Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Biotechnol Bioeng ; 118(1): 442-452, 2021 01.
Article in English | MEDLINE | ID: mdl-32990953

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as an exciting new tool for cardiac research and can serve as a preclinical platform for drug development and disease modeling studies. However, these aspirations are limited by current culture methods in which hPSC-CMs resemble fetal human cardiomyocytes in terms of structure and function. Herein we provide a novel in vitro platform that includes patterned extracellular matrix with physiological substrate stiffness and is amenable to both mechanical and electrical analysis. Micropatterned lanes promote the cellular and myofibril alignment of hPSC-CMs while the addition of micropatterned bridges enable formation of a functional cardiac syncytium that beats synchronously over a large two-dimensional area. We investigated the electrophysiological properties of the patterned cardiac constructs and showed they have anisotropic electrical impulse propagation, as occurs in the native myocardium, with speeds 2x faster in the primary direction of the pattern as compared to the transverse direction. Lastly, we interrogated the mechanical function of the pattern constructs and demonstrated the utility of this platform in recording the strength of cardiomyocyte contractions. This biomimetic platform with electrical and mechanical readout capabilities will enable the study of cardiac disease and the influence of pharmaceuticals and toxins on cardiomyocyte function. The platform also holds potential for high throughput evaluation of drug safety and efficacy, thus furthering our understanding of cardiovascular disease and increasing the translational use of hPSC-CMs.


Subject(s)
Electrophysiological Phenomena , Giant Cells/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Cell Line , Humans , Induced Pluripotent Stem Cells/metabolism
2.
Br J Pharmacol ; 149(5): 481-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16967046

ABSTRACT

BACKGROUND AND PURPOSE: Fluoxetine (Prozac) is a widely prescribed drug in adults and children, and it has an active metabolite, norfluoxetine, with a prolonged elimination time. Although uncommon, Prozac causes QT interval prolongation and arrhythmias; a patient who took an overdose of Prozac exhibited a prolonged QT interval (QTc 625 msec). We looked for possible mechanisms underlying this clinical finding by analysing the effects of fluoxetine and norfluoxetine on ion channels in vitro. EXPERIMENTAL APPROACH: We studied the effects of fluoxetine and norfluoxetine on the electrophysiology and cellular trafficking of hERG K+ and SCN5A Na+ channels heterologously expressed in HEK293 cells. KEY RESULTS: Voltage clamp analyses employing square pulse or ventricular action potential waveform protocols showed that fluoxetine and norfluoxetine caused direct, concentration-dependent, block of hERG current (IhERG). Biochemical studies showed that both compounds also caused concentration-dependent reductions in the trafficking of hERG channel protein into the cell surface membrane. Fluoxetine had no effect on SCN5A channel or HEK293 cell endogenous current. Mutations in the hERG channel drug binding domain reduced fluoxetine block of IhERG but did not alter fluoxetine's effect on hERG channel protein trafficking. CONCLUSIONS AND IMPLICATIONS: Our findings show that both fluoxetine and norfluoxetine at similar concentrations selectively reduce IhERG by two mechanisms, (1) direct channel block, and (2) indirectly by disrupting channel protein trafficking. These two effects are not mediated by a single drug binding site. Our findings add complexity to understanding the mechanisms that cause drug-induced long QT syndrome.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Fluoxetine/adverse effects , Long QT Syndrome/chemically induced , Adult , Antidepressive Agents, Second-Generation/adverse effects , Antidepressive Agents, Second-Generation/pharmacology , Blotting, Western , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/physiology , Cisapride/pharmacology , Dose-Response Relationship, Drug , Drug Overdose , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Female , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Humans , Long QT Syndrome/metabolism , Long QT Syndrome/physiopathology , Membrane Potentials/drug effects , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation/genetics , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Piperidines/pharmacology , Protein Transport/drug effects , Pyridines/pharmacology , Sodium Channels/genetics , Sodium Channels/metabolism , Time Factors , Transfection
3.
WMJ ; 105(3): 49-54, 2006 May.
Article in English | MEDLINE | ID: mdl-16749326

ABSTRACT

Transient left ventricular apical ballooning is a newly defined syndrome characterized by sudden onset of chest symptoms, electrocardiographic changes characteristic of myocardial ischemia, transient left ventricular dysfunction-particularly in the apical region, low-grade troponin elevation, and no significant coronary stenosis by angiogram. This syndrome is also referred to as Takotsubo cardiomyopathy, "Ampulla" cardiomyopathy, Human Stress cardiomyopathy, and Broken Heart Syndrome. Emergency physicians, family physicians, general internists, and cardiologists may all encounter this syndrome at the point of contact. The similarity to acute coronary syndrome requires all clinicians who may potentially care for these patients to familiarize themselves with this newly recognized disease. We provide a recent case and review the current literature surrounding this syndrome.


Subject(s)
Cardiomyopathies/diagnosis , Aged , Cardiomyopathies/blood , Coronary Angiography , Diagnosis, Differential , Echocardiography , Electrocardiography , Female , Humans , Myocardial Ischemia/diagnosis , Syndrome , Troponin I/blood , Ventricular Dysfunction, Left/diagnosis , Ventricular Premature Complexes/diagnosis
4.
JAMA ; 286(18): 2264-9, 2001 Nov 14.
Article in English | MEDLINE | ID: mdl-11710892

ABSTRACT

CONTEXT: Fatal arrhythmias from occult long QT syndrome may be responsible for some cases of sudden infant death syndrome (SIDS). Because patients who have long QT syndrome with sodium channel gene (SCN5A) defects have an increased frequency of cardiac events during sleep, and a recent case is reported of a sporadic SCN5A mutation in an infant with near SIDS, SCN5A has emerged as the leading candidate ion channel gene for SIDS. OBJECTIVE: To determine the prevalence and functional properties of SCN5A mutations in SIDS. DESIGN, SETTING, AND SUBJECTS: Postmortem molecular analysis of 93 cases of SIDS or undetermined infant death identified by the Medical Examiner's Office of the Arkansas State Crime Laboratory between September 1997 and August 1999. Genomic DNA was extracted from frozen myocardium and subjected to SCN5A mutational analyses. Missense mutations were incorporated into the human heart sodium channel alpha subunit by mutagenesis, transiently transfected into human embryonic kidney cells, and characterized electrophysiologically. MAIN OUTCOME MEASURES: Molecular and functional characterization of SCN5A defects. RESULTS: Two of the 93 cases of SIDS possessed SCN5A mutations: a 6-week-old white male with an A997S missense mutation in exon 17 and a 1-month old white male with an R1826H mutation in exon 28. These 2 distinct mutations occurred in highly conserved regions of the sodium channel and were absent in 400 control patients (800 alleles). Functionally, the A997S and R1826H mutant channels expressed a sodium current characterized by slower decay and a 2- to 3-fold increase in late sodium current. CONCLUSION: Approximately 2% of this prospective, population-based cohort of SIDS cases had an identifiable SCN5A channel defect, suggesting that mutations in cardiac ion channels may provide a lethal arrhythmogenic substrate in some infants at risk for SIDS.


Subject(s)
Sodium Channels/genetics , Sudden Infant Death/genetics , Autopsy , Cohort Studies , DNA/isolation & purification , DNA Mutational Analysis , Electrophysiology , Female , Gene Expression , Humans , Infant , Long QT Syndrome/genetics , Male , Mutation , Myocardium/pathology , NAV1.5 Voltage-Gated Sodium Channel , Phenotype , Polymorphism, Genetic , Risk Factors
5.
Proc Natl Acad Sci U S A ; 98(20): 11760-4, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562480

ABSTRACT

ATP-sensitive potassium channels (K(ATP)) are involved in a diverse array of physiologic functions including protection of tissue against ischemic insult, regulation of vascular tone, and modulation of insulin secretion. To improve our understanding of the role of K(ATP) in these processes, we used a gene-targeting strategy to generate mice with a disruption in the muscle-specific K(ATP) regulatory subunit, SUR2. Insertional mutagenesis of the Sur2 locus generated homozygous null (Sur2(-/-)) mice and heterozygote (Sur2(+/-)) mice that are viable and phenotypically similar to their wild-type littermates to 6 weeks of age despite, respectively, half or no SUR2 mRNA expression or channel activity in skeletal muscle or heart. Sur2(-/-) animals had lower fasting and fed serum glucose, exhibited improved glucose tolerance during a glucose tolerance test, and demonstrated a more rapid and severe hypoglycemia after administration of insulin. Enhanced glucose use was also observed during in vivo hyperinsulinemic euglycemic clamp studies during which Sur2(-/-) mice required a greater glucose infusion rate to maintain a target blood glucose level. Enhanced insulin action was intrinsic to the skeletal muscle, as in vitro insulin-stimulated glucose transport was 1.5-fold greater in Sur2(-/-) muscle than in wild type. Thus, membrane excitability and K(ATP) activity, to our knowledge, seem to be new components of the insulin-stimulated glucose uptake mechanism, suggesting possible future therapeutic approaches for individuals suffering from diabetes mellitus.


Subject(s)
ATP-Binding Cassette Transporters , Glucose/metabolism , Insulin/pharmacology , Muscle Proteins , Muscle, Skeletal/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Receptors, Drug/physiology , Analysis of Variance , Animals , Biological Transport , Blood Glucose/metabolism , Deoxyglucose/pharmacokinetics , Exons , Glucose Clamp Technique , Glucose Tolerance Test , Glucose Transporter Type 4 , Insulin/blood , Introns , Mice , Mice, Knockout , Monosaccharide Transport Proteins/genetics , Muscle, Skeletal/drug effects , Polymerase Chain Reaction , Potassium Channels/deficiency , Potassium Channels/genetics , RNA, Messenger/metabolism , Receptors, Drug/deficiency , Receptors, Drug/genetics , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Sulfonylurea Receptors , Triglycerides/blood , Weight Gain
6.
Pflugers Arch ; 439(6): 814-21, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10784357

ABSTRACT

The class IB antiarrhythmic drug, lidocaine, interacts strongly with depolarized sodium (Na) channels, an action that is thought to underlie its clinical efficacy. Previously, we have reported Na channel gating current (Ig) experiments with a quaternary form of lidocaine, QX-222, which binds preferentially to open Na channels and modifies the gating-charge/voltage (Q/V) relationship of cardiac Na channels by reducing maximal gating charge (Qmax) and lessening its voltage dependence. We report here investigations with lidocaine itself on Ig of native canine and cloned human cardiac Na channels. Although the state dependence of lidocaine binding to Na channels differs from that of quaternary drugs, Ig measurements demonstrated that lidocaine produced changes in the Q/V relationships similar to those elicited by QX-222, with a reduction in Qmax by 33% and a corresponding decrease in the slope factor. Concentration/response curves for the reduction in gating charge by lidocaine matched those for the block of sodium current (I(Na)), as would be expected if modification of Na channel voltage sensors by lidocaine underlied its action. The application of site-3 toxins, which inhibit movement of the voltage sensor associated with inactivation, to lidocaine-bound Na channels elicits an additional reduction in Qmax suggesting that lidocaine does not "stabilize" the Na channel in an inactivated state. We conclude that lidocaine blocks I(Na) by modification of the Na channel's voltage sensors predominately associated with channel activation leading to channel opening.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Ion Channel Gating/drug effects , Lidocaine/pharmacology , Myocardium/metabolism , Sodium Channels/drug effects , Sodium Channels/metabolism , Animals , Anti-Arrhythmia Agents/metabolism , Cell Line , Dogs , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiology , Humans , Ion Channel Gating/physiology , Lidocaine/metabolism , Saxitoxin/pharmacology , Sodium Channel Blockers , Sodium Channels/physiology
7.
Mol Pharmacol ; 57(1): 101-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617684

ABSTRACT

Flecainide block of Na(+) current (I(Na)) was investigated in wild-type (WT) or the long QT syndrome 3 (LQT3) sodium channel alpha subunit mutation with three amino acids deleted (DeltaKPQ) stably transfected into human embryonic kidney 293 cells using whole-cell, patch-clamp recordings. Flecainide (1-300 mM) caused tonic and use-dependent block (UDB) of I(Na) in a concentration-dependent manner. Compared with WT, DeltaKPQ I(Na) was more sensitive to flecainide, and flecainide preferentially inhibited late I(Na) (mean current between 20 and 23.5 ms after depolarization) compared with peak I(Na). The IC(50) value of peak and late I(Na) for WT was 127 +/- 6 and 44 +/- 2 microM (n = 20) and for DeltaKPQ was 80 +/- 9 and 19 +/- 2 microM (n = 31) respectively. UDB of peak I(Na) was greater and developed more slowly during pulse trains for DeltaKPQ than for WT. The IC(50) value for UDB of peak I(Na) for WT was 29 +/- 4 microM (n = 20) and for DeltaKPQ was 11 +/- 1 microM (n = 26). For DeltaKPQ, UDB of late I(Na) was greater than for peak I(Na). Recovery from block was slower for DeltaKPQ than for WT. We conclude that DeltaKPQ interacts differently with flecainide than with WT, leading to increased block and slowed recovery, especially for late I(Na). These data provide insights into mechanisms for flecainide block and provide a rationale at the cellular and molecular level that open channel block may be a useful pharmacological property for treatment of LQT3.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Long QT Syndrome/metabolism , Sodium Channel Blockers , Binding Sites , Cells, Cultured , Electrocardiography/drug effects , Electrophysiology , Humans , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Models, Molecular , Mutation , Quality Control , Sodium Channels/genetics , Sodium Channels/physiology
8.
Cardiovasc Res ; 42(2): 503-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10533585

ABSTRACT

OBJECTIVE: The affinity of lidocaine for the alpha-subunit of the Na channel has been reported to be greater for heart than for non-heart alpha-subunits, and also to be no different. Lidocaine block has a complex voltage dependence caused by a higher affinity for the inactivated state over the resting state. Inactivation kinetics, however, depend upon the alpha-subunit isoform and the presence of the auxiliary beta 1-subunit and will affect measures of block. METHODS: We studied the voltage dependence of lidocaine block of Na currents by a two microelectrode voltage clamp in oocytes injected with RNA for the Na channel alpha-subunits of human heart (hH1a) or a rat skeletal muscle (rSkM1) alone, or coexpressed with the beta 1-subunit. RESULTS: The midpoints of availability for a 25-s conditioning potential in control solutions were -65 mV for rSkM1, -50 for rSkM1 + beta 1, -78 mV for hH1a and -76 for hH1a + beta 1. The Kd of tonic lidocaine block was measured at -90, -100, -110, -120 and -130 mV in the same oocytes. The apparent Kd for both isoforms +/- beta 1 became greater with more negative holding potentials, but tended to reach different plateaus at -130 mV (Kd = 2128 microM for rSkM1, 1760 microM for rSkM1 + beta 1, 433 for hH1a, and 887 microM for hH1a + beta 1). Inactivated state affinities, assessed by fitting the shift in the Boltzmann midpoint of the availability relationship to the modulated receptor model, were 4 microM for rSkM1, 1 microM for rSkM1 + beta 1, 7 microM for hH1a and 9 microM for hH1a + beta 1. CONCLUSION: The heart Na channel alpha-subunits expressed in oocytes have an intrinsically higher rest state affinity for lidocaine compared to rSkM1 after the voltage- and state dependence of block are considered. Coexpression with beta 1 modestly increased the rest affinity of lidocaine for rSkM1, but had the opposite effect for hH1a.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Lidocaine/pharmacology , Oocytes/metabolism , Sodium Channels/metabolism , Animals , Binding, Competitive , Dose-Response Relationship, Drug , Female , Muscle, Skeletal/metabolism , Myocardium/metabolism , Oocytes/drug effects , Patch-Clamp Techniques , Protein Binding , Protein Isoforms/metabolism , Rats , Xenopus
9.
J Gen Physiol ; 114(2): 251-69, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10436001

ABSTRACT

Anionic phospholipids modulate the activity of inwardly rectifying potassium channels (Fan, Z., and J.C. Makielski. 1997. J. Biol. Chem. 272:5388-5395). The effect of phosphoinositides on adenosine triphosphate (ATP) inhibition of ATP-sensitive potassium channel (K(ATP)) currents was investigated using the inside-out patch clamp technique in cardiac myocytes and in COS-1 cells in which the cardiac isoform of the sulfonylurea receptor, SUR2, was coexpressed with the inwardly rectifying channel Kir6.2. Phosphoinositides (1 mg/ml) increased the open probability of K(ATP) in low [ATP] (1 microM) within 30 s. Phosphoinositides desensitized ATP inhibition with a longer onset period (>3 min), activating channels inhibited by ATP (1 mM). Phosphoinositides treatment for 10 min shifted the half-inhibitory [ATP] (K(i)) from 35 microM to 16 mM. At the single-channel level, increased [ATP] caused a shorter mean open time and a longer mean closed time. Phosphoinositides prolonged the mean open time, shortened the mean closed time, and weakened the [ATP] dependence of these parameters resulting in a higher open probability at any given [ATP]. The apparent rate constants for ATP binding were estimated to be 0.8 and 0.02 mM(-1) ms(-1) before and after 5-min treatment with phosphoinositides, which corresponds to a K(i) of 35 microM and 5.8 mM, respectively. Phosphoinositides failed to desensitize adenosine inhibition of K(ATP). In the presence of SUR2, phosphoinositides attenuated MgATP antagonism of ATP inhibition. Kir6.2DeltaC35, a truncated Kir6.2 that functions without SUR2, also exhibited phosphoinositide desensitization of ATP inhibition. These data suggest that (a) phosphoinositides strongly compete with ATP at a binding site residing on Kir6.2; (b) electrostatic interaction is a characteristic property of this competition; and (c) in conjunction with SUR2, phosphoinositides render additional, complex effects on ATP inhibition. We propose a model of the ATP binding site involving positively charged residues on the COOH-terminus of Kir6.2, with which phosphoinositides interact to desensitize ATP inhibition.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Phosphatidylinositols/pharmacology , Potassium Channel Blockers , Potassium Channels, Inwardly Rectifying , ATP-Binding Cassette Transporters , Adenosine Triphosphate/pharmacology , Algorithms , Animals , COS Cells , Electric Stimulation , Electrophysiology , KATP Channels , Kinetics , Membrane Potentials/physiology , Mixed Function Oxygenases/metabolism , Molecular Probes , Patch-Clamp Techniques , Potassium Channels/metabolism
10.
J Biol Chem ; 274(19): 13656-65, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224138

ABSTRACT

ATP-sensitive potassium channels (KATP) are implicated in a diverse array of physiological functions. Previous work has shown that alternative usage of exons 14, 39, and 40 of the muscle-specific KATP channel regulatory subunit, sur2, occurs in tissue-specific patterns. Here, we show that exon 17 of the first nucleotide binding fold of sur2 is also alternatively spliced. RNase protection demonstrates that SUR2(Delta17) predominates in skeletal muscle and gut and is also expressed in bladder, fat, heart, lung, liver, and kidney. Polymerase chain reaction and restriction digest analysis of sur2 cDNA demonstrate the existence of at least five sur2 splice variants as follows: SUR2(39), SUR2(40), SUR2(Delta17/39), SUR2(Delta17/40), and SUR2(Delta14/39). Electrophysiological recordings of excised, inside-out patches from COS cells cotransfected with Kir6.2 and the sur2 variants demonstrated that exon 17 splicing alters KATP sensitivity to ATP block by 2-fold from approximately 40 to approximately 90 microM for exon 17 and Delta17, respectively. Single channel kinetic analysis of SUR2(39) and SUR2(Delta17/39) demonstrated that both exhibited characteristic KATP kinetics but that SUR2(Delta17/39) exhibited longer mean burst durations and shorter mean interburst dwell times. In sum, alternative splicing of sur2 enhances the observed diversity of KATP and may contribute to tissue-specific modulation of ATP sensitivity.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/pharmacology , Alternative Splicing , Exons , Mixed Function Oxygenases/genetics , Potassium Channels/drug effects , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Mice , Mixed Function Oxygenases/chemistry , Molecular Sequence Data , Potassium Channels/physiology , Potassium Channels, Inwardly Rectifying , Receptors, Drug , Recombinant Proteins/drug effects , Sulfonylurea Receptors
11.
Circ Res ; 84(9): 989-98, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10325236

ABSTRACT

Calcium channel antagonists have diverse effects on cardiac electrophysiology. We studied the effects of verapamil, diltiazem, and nifedipine on HERG K+ channels that encode IKr in native heart cells. In our experiments, verapamil caused high-affinity block of HERG current (IC50=143.0 nmol/L), a value close to those reported for verapamil block of L-type Ca2+ channels, whereas diltiazem weakly blocked HERG current (IC50=17.3 micromol/L), and nifedipine did not block HERG current. Verapamil block of HERG channels was use and frequency dependent, and verapamil unbound from HERG channels at voltages near the normal cardiac cell resting potential or with drug washout. Block of HERG current by verapamil was reduced by lowering pHO, which decreases the proportion of drug in the membrane-permeable neutral form. N-methyl-verapamil, a membrane-impermeable, permanently charged verapamil analogue, blocked HERG channels only when applied intracellularly. Verapamil antagonized dofetilide block of HERG channels, which suggests that they may share a common binding site. The C-type inactivation-deficient mutations, Ser620Thr and Ser631Ala, reduced verapamil block, which is consistent with a role for C-type inactivation in high-affinity drug block, although the Ser620Thr mutation decreased verapamil block 20-fold more than the Ser631Ala mutation. Our findings suggest that verapamil enters the cell membrane in the neutral form to act at a site within the pore accessible from the intracellular side of the cell membrane, possibly involving the serine at position 620. Thus, verapamil shares high-affinity HERG channel blocking properties with other class III antiarrhythmic drugs, and this may contribute to its antiarrhythmic mechanism.


Subject(s)
Anti-Arrhythmia Agents/metabolism , Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Verapamil/metabolism , Verapamil/pharmacology , Cell Line , Diltiazem/pharmacology , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/physiology , Mutation/physiology , Nifedipine/pharmacology , Phenethylamines/antagonists & inhibitors , Phenethylamines/pharmacology , Potassium Channels/genetics , Potassium Channels/physiology , Sulfonamides/antagonists & inhibitors , Sulfonamides/pharmacology , Transcriptional Regulator ERG , Verapamil/analogs & derivatives
12.
Am J Physiol ; 275(6): H2016-24, 1998 12.
Article in English | MEDLINE | ID: mdl-9843800

ABSTRACT

Na+ current (INa) through wild-type human heart Na+ channels (hH1) is important for normal cardiac excitability and conduction, and it participates in the control of repolarization and refractoriness. INa kinetics depend strongly on temperature, but INa for hH1 has been studied previously only at room temperature. We characterized early INa (the peak and initial decay) and late INa of the wild-type hH1 channel and a mutant channel (DeltaKPQ) associated with congenital long Q-T syndrome. Channels were stably transfected in HEK-293 cells and studied at 23 and 33 degreesC using whole cell patch clamp. Activation and inactivation kinetics for early INa were twofold faster at higher temperature for both channels and shifted activation and steady-state inactivation in the positive direction, especially for DeltaKPQ. For early INa (<24 ms), DeltaKPQ decayed faster than the wild type for voltages negative to -20 mV but slower for more positive voltages, suggesting a reduced voltage dependence of fast inactivation. Late INa at 240 ms was significantly greater for DeltaKPQ than for the wild type at both temperatures. The majority of late INa for DeltaKPQ was not persistent; rather, it decayed slowly, and this late component exhibited slower recovery from inactivation compared with peak INa. Additional kinetic changes for early and peak INa for DeltaKPQ compared with the wild type at both temperatures were 1) reduced voltage dependence of steady-state inactivation with no difference in midpoint, 2) positive shift for activation kinetics, and 3) more rapid recovery from inactivation. This study represents the first description of human Na+ channel kinetics near physiological temperature and also demonstrates complex gating changes in the DeltaKPQ that are present at 33 degreesC and that may underlie the electrophysiological and clinical phenotype of congenital long Q-T Na+ channel syndromes.


Subject(s)
Long QT Syndrome/metabolism , Mutation/physiology , Myocardium/metabolism , Sodium Channels/genetics , Sodium Channels/physiology , Temperature , Cell Line , Electric Conductivity , Homeostasis/physiology , Humans , Kinetics , Long QT Syndrome/genetics , Reaction Time/physiology , Time Factors
13.
Biophys J ; 74(1): 230-41, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9449325

ABSTRACT

We have established stably transfected HEK 293 cell lines expressing high levels of functional human ether-a go-go-related gene (HERG) channels. We used these cells to study biochemical characteristics of HERG protein, and to study electrophysiological and pharmacological properties of HERG channel current at 35 degrees C. HERG-transfected cells expressed an mRNA band at 4.0 kb. Western blot analysis showed two protein bands (155 and 135 kDa) slightly larger than the predicted molecular mass (127 kDa). Treatment with N-glycosidase F converted both bands to smaller molecular mass, suggesting that both are glycosylated, but at different levels. HERG current activated at voltages positive to -50 mV, maximum current was reached with depolarizing steps to -10 mV, and the current amplitude declined at more positive voltages, similar to HERG channel current expressed in other heterologous systems. Current density at 35 degrees C, compared with 23 degrees C, was increased by more than twofold to a maximum of 53.4 +/- 6.5 pA/pF. Activation, inactivation, recovery from inactivation, and deactivation kinetics were rapid at 35 degrees C, and more closely resemble values reported for the rapidly activating delayed rectifier K+ current (I(Kr)) at physiological temperatures. HERG channels were highly selective for K+. When we used an action potential clamp technique, HERG current activation began shortly after the upstroke of the action potential waveform. HERG current increased during repolarization to reach a maximum amplitude during phases 2 and 3 of the cardiac action potential. HERG contributed current throughout the return of the membrane to the resting potential, and deactivation of HERG current could participate in phase 4 depolarization. HERG current was blocked by low concentrations of E-4031 (IC50 7.7 nM), a value close to that reported for I(Kr) in native cardiac myocytes. Our data support the postulate that HERG encodes a major constituent of I(Kr) and suggest that at physiological temperatures HERG contributes current throughout most of the action potential and into the postrepolarization period.


Subject(s)
Action Potentials/physiology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Trans-Activators , Anti-Arrhythmia Agents/pharmacology , Cell Line , Cell Membrane/physiology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Kidney , Kinetics , Membrane Potentials/drug effects , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium Channels/biosynthesis , Potassium Channels/drug effects , Pyridines/pharmacology , Recombinant Proteins/biosynthesis , Temperature , Time Factors , Transcriptional Regulator ERG , Transfection
14.
J Biol Chem ; 272(9): 5388-95, 1997 Feb 28.
Article in English | MEDLINE | ID: mdl-9038137

ABSTRACT

The ATP-sensitive potassium channel (KATP) controls insulin release in pancreatic beta-cells and also modulates important functions in other cell types. In this study we report that anionic phospholipids activated KATP in pancreatic beta-cells, cardiac myocytes, skeletal muscle cells, and a cloned KATP composed of two subunits (SUR/Kir6. 2) stably expressed in a mammalian cell line. The effectiveness was proportional to the number of negative charges on the head group of the anionic phospholipid. Screening negative charges with polyvalent cations antagonized the effect. Enzymatic treatment with phospholipases that reduced charge on the lipids also reduced or eliminated the effect. These results suggest that intact phospholipids with negative charges are the critical requirement for activation of KATP, in distinction from the usual cell signaling pathway through phospholipids that requires cleavage. Mutations of two positively charged amino acid residues at the C terminus of Kir6. 2 accelerated loss of channel activity and reduced the activating effects of phospholipids, suggesting involvement of this region in the activation. Metabolism of anionic phospholipids in plasmalemmal membrane may be a novel and general mechanism for regulation of KATP and perhaps other ion channels in the family of inward rectifiers.


Subject(s)
Phospholipids/pharmacology , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , ATP-Binding Cassette Transporters , Amino Acid Sequence , Animals , Cattle , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , KATP Channels , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Mutagenesis , Myocardium/metabolism , Potassium Channels/genetics , Rats , Rats, Wistar
15.
J Physiol ; 496 ( Pt 1): 275-86, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8910215

ABSTRACT

1. Two mutant human skeletal muscle voltage-gated Na+ channel alpha-subunits (hSkM1), with mutations found in patients with hereditary paramyotonia congenita (T1313M on the III-IV linker and R1448C on the outside of S4 of repeat IV), and wild-type hSkM1 channels were expressed in a human embryonic kidney cell lines (tsA201) using recombinant cDNA. 2. Compared with wild-type, both mutants exhibited altered inactivation phenotypes. Current decay was slowed for both, but voltage-dependent availability from inactivation was shifted in the negative direction for R1448C and in the positive direction for T1313M. 3. The hypothesis that a local anaesthetic, lidocaine (lignocaine), binds primarily to the inactivated state to block the channel was reassessed by testing lidocaine block of these two mutants and the wild-type channel. 4. T1313M showed reduced phasic block, but R1448C showed increased phasic block for trains of depolarizations. 5. Rest block (from -120 mV) was increased for R1448C (IC50 approximately equal to 0.2 mM) and decreased for T1313M (IC50 approximately equal to 1.3 mM) compared with wild-type (IC50 approximately 0.5 mM), but these differences were diminished at a holding potential of -150 mV, suggesting that the differences were caused by binding to the inactivated state rather than a different affinity of lidocaine for the resting state. 6. Inactivated state affinity measured from lidocaine-induced shifts in voltage-dependent availability was reduced for T1313M (Kd = 63 microM) but little changed for R1448C (Kd = 14 microM) compared with wild-type (Kd = 11 microM). Two pulse recovery protocols showed faster recovery from lidocaine block for T1313M and slower recovery for R1448C. Together these accounted for the opposite effects on lidocaine phasic block observed for the mutant channels. 7. Neither mutation is located at a putative lidocaine binding site in domain 4 S6, yet both affected lidocaine block. The data suggest that R1448C altered phasic lidocaine block mainly through altered kinetics, but T1313M altered block through a change in affinity for the inactivated state. These findings have implications for drug therapy of paramyotonia congenita, and also provide an insight into structural requirements for drug affinity.


Subject(s)
Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Myotonia Congenita/genetics , Sodium Channels/metabolism , Cell Line , Electrophysiology , Humans , Membrane Potentials/physiology , Mutation , Myotonia Congenita/metabolism , Patch-Clamp Techniques , Phenotype , Sodium Channels/drug effects , Structure-Activity Relationship
16.
Jpn Heart J ; 37(5): 733-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8973385

ABSTRACT

The heart Na channel, although resembling other voltage-gated Na channels, has important functional and structural differences. For heart channels expressed in oocytes, the midpoint of the inactivation relationship was 13 mV negative to that of rat skeletal muscle Na channels, and sensitivity to tonic lidocaine block was approximately 5 times more sensitive for heart. Co-expression with the beta subunit increased the difference in inactivation midpoint to 24 mV, largely by changing the midpoint of the rat skeletal muscle channel by 10 mV in the positive direction. Co-expression with beta 1 decreased lidocaine sensitivity for heart but not for skeletal muscle Na channels, and decreased but did not eliminate the greater heart sensitivity to lidocaine block. The differences in inactivation are likely to account for some, but not all, of the differences in lidocaine sensitivity. This cardiac phenotype is important for the role the channel plays in cardiac physiology and pathophysiology, and also may lead to elucidation of structure-function relationships.


Subject(s)
Anesthetics, Local/pharmacology , Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Lidocaine/pharmacology , Sodium Channels/physiology , Animals , Electric Conductivity , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Phenotype , Rats , Sodium Channels/drug effects
17.
Am J Physiol ; 271(2 Pt 2): H790-7, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8770124

ABSTRACT

Single Na channels from rat and rabbit ventricular cells were studied with use of the excised inside-out patch-clamp technique. To investigate local anesthetic interactions with Na channels modified by the ischemic metabolite lysophosphatidylcholine (LPC), the quaternary ammonium lidocaine derivative QX-222 [2-(trimethylamino)-N-(2,6-dimethylphenyl)acetamide] was applied to the cytoplasmic side of patches from untreated cells and from those treated with LPC for approximately 1 h. Single-channel amplitudes and kinetics for unmodified channels were similar to those reported previously for cardiac cells with a single-component, mean-channel open time. LPC-modified channels showed prolonged open channel bursting with a two-component, mean open time, suggesting two open states. Conductance sublevels to the 60-70% level of the main conductance were found in both unmodified and LPC-modified channels and also with and without QX-222 present. QX-222 reversibly shortened the open time of the unmodified channel and for both open times of the LPC-modified channel without decreasing single-channel amplitude. Calculated association rates for QX-222 with the channel were found to be greater for the open states of the modified channel than those for the unmodified channel. Thus the lidocaine analogue QX-222 interacts with and blocks the open state of both unmodified and LPC-modified, cardiac Na channels. The blocking effect on LPC-modified channels would be predicted to be greater both because of the longer dwell time in the high-affinity open states for modified channels and also because of an intrinsically greater association rate in the modified channels.


Subject(s)
Lidocaine/analogs & derivatives , Lysophosphatidylcholines/pharmacology , Myocardium/metabolism , Sodium Channel Blockers , Sodium Channels/drug effects , Animals , Electric Conductivity , Lidocaine/pharmacology , Myocardium/cytology , Patch-Clamp Techniques , Rabbits , Rats , Sodium Channels/physiology
18.
J Mol Cell Cardiol ; 28(4): 743-53, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8732502

ABSTRACT

Lysophosphatidylcholine (LPC), an ischemic metabolite implicated in arrhythmogenesis, has been shown to modulate aspects of Na+ channel gating, but its effects on steady-state availability (h infinity), recovery from inactivation, and the timing of onset and possible reversibility, have not been characterized. We studied Na current (INa) by the whole-cell patch clamp technique on isolated rat ventricular myocytes at 22 degrees C with reduced Na+ (45 mM out, 5 mM in) from a holding potential of -150 mV. Changes in the electrophysiological parameters were measured after LPC 10 microM was added to the bath and compared to time controls (TC) taken from the time of seal formation. LPC decreased peak current for a test potential to -30 mV by about 20%. The peak current voltage relationship shifted in a positive direction by about 5 mV after LPC as compared to a small 2 mV negative shift in TC cells. LPC shifted the steady-state availability curve in the hyperpolarizing direction by about 6 mV. LPC perfusion caused a slowing of the decay of INa, and also a slowing of recovery from inactivation. Onset of the effects occurred within 6 min after adding LPC to the bath and were statistically significant with respect to TC cells between 12 and 16 min. In three cells, some of the effects on INa were either arrested or partially reversed by washout and cell survival was less than 20 min if LPC was not removed from the bath. These LPC induced changes in INa would tend to slow conduction and increase refractoriness, effects also seen in acutely ischemic myocardium. We therefore conclude that LPC action on INa may potentiate the arrhythmogenic substrate and that the onset of these changes are sufficiently rapid to play a role in the electrical instability of acute ischemia.


Subject(s)
Action Potentials/drug effects , Lysophosphatidylcholines/pharmacology , Myocardium/metabolism , Sodium Channels/metabolism , Sodium Channels/physiology , Animals , Arrhythmias, Cardiac/metabolism , Cells, Cultured , Myocardial Ischemia/metabolism , Myocardium/cytology , Rats
19.
Biochem Biophys Res Commun ; 220(3): 532-8, 1996 Mar 27.
Article in English | MEDLINE | ID: mdl-8607800

ABSTRACT

The ATP-sensitive potassium channel of insulin-secreting pancreatic beta-cells is a complex of Kir6.2, a member of the inwardly rectifying potassium channel superfamily, and the sulfonylurea receptor. We have isolated cDNA clones encoding rat Kir6.2. Co-expression of rat Kir6.2 and sulfonylurea receptor in human embryonic kidney cells generated a potassium current with the properties of the beta-cell ATP-sensitive potassium channel. A quantitative reverse transcriptase-polymerase chain reaction assay indicated that Kir6.2 and sulfonylurea receptor mRNAs were abundantly expressed in rat islets and that expression of Kir6.2 mRNA was reduced by >70% in islets from Zucker diabetic fatty male rats, whereas there was no significant change in sulfonylurea receptor mRNA levels. Thus, decreased expression of Kir6.2 could contribute to the beta-cell dysfunction which characterizes diabetes mellitus in this animal model.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus/physiopathology , Gene Expression , Islets of Langerhans/metabolism , Obesity , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Line , Cloning, Molecular , DNA Primers , Diabetes Mellitus/genetics , Diabetes Mellitus, Type 2/genetics , Glyburide/pharmacology , Humans , Islets of Langerhans/physiopathology , Male , Membrane Potentials/drug effects , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Potassium Channels/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Zucker , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Reference Values , Sequence Homology, Amino Acid
20.
Mol Pharmacol ; 49(1): 30-9, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8569709

ABSTRACT

Coexpression of the rat beta 1 subunit with rat brain and skeletal muscle sodium channel alpha subunits in Xenopus oocytes normalizes currents by accelerating sodium current decay kinetics, shifting steady state availability relationships, and accelerating recovery from inactivation. Unlike brain and skeletal muscle, the heart alpha subunit expressed without beta 1 has native-like decay kinetics in oocytes. Messenger RNA for beta 1 has been found in heart, but whether and how it affects cardiac sodium channel function are unclear. We studied coexpression of human heart alpha subunit with beta 1 in Xenopus oocytes using two microelectrode voltage-clamp and macropatch techniques. Coexpression with beta 1 caused a significant positive shift of 3-7 mV in the midpoint of the steady state inactivation relationship but did not affect single-channel conductance, activation, current decay, or recovery from inactivation. Sensitivity to lidocaine block, however, was decreased for both resting state block (Kd = 0.5-1.3 mM) and phasic block in response to pulse trains, but inactivated state block was not affected (Kd = approximately 10 microM). Coexpression with beta 1 increased the rate of recovery from lidocaine block, which accounted for the major part of the observed differences in tonic and phasic block. A beta 1 construct with the cytoplasmic tail removed also produced these effects, demonstrating that the beta 1 cytoplasmic tail was not involved in altering lidocaine block. We conclude that the beta 1 subunit is capable of affecting function of the cardiac sodium channel in oocytes by decreasing tonic and phasic lidocaine block with small effects on gating.


Subject(s)
Lidocaine/pharmacology , Myocardium/metabolism , Sodium Channel Blockers , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Humans , Membrane Potentials , Molecular Sequence Data , Oocytes , Rats , Sodium Channels/genetics , Sodium Channels/physiology , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...