Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 39662, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045073

ABSTRACT

Pain places a devastating burden on patients and society and current pain therapeutics exhibit limitations in efficacy, unwanted side effects and the potential for drug abuse and diversion. Although genetic evidence has clearly demonstrated that the voltage-gated sodium channel, Nav1.7, is critical to pain sensation in mammals, pharmacological inhibitors of Nav1.7 have not yet fully recapitulated the dramatic analgesia observed in Nav1.7-null subjects. Using the tarantula venom-peptide ProTX-II as a scaffold, we engineered a library of over 1500 venom-derived peptides and identified JNJ63955918 as a potent, highly selective, closed-state Nav1.7 blocking peptide. Here we show that JNJ63955918 induces a pharmacological insensitivity to pain that closely recapitulates key features of the Nav1.7-null phenotype seen in mice and humans. Our findings demonstrate that a high degree of selectivity, coupled with a closed-state dependent mechanism of action is required for strong efficacy and indicate that peptides such as JNJ63955918 and other suitably optimized Nav1.7 inhibitors may represent viable non-opioid alternatives for the pharmacological treatment of severe pain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/metabolism , Spider Venoms/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Cell Line , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Male , Pain/prevention & control , Rats, Sprague-Dawley , Spider Venoms/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry
2.
Future Med Chem ; 4(5): 661-79, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22458684

ABSTRACT

Ion channels are targets of many therapeutically useful agents, and worldwide sales of ion channel-targeted drugs are estimated to be approximately US$12 billion. Nevertheless, considering that over 400 genes encoding ion channel subunits have been identified, ion channels remain significantly under-exploited as therapeutic targets. This is at least partly due to limitations in high-throughput assay technologies that support screening and lead optimization. Will the recent developments in automated electrophysiology rectify this situation? What are the other major limitations and can they be overcome? In this article, we review the status of ion channel drug discovery, discuss current challenges and propose alternative approaches that may facilitate the discovery of new drugs in the future.


Subject(s)
Drug Discovery , Ion Channels/chemistry , Biological Products/chemistry , Clinical Trials as Topic , High-Throughput Screening Assays , Humans , Ion Channels/metabolism , Ligands , Structure-Activity Relationship
3.
Methods Find Exp Clin Pharmacol ; 32(8): 557-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21132125

ABSTRACT

This study examined the efficacy of a novel TRPV1 antagonist, JNJ-17203212, in two experimental rat models that exhibit a hypersensitive visceral motor response (VMR) to colorectal distension (CRD). In the first model, intraluminal administration of acetic acid (1% solution) into the distal colon produced an acute colonic hypersensitivity. In the second model, intraluminal administration of 2,4,6-trinitrobenzenesulfonic acid (TNBS) into the distal colon produced a chronic, post-inflammatory colonic hypersensitivity 30 days post-TNBS administration. Throughout this study, colonic sensitivity was assessed via quantification of VMR to CRD in rats following a single, oral administration of JNJ-17203212 (3, 10 or 30 mg/kg) or vehicle. Intraluminal administration of acetic acid and TNBS resulted in increased VMR to CRD when compared to controls. In both groups, VMR to CRD was significantly reduced by administration of JNJ-17203212 at 30 mg/kg. The results of this study show that the selective TRPV1 antagonist, JNJ-17203212, reduces sensitivity to luminal distension in both an acute, noninflammatory and a chronic, post-inflammatory rodent model of colonic hypersensitivity. These data indicate that TRPV1 is involved in the pathogenesis of visceral hypersensitivity and that JNJ-17203212 may be a potential therapeutic agent for functional bowel disorders characterized by abdominal hypersensitivity, such as irritable bowel syndrome.


Subject(s)
Aminopyridines/pharmacology , Irritable Bowel Syndrome/drug therapy , Piperazines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Acetic Acid , Administration, Oral , Aminopyridines/administration & dosage , Animals , Colon/drug effects , Colon/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Irritable Bowel Syndrome/physiopathology , Male , Piperazines/administration & dosage , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid
4.
Am J Physiol Cell Physiol ; 298(6): C1457-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20457836

ABSTRACT

TRPA1 is a nonselective cation channel belonging to the transient receptor potential (TRP) family that is expressed in peripheral sensory neurons and may play important roles in pain perception and inflammation. We found that agonist stimulation of TRPA1, along with other members of the TRP family (TRPV1-4 and TRPM8), can induce the appearance of a large pore permeable to large organic cations such as Yo-Pro (YP) and N-methyl-d-glucamine, in an agonist and divalent cation-dependent manner. YP uptake was not inhibited by a panel of putative gap junction/pannexin blockers, suggesting that gap junction proteins are not required in this process. Our data suggest that changes in the TRP channel selectivity filter itself result in a progressive but reversible pore dilation process, a process that is under strong regulation by external calcium ions. Our data suggest that calcium plays a novel role in setting the amount of time TRPA1 channels spend in a dilated state providing a mechanism that may limit sensory neuron activation by painful or irritating substances.


Subject(s)
Calcium Channels/metabolism , Cell Membrane Permeability , Cell Membrane/metabolism , Ion Channel Gating , Nerve Tissue Proteins/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Benzamides/pharmacology , Benzoxazoles/metabolism , CHO Cells , Calcium/metabolism , Calcium Channels/genetics , Carbamates/pharmacology , Cell Membrane/drug effects , Cell Membrane Permeability/drug effects , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Farnesol/analogs & derivatives , Farnesol/pharmacology , Humans , Ion Channel Gating/drug effects , Isothiocyanates/pharmacology , Kinetics , Meglumine/metabolism , Membrane Transport Modulators/pharmacology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Purinergic P2 Receptor Agonists , Quinolinium Compounds/metabolism , Rats , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X7 , Salicylates/pharmacology , TRPA1 Cation Channel , TRPM Cation Channels/agonists , TRPM Cation Channels/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Transfection , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/genetics
5.
Curr Pharm Des ; 15(18): 2149-68, 2009.
Article in English | MEDLINE | ID: mdl-19519450

ABSTRACT

This article reviews evidence that hyperpolarization-activated, cation nonselective (HCN) channels, the molecular basis of the Ih current, potentially represent valid targets for novel analgesic agents. Ih is a prominent current in many peripheral sensory nerves, with highest current density typically found in large diameter neurons. Recent data suggest that Ih may represent a valid target for the treatment of spontaneous pain and allodynia associated with nerve injury. The majority of available electrophysiological and molecular evidence suggests that fast activating, weakly cyclic adenosine monophosphate (cAMP) sensitive HCN1-based channels may make a significant contribution to Ih, especially in large diameter, mechanosensitive fibers, where the Ih current appears to support abnormal spontaneous firing after nerve injury. In contrast, HCN4 channels seem to play the predominant role in cardiac pacemaker tissue. These observations raise the possibility that HCN1 selective blockers may inhibit pain associated with nerve injury without dramatic effects on heart rate. Development of novel HCN blocking analgesics presents a number of significant technical challenges. Although a number of HCN blockers are available, such as ZD-7288, ivabradine, and others, these drugs inhibit all HCN isoforms with the same potency. As a result, these compounds have powerful effects on heart rate, severely limiting their utility for non-cardiac indications such as pain. Selectivity challenges, mechanisms of compound interaction with the channel, and assay methods are described in detail.


Subject(s)
Analgesics/pharmacology , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Cyclic Nucleotide-Gated Cation Channels/physiology , Drug Discovery/methods , Analgesics/chemistry , Animals , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Models, Biological , Molecular Structure , Pain/drug therapy , Pain/physiopathology , Potassium Channels/physiology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology
6.
Curr Pharm Des ; 15(15): 1773-98, 2009.
Article in English | MEDLINE | ID: mdl-19442190

ABSTRACT

Kv7.x channels are a family of six transmembrane domain, single pore-loop, voltage-gated K(+) channels. Five members of the family have been identified to date, including the cardiac channel Kv7.1 (formerly known as KvLQT1) and four neuronal Kv7.x channels, Kv7.2-5. Heteromeric channels containing Kv7.3 and either Kv7.2 or Kv7.5 are thought to underlie the neuronal M-current, a non-inactivating, slowly deactivating, sub-threshold current that has long been known to exert a powerful stabilizing influence on neuronal excitability. Modulators of these channels have the potential to influence neuronal activity in various tissues and are of much interest as therapeutic drug targets for the treatment of a variety of clinical disorders, such as epilepsy and pain. The purpose of the present article is to review the molecular, functional and behavioral evidence validating Kv7.x as drug targets for the treatment of pain. In addition, an update on pre-clinical Kv7 drug discovery efforts will be presented, along with a summary of on-going clinical trials with Kv7 channel activators.


Subject(s)
Analgesics/pharmacology , KCNQ Potassium Channels/drug effects , Pain/drug therapy , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Delivery Systems , Drug Design , Drug Evaluation, Preclinical , Humans , KCNQ Potassium Channels/metabolism , Neurons/metabolism , Pain/physiopathology
8.
Mol Pharmacol ; 73(3): 977-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089837

ABSTRACT

KCNQ2 (Kv7.2) and KCNQ3 (Kv7.3) are voltage-gated K(+) channel subunits that underlie the neuronal M current. In humans, mutations in these genes lead to a rare form of neonatal epilepsy (Biervert et al., 1998; Singh et al., 1998), suggesting that KCNQ2/Q3 channels may be attractive targets for novel antiepileptic drugs. In the present study, we have identified the compound N-(6-chloro-pyridin-3-yl)-3,4-difluoro-benzamide (ICA-27243) as a selective activator of the neuronal M current and KCNQ2/Q3 channels. In SH-SY5Y human neuroblastoma cells, ICA-27243 produced membrane potential hyperpolarization that could be prevented by coadministration with the M-current inhibitors 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE-991) and linopirdine. ICA-27243 enhanced both (86)Rb(+) efflux (EC(50) = 0.2 microM) and whole-cell currents in Chinese hamster ovary cells stably expressing heteromultimeric KCNQ2/Q3 channels (EC(50) = 0.4 microM). Activation of KCNQ2/Q3 channels was associated with a hyperpolarizing shift of the voltage dependence of channel activation (V((1/2)) shift of -19 mV at 10 microM). In contrast, ICA-27243 was less effective at activating KCNQ4 and KCNQ3/Q5 and was selective over a wide range of neurotransmitter receptors and ion channels such as voltage-dependent sodium channels and GABA-gated chloride channels. ICA-27243 (1-10 microM) was found to reversibly suppress seizure-like activity in an ex vivo hippocampal slice model of epilepsy and demonstrated in vivo anticonvulsant activity (ED(50) = 8.4 mg/kg) in the mouse maximal electroshock epilepsy model. In conclusion, ICA-27243 represents the first member of a novel chemical class of selective KCNQ2/Q3 activators with anticonvulsant-like activity in experimental models of epilepsy.


Subject(s)
Benzamides/pharmacology , KCNQ2 Potassium Channel/drug effects , KCNQ3 Potassium Channel/drug effects , Pyridines/pharmacology , Animals , CHO Cells , Cell Culture Techniques , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electrophysiology , Hippocampus/metabolism , Humans , Inhibitory Concentration 50 , Kidney/cytology , Male , Membrane Potentials/drug effects , Microelectrodes , Neuroblastoma/pathology , Patch-Clamp Techniques , Plasmids , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
9.
Br J Pharmacol ; 132(2): 381-4, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11159685

ABSTRACT

Heteromeric KCNQ5/Q3 channels were stably expressed in Chinese Hamster ovary cells and characterized using the whole cell voltage-clamp technique. KCNQ5/Q3 channels were activated by the novel anticonvulsant, retigabine (EC(50) 1.4 microM) by a mechanism that involved drug-induced, leftward shifts in the voltage-dependence of channel activation (-31.8 mV by 30 microM retigabine). KCNQ5/Q3 channels were inhibited by linopirdine (IC(50) 7.7 microM) and barium (IC(50) 0.46 mM), at concentrations similar to those required to inhibit native M-currents. These findings identify KCNQ5/Q3 channels as a molecular target for retigabine and raise the possibility that activation of KCNQ5/Q3 channels may be responsible for some of the anti-convulsant activity of this agent. Furthermore, the sensitivity of KCNQ5/Q3 channels to linopirdine supports the possibility that potassium channels comprised of KCNQ5 and KCNQ3 may make a contribution to native M-currents.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , Anticonvulsants/pharmacology , Barium/pharmacology , CHO Cells , Carbamates/pharmacology , Cricetinae , Electrophysiology , Indoles/pharmacology , KCNQ3 Potassium Channel , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Plasmids , Potassium Channels/agonists , Potassium Channels/genetics , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
10.
Mol Pharmacol ; 58(3): 591-600, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953053

ABSTRACT

Retigabine [N-(2-amino-4-[fluorobenzylamino]-phenyl) carbamic acid; D-23129] is a novel anticonvulsant, unrelated to currently available antiepileptic agents, with activity in a broad range of seizure models. In the present study, we sought to determine whether retigabine could enhance current through M-like currents in PC12 cells and KCNQ2/Q3 K(+) channels expressed in Chinese hamster ovary cells (CHO-KCNQ2/Q3). In differentiated PC12 cells, retigabine enhanced a linopirdine-sensitive current. The effect of retigabine was associated with a slowing of M-like tail current deactivation in these cells. Retigabine (0.1 to 10 microM) induced a potassium current and hyperpolarized CHO cells expressing KCNQ2/Q3 cells but not in wild-type cells. Retigabine-induced currents in CHO-KCNQ2/Q3 cells were inhibited by 60.6 +/- 11% (n = 4) by the KCNQ2/Q3 blocker, linopirdine (10 microM), and 82.7 +/- 5.4% (n = 4) by BaCl(2) (10 mM). The mechanism by which retigabine enhanced KCNQ2/Q3 currents involved large, drug-induced, leftward shifts in the voltage dependence of channel activation (-33.1 +/- 2.6 mV, n = 4, by 10 microM retigabine). Retigabine shifted the voltage dependence of channel activation with an EC(50) value of 1.6 +/- 0.3 microM (slope factor was 1.2 +/- 0.1, n = 4 to 5 cells per concentration). Retigabine (0.1 to 10 microM) also slowed the rate of channel deactivation, predominantly by increasing the contribution of a slowly deactivating tail current component. Our findings identify KCNQ2/Q3 channels as a molecular target for retigabine and suggest that activation of KCNQ2/Q3 channels may be responsible for at least some of the anticonvulsant activity of this agent.


Subject(s)
Anticonvulsants/pharmacology , Carbamates/pharmacology , Phenylenediamines/pharmacology , Potassium Channels/metabolism , Animals , Barium/pharmacology , CHO Cells , Cell Differentiation , Cell Line , Cricetinae , Drug Interactions , Electrophysiology , Humans , Indoles/pharmacology , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , PC12 Cells , Potassium/metabolism , Potassium/physiology , Potassium Channels/drug effects , Potassium Channels, Voltage-Gated , Pyridines/pharmacology , Rats , Transfection
11.
Am J Physiol Heart Circ Physiol ; 278(4): H1105-16, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10749704

ABSTRACT

Previous studies have established that reductions in repolarizing currents occur in heart disease and can contribute to life-threatening arrhythmias in myocardium. In this study, we investigated whether the thyroid hormone analog 3, 5-diiodothyropropionic acid (DITPA) could restore repolarizing transient outward K(+) current (I(to)) density and gene expression in rat myocardium after myocardial infarction (MI). Our findings show that I(to) density was reduced after MI (14.0 +/- 1.0 vs. 10.2 +/- 0.9 pA/pF, sham vs. post-MI at +40 mV). mRNA levels of Kv4.2 and Kv4.3 genes were decreased but Kv1.4 mRNA levels were increased post-MI. Corresponding changes in Kv4.2 and Kv1.4 protein were also observed. Chronic treatment of post-MI rats with 10 mg/kg DITPA restored I(to) density (to 15.2 +/- 1.1 pA/pF at +40 mV) as well as Kv4.2 and Kv1.4 expression to levels observed in sham-operated controls. Other membrane currents (Na(+), L-type Ca(2+), sustained, and inward rectifier K(+) currents) were unaffected by DITPA treatment. Associated with the changes in I(to) expression, action potential durations (current-clamp recordings in isolated single right ventricular myocytes and monophasic action potential recordings from the right free wall in situ) were prolonged after MI and restored with DITPA treatment. Our results demonstrate that DITPA restores I(to) density in the setting of MI, which may be useful in preventing complications associated with I(to) downregulation.


Subject(s)
Action Potentials/drug effects , Diiodothyronines/pharmacology , Myocardial Infarction/drug therapy , Potassium Channels, Voltage-Gated , Propionates/pharmacology , Animals , Dose-Response Relationship, Drug , Electrophysiology , Gene Expression/drug effects , Heart Ventricles/chemistry , Heart Ventricles/cytology , Heart Ventricles/metabolism , Kv1.4 Potassium Channel , Male , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/metabolism , Myocardium/chemistry , Myocardium/cytology , Myocardium/metabolism , Potassium/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Receptors, Thyroid Hormone/physiology , Shal Potassium Channels
12.
J Biol Chem ; 275(9): 6453-61, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10692449

ABSTRACT

We present the cloning and characterization of two novel calcium-activated potassium channel beta subunits, hKCNMB3 and hKCNMB4, that are enriched in the testis and brain, respectively. We compare and contrast the steady state and kinetic properties of these beta subunits with the previously cloned mouse beta1 (mKCNMB1) and the human beta2 subunit (hKCNMB2). Once inactivation is removed, we find that hKCNMB2 has properties similar to mKCNMB1. hKCNMB2 slows Hslo1 channel gating and shifts the current-voltage relationship to more negative potentials. hKCNMB3 and hKCNMB4 have distinct effects on slo currents not observed with mKCNMB1 and hKCNMB2. Although we found that hKCNMB3 does interact with Hslo channels, its effects on Hslo1 channel properties were slight, increasing Hslo1 activation rates. In contrast, hKCNMB4 slows Hslo1 gating kinetics, and modulates the apparent calcium sensitivity of Hslo1. We found that the different effects of the beta subunits on some Hslo1 channel properties are calcium-dependent. mKCNMB1 and hKCNMB2 slow activation at 1 microM but not at 10 microM free calcium concentrations. hKCNMB4 decreases Hslo1 channel openings at low calcium concentrations but increases channel openings at high calcium concentrations. These results suggest that beta subunits in diverse tissue types fine-tune slo channel properties to the needs of a particular cell.


Subject(s)
Potassium Channels, Calcium-Activated , Potassium Channels/genetics , Amino Acid Sequence , Brain/metabolism , Calcium/pharmacology , Cloning, Molecular , Humans , Ion Channel Gating , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channel beta Subunits , Large-Conductance Calcium-Activated Potassium Channels , Male , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Patch-Clamp Techniques , Potassium Channels/chemistry , RNA, Messenger/metabolism , Sequence Alignment , Testis/metabolism
13.
Circ Res ; 85(11): 1067-76, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10571538

ABSTRACT

Action potential duration is prolonged in many forms of heart disease, often as a result of reductions in Ca(2+)-independent transient outward K(+) currents (ie, I(to)). To examine the effects of a primary reduction in I(to) current in the heart, transgenic mice were generated that express a dominant-negative N-terminal fragment of the K(v)4.2 pore-forming potassium channel subunit under the control of the mouse alpha-myosin heavy chain promoter. Two of 6 founders died suddenly, and only 1 mouse successfully transmitted the transgene in mendelian fashion. Electrophysiological analysis at 2 to 4 weeks of age demonstrated that I(to) density was specifically reduced and action potential durations were prolonged in a subset of transgenic myocytes. The heterogeneous reduction in I(to) was accompanied by significant prolongation of monophasic action potentials. In vivo hemodynamic studies at this age revealed significant elevations in the mean arterial pressure, peak systolic ventricular pressures, and +/-dP/dt, indicative of enhanced contractility. Surprisingly, by 10 to 12 weeks of age, transgenic mice developed clinical and hemodynamic evidence of congestive heart failure. Failing transgenic hearts displayed molecular and cellular remodeling, with evidence of hypertrophy, chamber dilatation, and interstitial fibrosis, and individual myocytes showed sharp reductions in I(to) and I(K1) densities, action potential duration prolongation, and increased cell capacitance. Our results confirm that K(v)4.2 subunits contribute to I(to) in the mouse and demonstrate that manipulation of cardiac excitability may secondarily influence contractile performance.


Subject(s)
Myocardium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium/metabolism , Action Potentials , Animals , Cardiomegaly/complications , Cardiomegaly/genetics , Echocardiography , Electrocardiography , Genes, Dominant , Heart Failure/etiology , Heart Failure/genetics , Hemodynamics , Ion Transport , Mice , Mice, Inbred CBA , Mice, Transgenic , Myocardial Contraction , Myosin Heavy Chains/genetics , Phenotype , Potassium Channels/biosynthesis , Potassium Channels/deficiency , Promoter Regions, Genetic , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Shal Potassium Channels , Ventricular Remodeling
14.
Cell Physiol Biochem ; 9(1): 11-28, 1999.
Article in English | MEDLINE | ID: mdl-10352341

ABSTRACT

The aim of the present study was to compare the biophysical properties and Cd2+ sensitivity of Kv4.2 and Kv1.4 in Xenopus oocytes with those of native transient outward potassium currents in rat and rabbit ventricular myocytes. In Xenopus oocytes, Kv4.2 inactivated at hyperpolarized voltages (V(1/2)inact = -58.4 +/- 0.96 mV, n = 12) and recovered from inactivation rapidly (time constant = 224 +/- 23 ms, n = 3). Cd2+ induced large (approx. 30 mV with 500 microM Cd2+), concentration-dependent rightward shifts in Kv4.2 steady-state activation and inactivation. Kv1.4 inactivated over more depolarized voltages than Kv4.2 (V(1/2)inact = -49.3 +/- 1.4 mV, n = 12). Recovery from inactivation of Kv1.4 was dominated by a large slow component (time constant = 9,038 +/- 1,178 ms, n = 4). Cd2+ exerted only modest effects on Kv1.4 gating, with 500 microM Cd2+ shifting the voltage dependence of steady-state activation and inactivation by approximately 12 mV. We show that the biophysical properties and Cd2+ sensitivity of rat ventricular Ito resemble those of heterologously expressed Kv4.2. These findings support previous suggestions that Kv4.2 is an important molecular component of Ito in adult rat heart. In addition, our findings show that Ito in rabbit ventricular myocytes and Kv1.4-based currents in Xenopus oocytes share similar biophysical properties and sensitivity to Cd2+, suggesting that Kv1.4 may underlie Ito in rabbit ventricle. However, a number of discrepancies exist between the properties of native currents and their putative molecular counterparts, suggesting that additional proteins and/or modulatory factors may also play a role in determining the biophysical and pharmacological properties of these native currents.


Subject(s)
Cadmium/pharmacology , Heart/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , Cells, Cultured , Female , Heart/drug effects , Heart Ventricles , Kv1.4 Potassium Channel , Membrane Potentials , Myocardium/cytology , Oocytes/drug effects , Oocytes/physiology , Potassium Channels/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Shal Potassium Channels , Xenopus laevis
15.
Circ Res ; 84(11): 1302-9, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10364568

ABSTRACT

Heart failure is the leading cause of mortality in patients with transfusional iron (Fe) overload in which myocardial iron uptake ensues via a transferrin-independent process. We examined the ability of L-type Ca2+ channel modifiers to alter Fe2+ uptake by isolated rat hearts and ventricular myocytes. Perfusion of rat hearts with 100 nmol/L 59Fe2+ and 5 mmol/L ascorbate resulted in specific 59Fe2+ uptake of 20.4+/-1.9 ng of Fe per gram dry wt. Abolishing myocardial electrical excitability with 20 mmol/L KCl reduced specific 59Fe2+ uptake by 60+/-7% (P<0.01), which suggested that a component of myocardial Fe2+ uptake depends on membrane voltage. Accordingly, 59Fe2+ uptake was inhibited by 10 micromol/L nifedipine (45+/-12%, P<0.02) and 100 micromol/L Cd2+ (86+/-3%; P<0. 001) while being augmented by 100 nmol/L Bay K 8644 (61+/-18%, P<0. 01) or 100 nmol/L isoproterenol (40+/-12%, P<0.05). By contrast, uptake of 100 nmol/L ferric iron (59Fe3+) was significantly lower (1. 4+/-0.3 ng Fe per gram dry wt; P<0.001) compared with divalent iron. These data suggest that a component of Fe2+ uptake into heart occurs via the L-type Ca2+ channel in myocytes. To investigate this further, the effects of Fe2+ on cardiac myocyte L-type Ca2+ currents were measured. In the absence of Ca2+, noninactivating nitrendipine-sensitive Fe2+ currents were recorded with 15 mmol/L [Fe2+]o. Low concentrations of Fe2+ enhanced Ca2+ current amplitude and slowed inactivation rates, which was consistent with Fe2+ entry into the cell, whereas higher Fe2+ levels caused dose-dependent decreases in peak current. Fe3+ had no effect on current amplitude or decay. Combined, our data suggest that myocardial Fe2+ uptake occurs via L-type Ca2+ channels and that blockade of these channels might be useful in the treatment of patients with excessive serum iron levels.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Heart/drug effects , Iron/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , In Vitro Techniques , Isoproterenol/pharmacology , Male , Myocardium/metabolism , Nifedipine/pharmacology , Patch-Clamp Techniques , Permeability , Rats , Rats, Sprague-Dawley
16.
J Physiol ; 517 ( Pt 1): 229-45, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10226162

ABSTRACT

1. Cardiac hypertrophy and prolongation of the cardiac action potential are hallmark features of heart disease. We examined the molecular mechanisms and the functional consequences of this action potential prolongation on calcium handling in right ventricular myocytes obtained from rats 8 weeks following ligation of the left anterior descending coronary artery (post-myocardial infarction (MI) myocytes). 2. Compared with myocytes from sham-operated rats (sham myocytes), post-MI myocytes showed significant reductions in transient outward K+ current (Ito) density (sham 19.7 +/- 1.1 pA pF-1 versus post-MI 11.0 +/- 1.3 pA pF-1; means +/- s.e.m.), inward rectifier K+ current density (sham -13.7 +/- 0.6 pA pF-1 versus post-MI -10.3 +/- 0.9 pA pF-1) and resting membrane potential (sham -84.4 +/- 1.3 mV versus post-MI -74.1 +/- 2.6 mV). Depressed Ito amplitude correlated with significant reductions in Kv4.2 and Kv4.3 mRNA and Kv4.2 protein levels. Kv1.4 mRNA and protein levels were increased and coincided with the appearance of a slow component of recovery from inactivation for Ito. 3. In current-clamp recordings, post-MI myocytes showed a significant increase in [Ca2+]i transient amplitude compared with sham myocytes. Using voltage-clamp depolarizations, no intrinsic differences in Ca2+ handling by the sarcoplasmic reticulum or in L-type Ca2+ channel density (ICa,L) were detected between the groups. 4. Stimulation of post-MI myocytes with an action potential derived from a sham myocyte reduced the [Ca2+] transient amplitude to the sham level and vice versa. 5. The net Ca2+ influx per beat via ICa,L was increased about 2-fold in myocytes stimulated with post-MI action potentials compared with sham action potentials. 6. Our findings demonstrate that reductions in K+ channel expression in post-MI myocytes prolong action potential duration resulting in elevated Ca2+ influx and [Ca2+]i transients.


Subject(s)
Calcium/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Potassium Channels/metabolism , Action Potentials , Animals , Calcium Channels/metabolism , Disease Models, Animal , Down-Regulation , Heart Ventricles/metabolism , Intracellular Fluid/metabolism , Ion Transport , Male , Membrane Potentials , Myocardial Infarction/genetics , Patch-Clamp Techniques , Potassium Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sarcoplasmic Reticulum/metabolism
17.
Am J Physiol ; 276(5): H1599-607, 1999 05.
Article in English | MEDLINE | ID: mdl-10330244

ABSTRACT

The aim of the present study was to assess differences in transient outward potassium current (Ito) between the right ventricular free wall and the interventricular septum of the adult rat ventricle and to evaluate the relative contributions of Kv4.2, Kv4.3, and Kv1.4 to Ito in these regions. The results show that Ito is composed of both rapidly and slowly recovering components in the right wall and septum. The fast component had a significantly higher density in the right free wall than in the septum, whereas the slow component did not differ between the two sites. Kv4.2 mRNA and protein levels were also highest in the right wall and correlated with Ito density, whereas Kv4.3 was expressed uniformly in these regions. The kinetics of the rapidly recovering component of Ito in myocytes was similar to that recorded in tsa-201 cells expressing Kv4.2 and Kv4.3 channels. Kv1.4 mRNA and protein expression correlated well with the density of the slowly recovering Ito, whereas the recovery kinetics of the slow component were identical to Kv1.4 expressed in tsa-201 cells. In conclusion, expression of Kv1.4, Kv4.2, and Kv4.3 differs between regions in rat hearts. Regionally specific differences in the genetic composition of Ito can account for the region-specific properties of this current.


Subject(s)
Heart Septum/chemistry , Myocardium/chemistry , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Animals , Cell Line , DNA Primers , Gene Expression/physiology , Heart Ventricles/chemistry , Kv1.4 Potassium Channel , Patch-Clamp Techniques , Potassium Channels/analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Shal Potassium Channels , Ventricular Function
18.
Cardiovasc Res ; 37(2): 312-23, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9614488

ABSTRACT

Action potential prolongation is a common finding in human heart failure and in animal models of cardiac hypertrophy. The mechanism of action potential prolongation involves altered expression of a variety of depolarising and hyperpolarising currents in the myocardium. In particular, decreased density of the transient outward potassium current seems to play a prominent role, regardless of species, precipitating factors or the severity of hypertrophy. The decreased density of the transient outward current appears to be caused by reduced transcription of Kv4.2 and Kv4.3 and may be caused in part by an inhibitory effect of alpha-adrenoceptor stimulation. During the early stage of the disease process, action potential prolongation may increase the amplitude of the intracellular calcium transient, causing positive inotropy. We argue therefore, that action prolongation may be a compensatory response which may acutely support the compromised cardiac output. In severe hypertrophy and end-stage heart failure however, despite continued action potential prolongation, the amplitude of the calcium transient becomes severely reduced. The mechanism underlying this event appears to involve reduced expression of calcium handling proteins, and these late events may herald the onset of failure. At present the events leading to the late changes in calcium handling are poorly understood. However, chronic activation of compensatory mechanisms including action potential prolongation may trigger these late events. In the present article we outline a hypothesis which describes a potential role for action potential prolongation, and the associated elevation in the levels of intracellular calcium, in maladaptive gene expression and the progression toward cardiac failure.


Subject(s)
Action Potentials , Calcium/metabolism , Heart Failure/etiology , Myocardium/metabolism , Animals , Calcium Channels/metabolism , Cardiomegaly/metabolism , Heart Failure/metabolism , Humans , Models, Cardiovascular , Shal Potassium Channels
19.
J Physiol ; 504 ( Pt 2): 271-86, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9365903

ABSTRACT

1. In rat heart, three K+ channel genes that encode inactivating transient outward (ITO)-like currents are expressed. During development the predominant K+ channel mRNA species switches from Kv1.4 to Kv4.2 and Kv4.3. However, no functional correlate of this isoform switch has been reported. We investigated action potential characteristics and ITO in cultured neonatal rat ventricular myocytes and adult rat hearts. We further examined whether the changes in K+ channel gene expression and the associated electrophysiology that occurs during development could be induced by thyroid hormone. 2. In myocytes isolated from right ventricle of adult rat heart, action potential duration was short and independent of rate of stimulation. The density of ITO was 21.5 +/- 1.8 pA pF-1 (n = 21). Recovery from inactivation was best described by a single exponential (tau fast = 31.7 +/- 2.7 ms, n = 13). The current remaining at the end of a 500 ms pulse (ISUS) was 6.2 +/- 0.5 pA pF-1 (n = 19). 3. In contrast to adult cells, action potential duration was prolonged and was markedly rate dependent in cultured neonatal rat ventricular myocytes. The current density of ITO measured in cultured ventricular myocytes from 1- to 2-day-old rats was 10.1 +/- 1.5 pA pF-1 (n = 17). The recovery from inactivation for ITO was best described by the sum of two exponentials (tau fast = 64.3 +/- 8.8 ms, 54.4 +/- 10.2%; tau slow = 8216 +/- 2396 ms, 37.4 +/- 7.9%; n = 5). ISUS was 4.4 +/- 0.6 pA pF-1 (n = 17). Steady-state activation and inactivation were similar in adult and neonatal ventricular myocytes. 4. In neonatal myocytes treated with thyroid hormone, tri-iodothyronine (T3, 100 nM), action potential duration was abbreviated and independent of stimulation rate. Whilst T3 did not significantly increase ITO density (24.0 +/- 2.9 pA pF-1; n = 21 in T3 treated cells cf. 20.1 +/- 3.0 pA pF-1; n = 37 in untreated controls), the recovery from inactivation of ITO was accelerated (tau fast = 39.2 +/- 3.6 ms, 82.2 +/- 8.9%, n = 9). T3 did however, increase ISUS current density (4.7 +/- 0.77 pA pF-1; n = 37 and 7.0 +/- 0.7 pA pF-1, n = 21, in control and T3 treated cells, respectively. 5. The effects of T3 (100 nM) were associated with a marked decrease in the expression of Kv1.4 at the mRNA and protein level, and an increase in the expression of Kv4.3 without changes in Kv4.2 mRNA levels. 6. The findings of the present study indicate that postnatal development involves a shortening of action potential duration and an increase in the density of ITO. Furthermore, we show that development is also associated with a loss of action potential rate dependence, and an acceleration in the rate of recovery of ITO. We propose that these functional effects occur as a consequence of the previously reported developmental Kv1.4 to Kv4.2/Kv4.3 isoform switch. In cultured neonatal myocytes, T3 induced many of the electrophysiological and molecular changes that normally occur during postnatal development, suggesting that this hormone may play an important role in postnatal electrophysiological development.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Myocardium/metabolism , Potassium Channels/genetics , Potassium/metabolism , Triiodothyronine/pharmacology , Action Potentials/drug effects , Aging , Animals , Anti-Arrhythmia Agents/pharmacology , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Flecainide/pharmacology , Gene Expression Regulation, Developmental/genetics , Heart Ventricles/drug effects , Heart Ventricles/growth & development , Myocardium/cytology , Potassium Channels/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleases/metabolism
20.
Exp Physiol ; 82(1): 85-98, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9023508

ABSTRACT

The aim of the present experiment was to determine whether the potassium channel opener 2-(2,2-bis(difluoromethyl)-6-nitro-3,4-dihydro-2H-1, 4-benzoxazine-4-yl)pyridine-N-oxide (ZM260384) was capable of accelerating the decline in skeletal muscle function during restricted blood flow in vivo. Cats (3.0-4.5 kg body weight) were anaesthetized with alphaxalone-alphadalone and breathed spontaneously following tracheotomy. Isometric tension was measured in the extensor digitorum longus-anterior tibialis (EDL-TA) muscle group. Ischaemia was induced by perfusing the hindlimb with the animal's own blood at a rate of 12.5 ml min-1 using a roller pump and stimulating the common peroneal nerve to induce repetitive submaximal tetanic contractions in the EDL-TA. The number of stimulation voltage increments required each minute to maintain a constant level of submaximal mechanical output and the time to exhaustion were used as indices of the rate of tension decline. The rate of tension decline in the ischaemic EDL-TA in the presence of ZM260384 at 3 mg kg-1, a maximally hypotensive dose predicted to be within the dose range required to exert direct effects on skeletal muscle, was measured and compared with the rate of tension decline in the presence of ZM260384 at 0.03 mg kg-1, also maximally hypotensive dose but below the predicted dose range for skeletal muscle effects. The number of voltage increments per minute was 1.93 +/- 0.07 and 1.48 +/- 0.14 (P < 0.05) in the presence of 3 and 0.03 mg kg-1 ZM260384, respectively. Time to exhaustion was 17.5 +/- 4.2 and 7.2 +/- 0.8 min (P < 0.05) in the presence of 3 and 0.03 mg kg-1 ZM260384, respectively. Given that there was no difference between these two groups in any haemodynamic variable measured, the results of the present study suggest that ZM260384 (3 mg kg-1) increases the rate of isometric force loss in ischemic skeletal muscle in vivo.


Subject(s)
Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Oxazines/pharmacology , Potassium Channels/metabolism , Anesthesia , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Cats , Dose-Response Relationship, Drug , Hindlimb/blood supply , Isometric Contraction/drug effects , Isometric Contraction/physiology , Male , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Potassium Channels/drug effects , Regional Blood Flow/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...