/physiology
20.
J Physiol ; 434: 23-40, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2023118

ABSTRACT

1. The contribution to the diastolic depolarization of the hyperpolarization-activated current, if, relative to other components was investigated in isolated rabbit sino-atrial (SA) node myocytes. 2. During the diastolic phase the membrane potential depolarized by 0.1096 +/- 0.014 V/s, which requires only about 3 pA of inward current in a cell with an average capacity of 30 pF. The problem of which ionic component is responsible for initiating the diastolic depolarization was investigated by analysing the composition and the properties of the net inward current in the diastolic range of voltages. 3. The measured instantaneous 'background' current activated during voltage clamp steps from a holding potential of -35 mV was outward positive to approximately -61 mV, and had a region of negative slope conductance from -45 to -35 mV. 4. The instantaneous component lost its rectifying behaviour in the presence of Ni2+ (100 microM) and nitrendipine (10 microM). These blockers of Ca(2+)-dependent currents modified the instantaneous I-V relation at voltages positive to -45 to -50 mV, thus implying that Ca2+ currents become important at less negative potentials than -50 mV, towards the very end of diastolic depolarization. 5. Possible errors introduced by voltage clamp analysis with the whole-cell method on the instantaneous current and on if measurement were evaluated. Leakage through the seal resistance caused the instantaneous I-V relation to be displaced in the inward direction at negative voltages. Correction for the seal leakage moved the reversal potential for the instantaneous current toward the negative direction from -61 to approximately -66 mV. Thus, no depolarization can be driven by the background current beyond -66 mV. 6. During voltage clamp analysis, lack of series-resistance compensation led to lack of intracellular voltage control, as was apparent using a second pipette on the same cell. This slowed activation of if and led to a 1.5- to 2-fold reduction of if size in the range -55 to -115 mV. Thus, uncorrected measurements of the instantaneous component and of if may concur to underestimate the role of if in pacemaking. 7. These results lead to the conclusion that in the SA node cells analysed, pacemaker activity is generated with the essential contribution of the hyperpolarization-activated current, if. Numerical computation of SA node cell activity using an extension of the DiFrancesco-Noble model shows that the if-activation hypothesis can account for the presence of spontaneous action potentials and their sensitivity to if changes.


Subject(s)
Action Potentials/physiology , Pacemaker, Artificial , Sinoatrial Node/physiology , Animals , Calcium/physiology , Cell Separation/methods , Diastole/physiology , Electric Stimulation , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nitrendipine/pharmacology , Rabbits , Sinoatrial Node/cytology
SELECTION OF CITATIONS
SEARCH DETAIL