Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
J Membr Biol ; 187(1): 15-25, 2002 May 01.
Article in English | MEDLINE | ID: mdl-12029374

ABSTRACT

Kcv is a K+-selective channel encoded by the Paramecium bursaria Chlorella virus 1 (PBVC-1). Expression of this protein, so far the smallest known functional K+ channel, in Xenopus oocytes reveals an instantaneous and a time-dependent component during voltage-clamp steps. These two components have an identical sensitivity to the inhibitor amantadine, implying that they reflect distinct kinetic features of the same channel. About 70% of the channels are always open; at hyperpolarizing voltages the time-dependent channels (30%) open in a voltage-dependent manner reaching half-maximal activation at about ?70 mV. At both extreme positive and negative voltages the open-channel conductance decreases in a voltage-dependent manner. To examine the mechanism underlying the voltage-dependence of Kcv we neutralized the two charged amino acids in the lipophilic N-terminus. However, this double mutation had no effect on the voltage-dependence of the channel, ruling against the possibility that these charged amino acids represent a membrane-embedded voltage sensor. We have considered whether a block by external divalent cations is involved in the voltage-dependence of the channel. The Kcv current was increased about 4-fold on reduction of external Ca2+ concentration by a factor of ten. This pronounced increase in current was observed on lowering Ca2+ but not Mg2+ and was voltage-independent. These data indicate a Ca2+-selective, but voltage-independent mechanism for regulation of channel conductance.


Subject(s)
Cell Membrane/physiology , Membrane Potentials/physiology , Models, Biological , Potassium Channels, Voltage-Gated/physiology , Potassium Channels/physiology , Viral Proteins , Animals , Clone Cells , Electric Conductivity , Gene Expression Regulation/physiology , Oocytes/physiology , Sensitivity and Specificity , Structure-Activity Relationship , Xenopus laevis/physiology
6.
News Physiol Sci ; 17: 32-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11821534

ABSTRACT

The "funny" (pacemaker) current has unusual characteristics, including activation on hyperpolarization, permeability to K(+) and Na(+), modulation by internal cAMP, and a tiny, single-channel conductance. In cardiac cells and neurons, pacemaker channels control repetitive activity and excitability. The recent cloning of HCN subunits provides new insight into the molecular basis for the funny channel properties.


Subject(s)
Ion Channels/physiology , Nerve Tissue Proteins , Animals , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating/physiology , Ion Channels/chemistry , Kinetics , Nucleotides, Cyclic/physiology , Potassium Channels , Protein Processing, Post-Translational
7.
Am J Physiol Heart Circ Physiol ; 281(3): H1252-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11514294

ABSTRACT

Although the neonatal sinus node beats at a faster rate than the adult, when a sodium current (I(Na)) present in the newborn is blocked, the spontaneous rate is slower in neonatal myocytes than in adult myocytes. This suggests a possible functional substitution of I(Na) by another current during development. We used ruptured [T-type calcium current (I(Ca,T))] and perforated [L-type calcium current (I(Ca,L))] patch clamps to study developmental changes in calcium currents in sinus node cells from adult and newborn rabbits. I(Ca,T) density did not differ with age, and no significant differences were found in the voltage dependence of activation or inactivation. I(Ca,L) density was lower in the adult than newborn (12.1 +/- 1.4 vs. 17.6 +/- 2.5 pA/pF, P = 0.049). However, activation and inactivation midpoints were shifted in opposite directions, reducing the potential contribution during late diastolic depolarization in the newborn (activation midpoints -17.3 +/- 0.8 and -22.3 +/- 1.4 mV in the newborn and adult, respectively, P = 0.001; inactivation midpoints -33.4 +/- 1.4 and -28.3 +/- 1.7 mV for the newborn and adult, respectively, P = 0.038). Recovery of I(Ca,L) from inactivation was also slower in the newborn. The results suggest that a smaller but more negatively activating and rapidly recovering I(Ca,L) in the adult sinus node may contribute to the enhanced impulse initiation at this age in the absence of I(Na).


Subject(s)
Aging/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Sinoatrial Node/metabolism , Animals , Animals, Newborn , Cells, Cultured , Electric Stimulation , Female , Membrane Potentials/physiology , Patch-Clamp Techniques , Rabbits , Sinoatrial Node/growth & development
8.
Pflugers Arch ; 442(2): 192-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11417213

ABSTRACT

We have reported previously that the sinoatrial node (SAN) in the newborn rabbit expresses a Na+ current (INa) with properties similar to the neuronal type-I isoform and that this current contributes to the net inward current flowing during diastolic depolarization. To characterize this current further we conducted cell-attached single-channel experiments in isolated newborn SAN myocytes. The Na+ channel was sensitive to divalent cation block and had a single-channel conductance of 25.6 pS in the absence of divalent cations. Kinetic compatibility between single-channel and previous whole-cell data was confirmed by measuring the time constant of current decay. At pacemaker potentials, time constants were of the order of tens of milliseconds. Additional experiments indicated that this slow inactivation arises because the Na+ channels expressed in the neonatal SAN tend to re-open frequently at potentials in the pacemaker range. We suggest that this is the mechanism by which a small tetrodotoxin (TTX)-sensitive current contributes to the total inward current flowing during slow diastolic depolarization in neonatal (but not adult) pacemaker myocytes.


Subject(s)
Animals, Newborn/physiology , Rabbits/physiology , Sinoatrial Node/metabolism , Sodium Channels/physiology , Animals , Cations, Divalent/pharmacology , Electric Conductivity , Kinetics , Patch-Clamp Techniques , Sinoatrial Node/cytology , Sodium Channel Blockers , Time Factors
9.
J Biol Chem ; 276(32): 29930-4, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11397812

ABSTRACT

The hyperpolarization-activated cyclic nucleotide-gated (HCN) family of "pacemaker" channels includes 4 isoforms, the kinetics and cAMP-induced modulation of which differ quantitatively. Because HCN isoforms are highly homologous in the central region, but diverge more substantially in the N and C termini, we asked whether these latter regions could contribute to the determination of channel properties. To this aim, we analyzed activation/deactivation kinetics and the response to cAMP of heterologously expressed isoforms mHCN1 and rbHCN4 and verified that mHCN1 has much faster kinetics and lower cAMP sensitivity than rbHCN4. We then constructed rbHCN4 chimeras by replacing either the N or the C terminus, or both, with the analogous domains from mHCN1. We found that: 1) replacement of the N terminus (chimera N1-4) did not substantially modify either the kinetics or cAMP dependence of wild-type channels; 2) replacement of the C terminus, on the contrary, resulted in a chimeric channel (4-C1), the kinetics of which were strongly accelerated compared with rbHCN4, and that was fully insensitive to cAMP; 3) replacement of both N and C termini led to the same results as replacement of the C terminus alone. These results indicate that the C terminus of rbHCN4 contributes to the regulation of voltage- and cAMP-dependent channel gating, possibly through interaction with other intracellular regions not belonging to the N terminus.


Subject(s)
Cyclic AMP/metabolism , Ion Channels/metabolism , Ion Channels/physiology , Muscle Proteins , Nerve Tissue Proteins , Animals , Cell Line , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Green Fluorescent Proteins , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/chemistry , Kinetics , Luminescent Proteins/metabolism , Potassium Channels , Protein Isoforms , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transfection
10.
J Biol Chem ; 276(31): 29233-41, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11328811

ABSTRACT

The pacemaker current I(f) of the sinoatrial node (SAN) is a major determinant of cardiac diastolic depolarization and plays a key role in controlling heart rate and its modulation by neurotransmitters. Substantial expression of two different mRNAs (HCN4, HCN1) of the family of pacemaker channels (HCN) is found in rabbit SAN, suggesting that the native channels may be formed by different isoforms. Here we report the cloning and heterologous expression of HCN1 from rabbit SAN and its specific localization in pacemaker myocytes. rbHCN1 is an 822-amino acid protein that, in human embryonic kidney 293 cells, displayed electrophysiological properties similar to those of I(f), suggesting that HCN1 can form a pacemaker channel. The presence of HCN1 in the SAN myocytes but not in nearby heart regions, and the electrophysiological properties of the channels formed by it, suggest that HCN1 plays a central and specific role in the formation of SAN pacemaker currents.


Subject(s)
Brain/physiology , Heart/physiology , Ion Channels/physiology , Muscle Proteins , Sinoatrial Node/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cyclic Nucleotide-Gated Cation Channels , Embryo, Mammalian , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/chemistry , Ion Channels/genetics , Kidney , Kinetics , Membrane Potentials/physiology , Molecular Sequence Data , Muscle, Skeletal/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Organ Specificity , Potassium Channels , RNA, Messenger/analysis , RNA, Messenger/genetics , Rabbits , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Transcription, Genetic , Transfection
11.
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
12.
13.
Am J Physiol Heart Circ Physiol ; 279(5): H2303-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11045966

ABSTRACT

Isolated newborn, but not adult, rabbit sinoatrial node (SAN) cells exhibit spontaneous activity that (unlike adult) are highly sensitive to the Na(+) current (I(Na)) blocker TTX. To investigate this TTX action on automaticity, cells were voltage clamped with ramp depolarizations mimicking the pacemaker phase of spontaneous cells (-60 to -20 mV, 35 mV/s). Ramps elicited a TTX-sensitive current in newborn (peak density 0.89 +/- 0.14 pA/pF, n = 24) but not adult (n = 5) cells. When depolarizing ramps were preceded by steplike depolarizations to mimic action potentials, ramp current decreased 54.6 +/- 8.0% (n = 3) but was not abolished. Additional experiments demonstrated that ramp current amplitude depended on the slope of the ramp and that TTX did not alter steady-state holding current at pacemaker potentials. This excluded a steady-state Na(+) window component and suggested a kinetic basis, which was investigated by measuring TTX-sensitive I(Na) during long step depolarizations. I(Na) exhibited a slow but complete inactivation time course at pacemaker voltages (tau = 33.9 +/- 3.9 ms at -50 mV), consistent with the rate-dependent ramp data. The data indicate that owing to slow inactivation of I(Na) at diastolic potentials, a small TTX-sensitive current flows during the diastolic depolarization in neonatal pacemaker myocytes.


Subject(s)
Diastole/physiology , Myocardium/metabolism , Sinoatrial Node/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Cells, Cultured , Ion Transport/drug effects , Myocardium/cytology , Patch-Clamp Techniques , Rabbits , Sodium Channel Blockers , Tetrodotoxin/pharmacology
14.
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
16.
Pflugers Arch ; 439(5): 618-26, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10764222

ABSTRACT

Human cDNA coding for the hyperpolarization-activated "pacemaker" channel HCN2 was expressed in Phoenix cells and yielded an inward current (IhHCN2) activated on hyperpolarization. The average IhHCN2 was half-activated at -83.1 mV and its kinetics could be described by second-order Hodgkin-Huxley gating. The time constant curve was bell-shaped and peaked at -82.2 mV. With 115 mM external Na+ and 30 mM external K+, IhHCN2 reversed at -17.1 mV, and had a mean conductance of 5.6 nS. Reducing the external K+ or Na+ concentration led to a concentration-dependent reduction of the IhHCN2 conductance and to a hyperpolarizing shift of reversal potential. External Cs+ ions (5 mM) blocked IhHCN2 in a voltage-dependent way according to a Woodhull-type block model, at an electrical distance of 0.66 from the external membrane surface, and with a dissociation constant of 15 mM at 0 mV. Increasing cytoplasmic cAMP using forskolin increased IhHCN2 by shifting the current activation curve to more positive voltages (11.7 mV). Exposure of the intracellular side of inside-out macro-patches to cAMP led to a depolarizing shift of the channel open probability curve (15.2 mV with 10 microM cAMP). These results indicate that although hHCN2 channels share several properties with native cardiac f-channels, differences also exist in permeability and block properties, suggesting that native channels may not be composed simply of homomeric constructs.


Subject(s)
Biological Clocks/physiology , Ion Channel Gating/physiology , Ion Channels/genetics , Ion Channels/metabolism , Muscle Proteins , Cell Line , Cesium/pharmacology , Cloning, Molecular , Colforsin/pharmacology , Cyclic AMP/pharmacology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/physiology , Myocardium/chemistry , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels , Sodium/pharmacology
17.
Biophys J ; 78(4): 1862-71, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733966

ABSTRACT

Effects of threonine substitution by glutamine at position 256 in the pore of the KAT1 channel have been investigated by voltage-clamp, using heterologous gene expression in Xenopus oocytes. The major discrepancy in T256Q from the wild-type channel (wt) was cation specific. While K(+) currents were reduced in a largely scalar fashion, the NH(4)(+) current exhibited slow, voltage-dependent inhibition during hyperpolarization. The same effects could be induced in wt, or intensified in T256Q, by addition of the impermeant cation methylammonium (MA(+)) to the bath. This stresses that both the mutation and MA(+) affect a mechanism already present in the wt. Assuming that current inhibition could be described as entry of the channel into an inactive state, we modeled in both wt and in T256Q the relaxation kinetics of the clamp currents by a C-O-I gating scheme, where C (closed) and I (inactivated) are nonconductive states, and O is an open state allowing K(+) and NH(4)(+) passage. The key reaction is the transition I-O. This cation-sensitive transition step ensures release of the channel from the inactive state and is approximately 30 times smaller in T256Q compared to wt. It can be inhibited by external MA(+) and is stimulated strongly by K(+) and weakly by NH(4)(+). This sensitivity of gating to external cations may prevent K(+) leakage from cation-starved cells.


Subject(s)
Plant Proteins/chemistry , Plant Proteins/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/chemistry , Potassium Channels/metabolism , Amino Acid Substitution , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins , Biophysical Phenomena , Biophysics , Cations , Electric Conductivity , Female , In Vitro Techniques , Ion Channel Gating , Kinetics , Mutagenesis, Site-Directed , Oocytes/metabolism , Plant Proteins/genetics , Potassium Channels/genetics , Protein Structure, Tertiary , Quaternary Ammonium Compounds/pharmacology , Xenopus
18.
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
19.
J Physiol ; 520 Pt 3: 737-44, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10545140

ABSTRACT

1. The hyperpolarization-activated If current was recorded in inside-out macropatches from sino-atrial (SA) node myocytes during exposure of their intracellular side to pronase, in an attempt to verify if cytoplasmic f-channel domains are involved in both voltage- and cAMP-dependent gating. 2. Superfusion with pronase caused a quick, dramatic acceleration of channel opening upon hyperpolarization and slowing, rapidly progressing into full blockade, of channel closing upon depolarization; these changes persisted after wash off of pronase and were irreversible, indicating proteolytic cleavage of channel regions which contribute to gating. 3. If recorded from patches normally responding to cAMP became totally insensitive to cAMP following pronase treatment, indicating partial or total removal of channel regions involved in the cAMP-dependent activation. 4. The fully activated I-V relationship was not modified by pronase, indicating that internal proteolysis did not affect the f-channel conductance. 5. The changes in If kinetics induced by pronase were due to a large depolarizing shift of the f-channel open probability curve (56.5 +/- 1.1 mV, n = 7). 6. These results are consistent with the hypothesis that cytoplasmic f-channel regions are implicated in dual voltage- and cAMP-dependent gating; also, since pronase does not abolish hyperpolarization-activated opening, an intrinsic voltage-dependent gating mechanism must exist which is inaccessible to proteolytic cleavage. A model scheme able to account for these data thus includes an intrinsic gating mechanism operating at depolarized voltages, and a blocking mechanism coupled to cAMP binding to the channel.


Subject(s)
Ion Channels/metabolism , Pronase/physiology , Sinoatrial Node/metabolism , Animals , Cyclic AMP/pharmacology , Electric Conductivity , Electrophysiology , Ion Channel Gating/physiology , Ion Channels/drug effects , Ion Channels/physiology , Kinetics , Pronase/pharmacology , Rabbits , Sinoatrial Node/cytology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...