Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Clin Pharmacol Ther ; 108(2): 201-211, 2020 08.
Article in English | MEDLINE | ID: mdl-32302411

ABSTRACT

Azithromycin (AZ) is a broad-spectrum macrolide antibiotic with a long half-life and a large volume of distribution. It is primarily used for the treatment of respiratory, enteric, and genitourinary bacterial infections. AZ is not approved for the treatment of viral infections, and there is no well-controlled, prospective, randomized clinical evidence to support AZ therapy in coronavirus disease 2019 (COVID-19). Nevertheless, there are anecdotal reports that some hospitals have begun to include AZ in combination with hydroxychloroquine or chloroquine (CQ) for treatment of COVID-19. It is essential that the clinical pharmacology (CP) characteristics of AZ be considered in planning and conducting clinical trials of AZ alone or in combination with other agents, to ensure safe study conduct and to increase the probability of achieving definitive answers regarding efficacy of AZ in the treatment of COVID-19. The safety profile of AZ used as an antibacterial agent is well established.1 This work assesses published in vitro and clinical evidence for AZ as an agent with antiviral properties. It also provides basic CP information relevant for planning and initiating COVID-19 clinical studies with AZ, summarizes safety data from healthy volunteer studies, and safety and efficacy data from phase II and phase II/III studies in patients with uncomplicated malaria, including a phase II/III study in pediatric patients following administration of AZ and CQ in combination. This paper may also serve to facilitate the consideration and use of a priori-defined control groups for future research.


Subject(s)
Antiviral Agents/pharmacology , Azithromycin/pharmacology , Azithromycin/therapeutic use , Betacoronavirus/drug effects , Antiviral Agents/therapeutic use , Azithromycin/adverse effects , Azithromycin/pharmacokinetics , Betacoronavirus/pathogenicity , Clinical Trials as Topic , Coronavirus Infections/drug therapy , Drug Therapy, Combination , Humans , Hydroxychloroquine/pharmacology , Lung/drug effects , Microbial Sensitivity Tests , SARS-CoV-2 , COVID-19 Drug Treatment
2.
J Pharmacol Toxicol Methods ; 61(2): 178-91, 2010.
Article in English | MEDLINE | ID: mdl-20172036

ABSTRACT

INTRODUCTION: Pharmacological inhibition of cardiac potassium channels encoded by hERG (human ether-à-go-go-related gene) is associated with QT interval prolongation and torsades de pointes arrhythmia. Electrophysiological assays of hERG channel inhibition are integral to the safety testing of novel drug candidates. This study was conducted to compare, for the high affinity hERG inhibitors dofetilide and cisapride, hERG blockade between action potential (AP) and conventional (step and step-ramp) screening waveforms. Furthermore, it evaluated dynamic (pulse-by-pulse) protocol-dependence of hERG channel inhibition by these drugs. METHODS: Whole-cell patch-clamp recordings were made at 37 degrees C from hERG-expressing HEK 293 cells. Half-maximal inhibitory concentrations (IC(50) values) for I(hERG) blockade were obtained using conventional voltage clamp and action potential clamp, using previously digitised ventricular and Purkinje fibre (PF) AP waveforms. RESULTS: A more marked variation in IC(50) values with different command waveforms was observed for cisapride (ranging from 7 to 72 nM) than for dofetilide (ranging from 4 to 15 nM), with higher IC(50)s obtained with AP than step or step-ramp commands. The two drugs differed little from one another in effects on voltage-dependent activation; however, I(hERG) blockade by each drug was initially voltage-dependent, but at steady-state was only voltage-dependent for cisapride. There was comparatively little difference between the two drugs in effects on I(hERG) availability or time constants of development of inactivation. Features of time-dependence of blockade and the use of protocols employing varying rest periods in drug or commands of alternating duration highlighted a pronounced ability of cisapride, but not dofetilide, to dissociate and reassociate from hERG on a pulse-by-pulse basis. DISCUSSION: Protocols described here that demonstrated dynamic variation (drug dissociation/reassociation) in hERG channel current blockade at 37 degrees C for cisapride may have future value for investigating drug interactions with the hERG channel. Downloadable digitised ventricular and PF AP waveforms that can be used in AP clamp experiments also accompany this article.


Subject(s)
Cisapride/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Phenethylamines/pharmacology , Potassium Channel Blockers/pharmacology , Sulfonamides/pharmacology , Action Potentials/drug effects , Axons/drug effects , Cell Line , Cisapride/metabolism , Data Interpretation, Statistical , Electrophysiology , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Patch-Clamp Techniques , Phenethylamines/metabolism , Potassium Channel Blockers/metabolism , Sulfonamides/metabolism , Temperature
3.
J Pharmacol Toxicol Methods ; 57(1): 9-22, 2008.
Article in English | MEDLINE | ID: mdl-17707659

ABSTRACT

INTRODUCTION: Prediction of the propensity of a compound to induce Torsades de Pointes continues to be a formidable challenge to the pharmaceutical industry. Development of an in vitro model for assessment of proarrhythmic potential offers the advantage of higher throughput and reduced compound quantity requirements when compared to in vivo studies. A rabbit isolated heart model (SCREENIT) has been reported to identify compounds with proarrhythmic potential based on the observance of compound-induced triangulation and instability of the monophasic action potential (MAP), ectopic beats, and reverse-use dependence of prolongation of the MAP duration. Previous reports have indicated that this model qualitatively identifies proarrhythmic compounds and suggest the use of this model to assign safety margins for human clinical use. The intent of this series of studies was to evaluate the impact of study design on the proarrhythmic concentration predicted by this model. METHODS: Nine compounds of varying proarrhythmic potential and a negative control were tested in a blinded fashion using a series of different experimental protocols: Compounds were tested at multiple concentration ranges and extended perfusion times were also evaluated. RESULTS: In general when the dataset is viewed as a whole, the model did identify proarrhythmic compounds, however the concentration at which action potential prolongation, triangulation, instability, reverse-use dependence and ectopic beats occurred often varied based on the concentration range selected. Further analysis using extended compound perfusion times demonstrated that variability may be due in part to lack of adequate equilibration of compound with the cardiac tissue. DISCUSSION: We report that the model correctly identified proarrhythmic agents in a qualitative manner, but that study design impacts the proarrhythmic concentration derived from the model.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiovascular Agents/adverse effects , Action Potentials/drug effects , Animals , Computer Simulation , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Electrophysiologic Techniques, Cardiac , Heart Conduction System/drug effects , Models, Biological , Predictive Value of Tests , Rabbits
4.
J Pharmacol Toxicol Methods ; 56(2): 194-202, 2007.
Article in English | MEDLINE | ID: mdl-17583537

ABSTRACT

INTRODUCTION: Purkinje fibre repolarisation assays are valuable tools for identifying compounds which affect cardiac ion channels. The throughput of compound testing in this assay is low therefore we designed a novel recording system to improve screening and animal tissue usage efficiencies. METHODS: The system was used to evaluate compounds using standard sharp microelectrode techniques. Animal tissue usage efficiencies were quantified by adding up the total number of Purkinje fibres from which recordings were attempted and dividing this by the number of experimental data sets generated, to arrive at a 'fibres per data set' ratio. Test compounds were dofetilide (3 x 10(-10) to 10(-8) M), cisapride (10(-8) to 3 x 10(-7) M), terodiline (10(-6) to 3 x 10(-5) M) and verapamil (3 x 10(-7) to 10(-5) M). RESULTS: Using the novel modified system, 21 data sets were generated from 29 fibres, compared to 24 data sets from 41 fibres using the conventional manual recording system, demonstrating a 24% improvement in the efficiency of animal tissue usage. Comparing data from the manual and modified systems revealed differences in absolute values for all parameters including APD90 (308.73 +/- 9.97 ms, n = 24, compared to 275.27 +/- 8.25 ms, n = 21, respectively; P < 0.05). Differences in the magnitude of changes in action potential parameters between the systems were also evident for all compounds including terodiline (1 x 10(-5) M) which caused a -27.1 +/- 16.5% reduction in APD50 in the manual system, compared to a - 55.2 +/- 2.2% reduction in the modified system. DISCUSSION: Although the value of the present study is limited by the small sample sizes, it has demonstrated utility of the modified system in improving efficiency of animal tissue usage. It offers potential utility in a higher throughput screening environment for examining the electrophysiological properties of novel compounds in native cardiac tissues, particularly where functional patch clamp data are limited.


Subject(s)
Electrophysiologic Techniques, Cardiac/methods , Heart/drug effects , Ion Channels/antagonists & inhibitors , Purkinje Fibers/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Butylamines/pharmacology , Calcium Channel Blockers/pharmacology , Cisapride/pharmacology , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Electric Stimulation , Electrophysiologic Techniques, Cardiac/instrumentation , Female , Heart/physiology , In Vitro Techniques , Ion Channels/physiology , Long QT Syndrome/diagnosis , Long QT Syndrome/physiopathology , Male , Microelectrodes , Phenethylamines/pharmacology , Purkinje Fibers/physiology , Reproducibility of Results , Sulfonamides/pharmacology , Verapamil/pharmacology
5.
Biochem Biophys Res Commun ; 351(1): 273-80, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17056009

ABSTRACT

The phenothiazine antipsychotic agent thioridazine has been linked with prolongation of the QT interval on the electrocardiogram, ventricular arrhythmias, and sudden death. Although thioridazine is known to inhibit cardiac hERG K(+) channels there is little mechanistic information on this action. We have investigated in detail hERG K(+) channel current (I(hERG)) blockade by thioridazine and identified a key molecular determinant of blockade. Whole-cell I(hERG) measurements were made at 37 degrees C from human embryonic kidney (HEK-293) cells expressing wild-type and mutant hERG channels. Thioridazine inhibited I(hERG) tails at -40mV following a 2s depolarization to +20mV with an IC(50) value of 80nM. Comparable levels of I(hERG) inhibition were seen with physiological command waveforms (ventricular and Purkinje fibre action potentials). Thioridazine block of I(hERG) was only weakly voltage-dependent, though the time dependence of I(hERG) inhibition indicated contingency of blockade upon channel gating. The S6 helix point mutation F656A almost completely abolished, and the Y652A mutation partially attenuated, I(hERG) inhibition by thioridazine. In summary, thioridazine is one of the most potent hERG K(+) channel blockers amongst antipsychotics, exhibiting characteristics of a preferential open/activated channel blocker and binding at a high affinity site in the hERG channel pore.


Subject(s)
Chlorpromazine/administration & dosage , Ether-A-Go-Go Potassium Channels/physiology , Ion Channel Gating/physiology , Kidney/physiology , Membrane Potentials/physiology , Antipsychotic Agents/administration & dosage , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/chemistry , Humans , Kidney/drug effects , Membrane Potentials/drug effects , Protein Structure, Tertiary , Structure-Activity Relationship
6.
J Membr Biol ; 211(1): 43-53, 2006 May.
Article in English | MEDLINE | ID: mdl-16988861

ABSTRACT

The regulation of ion channels and transporters by anionic phospholipids is currently very topical. G protein-gated K(+) channels from the Kir3.0 family are involved in slowing the heart rate, generating late inhibitory postsynaptic potentials and controlling hormone release from neuroendocrine cells. There is considerable functional precedent for the control of these channels by phosphatidylinositol 4,5-bisphosphate. In this study, we used a biochemical assay to investigate the lipid binding properties of Kir3.0 channel domains. We reveal a differential binding affinity to a range of phosphoinositides between the C termini of the Kir3.0 isoforms. Furthermore, the N terminus in addition to the C terminus of Kir3.4 is necessary to observe binding and is decreased by the mutations R72A, K195A and R196A but not K194A. Protein kinase C phosphorylation of the Kir3.1 C-terminal fusion protein decreases anionic phospholipid binding. The differential binding affinity has functional consequences as the inhibition of homomeric Kir3.1, occurring after M3 receptor activation, recovers over minutes while homomeric Kir3.2 does not.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Phosphatidylinositols/metabolism , Amino Acid Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Humans , Maltose-Binding Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding/physiology , Protein Kinase C/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
7.
Biochem Biophys Res Commun ; 341(2): 500-6, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16446155

ABSTRACT

The HERG potassium channel might have a non-canonical drug binding site, distinct from the channel's inner cavity, that could be responsible for elements of closed-state pharmacological inhibition of the channel. The macrolide antibiotic erythromycin is a drug that may block unconventionally because of its size. Here we used whole-cell patch-clamp recording at 37 degrees C from heterologously expressed HERG channels in a mammalian cell line to show that erythromycin either produces a rapid open-state-dependent HERG channel inhibition, or components of both open-state-dependent and closed-state-dependent inhibition. Alanine-substitution of HERG's canonical determinants of blockade revealed that Y652 was not important as a molecular determinant of blockade, and that mutation of F656 resulted in only weak attenuation of inhibition. In computer models of the channel, erythromycin could make several direct contacts with F656, but not with Y652, in the open-state model, and erythromycin was unable to fit into a closed-state channel model.


Subject(s)
Erythromycin/pharmacology , Ether-A-Go-Go Potassium Channels/chemistry , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Inhibitory Concentration 50 , Models, Biological , Models, Chemical , Models, Molecular , Mutation , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/chemistry , Protein Conformation , Protein Synthesis Inhibitors/pharmacology , Software , Temperature , Time Factors
8.
Br J Pharmacol ; 147(8): 905-16, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16474415

ABSTRACT

The fluoroquinolone antibiotic moxifloxacin has been associated with the acquired long QT syndrome and is used as a positive control in the evaluation of the QT-interval prolonging potential of new drugs. In common with other QT-prolonging agents, moxifloxacin is known to inhibit the hERG potassium K+ channel, but at present there is little mechanistic information available on this action. This study was conducted in order to characterise the inhibition of hERG current (I(hERG)) by moxifloxacin, and to determine the role in drug binding of the S6 aromatic amino-acid residues Tyr652 and Phe656. hERG currents were studied using whole-cell patch clamp (at room temperature and at 35-37 degrees C) in an HEK293 cell line stably expressing hERG channels. Moxifloxacin reversibly inhibited currents in a dose-dependent manner. We investigated the effects of different voltage commands to elicit hERG currents on moxifloxacin potency. Using a 'step-ramp' protocol, the IC50 was 65 microM at room temperature and 29 microM at 35 degrees C. When a ventricular action potential waveform was used to elicit currents, the IC50 was 114 microM. Block of hERG by moxifloxacin was found to be voltage-dependent, occurred rapidly and was independent of stimulation frequency. Mutagenesis of the S6 helix residue Phe656 to Ala failed to eliminate or reduce the moxifloxacin-mediated block whereas mutation of Tyr652 to Ala reduced moxifloxacin block by approximately 66%. Our data demonstrate that moxifloxacin blocks the hERG channel with a preference for the activated channel state. The Tyr652 but not Phe656 S6 residue is involved in moxifloxacin block of hERG, concordant with an interaction in the channel inner cavity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aza Compounds/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Quinolines/pharmacology , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Fluoroquinolones , Humans , Moxifloxacin , Ofloxacin/pharmacology
9.
Mol Pharmacol ; 69(2): 509-19, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16291873

ABSTRACT

Block of human ether-a-go-go related gene (hERG) K(+) channels by otherwise useful drugs is the most common cause of long QT syndrome, a disorder of cardiac repolarization that predisposes patients to potentially fatal arrhythmias. This undesirable long QT side effect has been a major reason for the withdrawal of medications from the pharmaceutical market. Understanding the molecular basis of hERG block is therefore essential to facilitate the design of safe drugs. Binding sites for hERG blockers have been mapped within the inner cavity of the channel and include aromatic residues in the S6 helix (Tyr-652, Phe-656) and residues in the pore helix (Thr-623, Ser-624, Val-625). We used mutagenesis of these residues, combined with an investigation of hERG block by close analogs of clofilium and ibutilide, to assess how specific alterations in drug structure affected potency and binding interactions. Although changing the basic nitrogen from quaternary to tertiary accelerated the onset of block, the IC(50) and kinetics for recovery from block were similar. In contrast, analogs with different para-substituents on the phenyl ring had significantly different potencies for wild-type hERG block. The highest potency was achieved with polar or electronegative para-substituents, whereas neutral para-substituents had potencies more than 100-fold lower. Results from mutagenesis and molecular modeling studies suggest that phenyl ring para-substituents influence drug interactions with Thr-623, Ser-624, and Tyr-652 and strongly affect binding affinity. Together, these findings suggest that modifying the para-substituent could be a useful strategy for reducing hERG potency and increasing the safety margin of compounds in development.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Drug Design , Ether-A-Go-Go Potassium Channels/chemistry , Quaternary Ammonium Compounds/chemistry , Sulfonamides/chemistry , Amino Acid Motifs , Amino Acids/chemistry , Amino Acids/genetics , Animals , Anti-Arrhythmia Agents/pharmacology , Binding Sites , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Humans , Models, Molecular , Molecular Structure , Mutagenesis , Oocytes/drug effects , Protein Conformation , Quaternary Ammonium Compounds/pharmacology , Structure-Activity Relationship , Sulfonamides/pharmacology , Xenopus laevis
10.
Am J Physiol Cell Physiol ; 289(3): C543-56, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15857907

ABSTRACT

G protein-gated inward rectifier (Kir3) channels are inhibited by activation of G(q/11)-coupled receptors and this has been postulated to involve the signaling molecules protein kinase C (PKC) and/or phosphatidylinositol 4,5-bisphosphate (PIP(2)). Their precise roles in mediating the inhibition of this family of channels remain controversial. We examine here their relative roles in causing inhibition of Kir3.1/3.2 channels stably expressed in human embryonic kidney (HEK)-293 cells after muscarinic M(3) receptor activation. In perforated patch mode, staurosporine prevented the G(q/11)-mediated, M(3) receptor, inhibition of channel activity. Recovery from M(3)-mediated inhibition was wortmannin sensitive. Whole cell currents, where the patch pipette was supplemented with PIP(2), were still irreversibly inhibited by M(3) receptor stimulation. When adenosine A(1) receptors were co-expressed, inclusion of PIP(2) rescued the A(1)-mediated response. Recordings from inside-out patches showed that catalytically active PKC applied directly to the intracellular membrane face inhibited the channels: a reversible effect modulated by okadaic acid. Generation of mutant heteromeric channel Kir3.1S185A/Kir3.2C-S178A, still left the channel susceptible to receptor, pharmacological, and direct kinase-mediated inhibition. Biochemically, labeled phosphate is incorporated into the channel. We suggest that PKC-delta mediates channel inhibition because recombinant PKC-delta inhibited channel activity, M(3)-mediated inhibition of the channel, was counteracted by overexpression of two types of dominant negative PKC-delta constructs, and, by using confocal microscopy, we have demonstrated translocation of green fluorescent protein-tagged PKC-delta to the plasma membrane on M(3) receptor stimulation. Thus Kir3.1/3.2 channels are sensitive to changes in membrane phospholipid levels but this is contingent on the activity of PKC-delta after M(3) receptor activation in HEK-293 cells.


Subject(s)
Potassium Channels, Inwardly Rectifying/physiology , Protein Kinase C/metabolism , Receptor Cross-Talk/physiology , Receptor, Muscarinic M3/physiology , Cell Line , Cell Membrane , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Kidney/cytology , Membrane Potentials/physiology , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Protein Kinase C-delta , Protein Kinase C-epsilon
11.
Am J Physiol Cell Physiol ; 287(1): C182-91, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15013952

ABSTRACT

Activation of G protein-gated inwardly rectifying K(+) (GIRK) channels, found in the brain, heart, and endocrine tissue, leads to membrane hyperpolarization that generates neuronal inhibitory postsynaptic potentials, slows the heart rate, and inhibits hormone release. During stimulation of G(i/o)-coupled receptors and subsequent channel activation, it has been observed that the current desensitizes. In this study we examined mechanisms underlying fast desensitization of cloned heteromeric neuronal Kir3.1+3.2A and atrial Kir3.1+3.4 channels and also homomeric Kir3.0 currents in response to stimulation of several G(i/o) G protein-coupled receptors (GPCRs) expressed in HEK-293 cells (adenosine A(1), adrenergic alpha(2A), dopamine D(2S), M(4) muscarinic, and GABA(B1b/2) receptors). We found that all agonist-induced currents displayed a similar degree of desensitization except the adenosine A(1) receptor, which exhibits an additional desensitizing component. Using the nonhydrolyzable GTP analog guanosine 5'-O-(3-thiotriphosphate) (GTPgammaS), we found that this is due to a receptor-dependent, G protein-independent process. Using Ca(2+) imaging we showed that desensitization is unlikely to be accounted for solely by phospholipase C activation and phosphatidylinositol 4,5-bisphosphate (PIP(2)) hydrolysis. We examined the contribution of the G protein cycle and found the following. First, agonist concentration is strongly correlated with degree of desensitization. Second, competitive inhibition of GDP/GTP exchange by using nonhydrolyzable guanosine 5'-O-(2-thiodiphosphate) (GDPbetaS) has two effects, a slowing of channel activation and an attenuation of the fast desensitization phenomenon. Finally, using specific Galpha subunits we showed that ternary complexes with fast activation rates display more prominent desensitization than those with slower activation kinetics. Together our data suggest that fast desensitization of GIRK currents is accounted for by the fundamental properties of the G protein cycle.


Subject(s)
GTP-Binding Proteins/metabolism , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/metabolism , Cell Line , Electric Conductivity , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Protein Isoforms/metabolism , Receptors, Cell Surface/metabolism
12.
Eur J Neurosci ; 18(8): 2110-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14622172

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are found in neurons, atrial myocytes and neuroendocrine cells. A characteristic feature is their activation by stimulation of Gi/o-coupled receptors. In central neurons, for example, they are activated by adenosine and GABA and, as such, they play an important role in neurotransmitter-mediated regulation of membrane excitability. The channels are tetrameric assemblies of Kir3.x subunits (Kir3.1-3.4 plus splice variants). In this study I have attempted to identify the channel subunits which contribute to the native GIRK current recorded from primary cultured rat hippocampal pyramidal neurons. Reverse transcriptase-polymerase chain reaction revealed the expression of mRNA for Kir3.1, 3.2A, 3.2C and 3.3 subunits and confocal immunofluorescence microscopy was used to investigate their expression patterns. Diffuse staining was observed on both cell somata and dendrites for Kir3.1 and Kir3.2A yet that for Kir3.2C was weaker and punctate. Whole-cell patch clamp recordings were used to record GIRK currents from hippocampal pyramidal neurons which were identified on the basis of inward rectification, dependence of reversal potential on external potassium concentration and sensitivity to tertiapin. The GIRK currents were enhanced by the stimulation of a number of Gi/o-coupled receptors and were inhibited by pertussis toxin. In order to ascertain which Kir3.x subunits were responsible for the native GIRK current I compared the properties with those of the cloned Kir3.1 + 3.2A and Kir3.1 + 3.2C channels heterologously expressed in HEK293 cells.


Subject(s)
GTP-Binding Proteins/physiology , Hippocampus/cytology , Neurons/physiology , Potassium Channels, Inwardly Rectifying/physiology , Potassium Channels/physiology , Adenosine/pharmacology , Analgesics/pharmacology , Animals , Animals, Newborn , Baclofen/pharmacology , Benzoxazines , Blotting, Northern , Calcium Channel Blockers/pharmacology , Carbachol/pharmacology , Cell Cycle Proteins/metabolism , Cells, Cultured , Cholinergic Agonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Free Radical Scavengers/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GABA Agonists/pharmacology , Hormones/pharmacology , Humans , Kidney , Membrane Potentials/drug effects , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Patch-Clamp Techniques/methods , Pertussis Toxin/pharmacology , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/classification , Protein Subunits/physiology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Serotonin/pharmacology , Somatostatin/pharmacology , Time Factors
13.
Proc Natl Acad Sci U S A ; 100(10): 6239-44, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12719528

ABSTRACT

G protein-gated inwardly rectifying K(+) (Kir) channels are found in neurones, atrial myocytes, and endocrine cells and are involved in generating late inhibitory postsynaptic potentials, slowing the heart rate and inhibiting hormone release. They are activated by G protein-coupled receptors (GPCRs) via the inhibitory family of G protein, G(i/o), in a membrane-delimited fashion by the direct binding of Gbetagamma dimers to the channel complex. In this study we are concerned with the kinetics of deactivation of the cloned neuronal G protein-gated K(+) channel, Kir3.1 + 3.2A, after stimulation of a number of GPCRs. Termination of the channel activity on agonist removal is thought to solely depend on the intrinsic hydrolysis rate of the G protein alpha subunit. In this study we present data that illustrate a more complex behavior. We hypothesize that there are two processes that account for channel deactivation: agonist unbinding from the GPCR and GTP hydrolysis by the G protein alpha subunit. With some combinations of agonist/GPCR, the rate of agonist unbinding is slow and rate-limiting, and deactivation kinetics are not modulated by regulators of G protein-signaling proteins. In another group, channel deactivation is generally faster and limited by the hydrolysis rate of the G protein alpha subunit. G protein isoform and interaction with G protein-signaling proteins play a significant role with this group of GPCRs.


Subject(s)
GTP-Binding Proteins/metabolism , Ion Channel Gating/physiology , Neurons/physiology , Potassium Channels/agonists , Potassium Channels/physiology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Guanosine Triphosphate/metabolism , Humans , Kinetics , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels, Inwardly Rectifying/agonists , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , Transfection , gamma-Aminobutyric Acid/pharmacology
14.
J Biol Chem ; 278(12): 10851-8, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12529316

ABSTRACT

Traditionally the consequences of activation of G-protein-coupled receptors (GPCRs) by an agonist are studied using biochemical assays. In this study we use live cells and take advantage of a G-protein-gated inwardly rectifying potassium channel (Kir3.1+3.2A) that is activated by the direct binding of Gbetagamma subunit to the channel complex to report, in real-time, using the patch clamp technique the activity of the "ternary complex" of agonist/receptor/G-protein. This analysis is further facilitated by the use of pertussis toxin-resistant fluorescent and non-fluorescent Galpha(i/o) subunits and a series of HEK293 cell lines stably expressing both channel and receptors (including the adenosine A(1) receptor, the adrenergic alpha(2A) receptor, the dopamine D(2S) receptor, the M4 muscarinic receptor, and the dimeric GABA-B(1b/2) receptor). We systematically analyzed the contribution of the various inputs to the observed kinetic response of channel activation. Our studies indicate that the combination of agonist, GPCR, and G-protein isoform uniquely specify the behavior of these channels and thus support the importance of the whole ternary complex at a kinetic level.


Subject(s)
Biosensing Techniques , Heterotrimeric GTP-Binding Proteins/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , Ion Channel Gating , Kinetics , Pertussis Toxin/pharmacology , Potassium Channels/chemistry , Receptors, Cell Surface/metabolism
15.
J Biol Chem ; 277(32): 28803-9, 2002 Aug 09.
Article in English | MEDLINE | ID: mdl-12048213

ABSTRACT

Signaling studies in living cells would be greatly facilitated by the development of functional fluorescently tagged G-protein alpha subunits. We have designed G(i/o)alpha subunits fused to the cyan fluorescent protein and assayed their function by studying the following two signal transduction pathways: the regulation of G-protein-gated inwardly rectifying K(+) channels (Kir3.0 family) and adenylate cyclase. Palmitoylation and myristoylation consensus sites were removed from G(i/o) alpha subunits (G(i1)alpha, G(i2)alpha, G(i3)alpha, and G(oA)alpha) and a mutation introduced at Cys(-4) rendering the subunit resistant to pertussis toxin. This construct was fused in-frame with cyan fluorescent protein containing a short peptide motif from GAP43 that directs palmitoylation and thus membrane targeting. Western blotting confirmed G(i/o)alpha protein expression. Confocal microscopy and biochemical fractionation studies revealed membrane localization. Each mutant G(i/o) alpha subunit significantly reduced basal current density when transiently expressed in a stable cell line expressing Kir3.1 and Kir3.2A, consistent with the sequestration of the Gbetagamma dimer by the mutant Galpha subunit. Moreover, each subunit was able to support A1-mediated and D2S-mediated channel activation when transiently expressed in pertussis toxin-treated cells. Overexpression of tagged G(i3)alpha and G(oA)alpha alpha subunits reduced receptor-mediated and forskolin-induced cAMP mobilization.


Subject(s)
GTP-Binding Proteins/metabolism , Microscopy, Fluorescence/methods , Adenylate Cyclase Toxin , Adenylyl Cyclases/metabolism , Binding Sites , Blotting, Western , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , DNA, Complementary/metabolism , Dimerization , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , Microscopy, Confocal , Mutation , Myristic Acid/metabolism , Open Reading Frames , Palmitic Acid/metabolism , Pertussis Toxin , Potassium/metabolism , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Transfection , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...