Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 98(2): 143-155, 2020 08.
Article in English | MEDLINE | ID: mdl-32616523

ABSTRACT

The two-pore domain potassium channel (K2P-channel) THIK-1 has several predicted protein kinase A (PKA) phosphorylation sites. In trying to elucidate whether THIK-1 is regulated via PKA, we expressed THIK-1 channels in a mammalian cell line (CHO cells) and used the phosphodiesterase inhibitor 3-isobutyl-1-methyl-xanthine (IBMX) as a pharmacological tool to induce activation of PKA. Using the whole-cell patch-clamp recording, we found that THIK-1 currents were inhibited by application of IBMX with an IC50 of 120 µM. Surprisingly, intracellular application of IBMX or of the second messenger cAMP via the patch pipette had no effect on THIK-1 currents. In contrast, extracellular application of IBMX produced a rapid and reversible inhibition of THIK-1. In patch-clamp experiments with outside-out patches, THIK-1 currents were also inhibited by extracellular application of IBMX. Expression of THIK-1 channels in Xenopus oocytes was used to compare wild-type channels with mutated channels. Mutation of the putative PKA phosphorylation sites did not change the inhibitory effect of IBMX on THIK-1 currents. Mutational analysis of all residues of the (extracellular) helical cap of THIK-1 showed that mutation of the arginine residue at position 92, which is in the linker between cap helix 2 and pore helix 1, markedly reduced the inhibitory effect of IBMX. This flexible linker region, which is unique for each K2P-channel subtype, may be a possible target of channel-specific blockers. SIGNIFICANCE STATEMENT: The potassium channel THIK-1 is strongly expressed in the central nervous system. We studied the effect of 3-isobutyl-1-methyl-xanthine (IBMX) on THIK-1 currents. IBMX inhibits breakdown of cAMP and thus activates protein kinase A (PKA). Surprisingly, THIK-1 current was inhibited when IBMX was applied from the extracellular side of the membrane, but not from the intracellular side. Our results suggest that IBMX binds directly to the channel and that the inhibition of THIK-1 current was not related to activation of PKA.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Arginine/genetics , Binding Sites/drug effects , CHO Cells , Cricetulus , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mutation , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Rats , Xenopus
2.
Pflugers Arch ; 467(5): 867-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25791628

ABSTRACT

Over the last decade, we have seen an enormous increase in the number of experimental studies on two-pore-domain potassium channels (K2P-channels). The collection of reviews and original articles compiled for this special issue of Pflügers Archiv aims to give an up-to-date summary of what is known about the physiology and pathophysiology of K2P-channels. This introductory overview briefly describes the structure of K2P-channels and their function in different organs. Its main aim is to provide some background information for the 19 reviews and original articles of this special issue of Pflügers Archiv. It is not intended to be a comprehensive review; instead, this introductory overview focuses on some unresolved questions and controversial issues, such as: Do K2P-channels display voltage-dependent gating? Do K2P-channels contribute to the generation of action potentials? What is the functional role of alternative translation initiation? Do K2P-channels have one or two or more gates? We come to the conclusion that we are just beginning to understand the extremely complex regulation of these fascinating channels, which are often inadequately described as 'leak channels'.


Subject(s)
Action Potentials/physiology , Cell Physiological Phenomena/physiology , Neurons/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Animals , Humans , Ion Channel Gating/physiology , Potassium Channels, Tandem Pore Domain/chemistry
4.
Pflugers Arch ; 467(5): 1105-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25559843

ABSTRACT

The intracellular transport of membrane proteins is controlled by trafficking signals: Short peptide motifs that mediate the contact with COPI, COPII or various clathrin-associated coat proteins. In addition, many membrane proteins interact with accessory proteins that are involved in the sorting of these proteins to different intracellular compartments. In the K2P channels, TASK-1 and TASK-3, the influence of protein-protein interactions on sorting decisions has been studied in some detail. Both TASK paralogues interact with the adaptor protein 14-3-3; TASK-1 interacts, in addition, with the adaptor protein p11 (S100A10) and the endosomal SNARE protein syntaxin-8. The role of these interacting proteins in controlling the intracellular traffic of the channels and the underlying molecular mechanisms are summarised in this review. In the case of 14-3-3, the interacting protein masks a retention signal in the C-terminus of the channel; in the case of p11, the interacting protein carries a retention signal that localises the channel to the endoplasmic reticulum; and in the case of syntaxin-8, the interacting protein carries an endocytosis signal that complements an endocytosis signal of the channel. These examples illustrate some of the mechanisms by which interacting proteins may determine the itinerary of a membrane protein within a cell and suggest that the intracellular traffic of membrane proteins may be adapted to the specific functions of that protein by multiple protein-protein interactions.


Subject(s)
Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Protein Transport/physiology , Animals , Endocytosis/physiology , Humans
5.
Pflugers Arch ; 467(5): 1069-79, 2015 May.
Article in English | MEDLINE | ID: mdl-25539776

ABSTRACT

We studied the potassium current flowing through TREK-1 channels in rat cardiac ventricular myocytes. We separated the TREK-1 current from other current components by blocking most other channels with a blocker cocktail. We tried to inhibit the TREK-1 current by activating protein kinase A (PKA) with a mixture of forskolin and isobutyl-methylxanthine (IBMX). Activation of PKA blocked an outwardly rectifying current component at membrane potentials positive to -40 mV. At 37 °C, application of forskolin plus IBMX reduced the steady-state outward current measured at positive voltages by about 52 %. Application of the potassium channel blockers quinidine or tetrahexylammonium also reduced the steady-state outward current by about 50 %. Taken together, our results suggest that the increase in temperature from 22 to 37 °C increased the TREK-1 current by a factor of at least 5 and that the average density of the TREK-1 current in rat cardiomyocytes at 37 °C is about 1.5 pA/pF at +30 mV. The contribution of TREK-1 to the action potential was assessed by using a dynamic patch clamp technique. After subtraction of simulated TREK-1 currents, action potential duration at 50 or 90 % repolarisation was increased by about 12 %, indicating that TREK-1 may be functionally important in rat ventricular muscle. During sympathetic stimulation, inhibition of TREK-1 channels via PKA is expected to prolong the action potential primarily in subendocardial myocytes; this may decrease the transmural dispersion of repolarisation and thus may serve to prevent the occurrence of arrhythmias.


Subject(s)
Membrane Potentials/drug effects , Myocytes, Cardiac/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Membrane Potentials/physiology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques/methods , Phosphodiesterase Inhibitors/pharmacology , Rats
6.
Mol Biol Cell ; 25(12): 1877-91, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24743596

ABSTRACT

The endosomal SNARE protein syntaxin-8 interacts with the acid-sensitive potassium channel TASK-1. The functional relevance of this interaction was studied by heterologous expression of these proteins (and mutants thereof) in Xenopus oocytes and in mammalian cell lines. Coexpression of syntaxin-8 caused a fourfold reduction in TASK-1 current, a corresponding reduction in the expression of TASK-1 at the cell surface, and a marked increase in the rate of endocytosis of the channel. TASK-1 and syntaxin-8 colocalized in the early endosomal compartment, as indicated by the endosomal markers 2xFYVE and rab5. The stimulatory effect of the SNARE protein on the endocytosis of the channel was abolished when both an endocytosis signal in TASK-1 and an endocytosis signal in syntaxin-8 were mutated. A syntaxin-8 mutant that cannot assemble with other SNARE proteins had virtually the same effect as wild-type syntaxin-8. Total internal reflection fluorescence microscopy showed formation and endocytosis of vesicles containing fluorescence-tagged clathrin, TASK-1, and/or syntaxin-8. Our results suggest that the unassembled form of syntaxin-8 and the potassium channel TASK-1 are internalized via clathrin-mediated endocytosis in a cooperative manner. This implies that syntaxin-8 regulates the endocytosis of TASK-1. Our study supports the idea that endosomal SNARE proteins can have functions unrelated to membrane fusion.


Subject(s)
Endocytosis , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Qa-SNARE Proteins/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Endosomes/metabolism , Female , HeLa Cells , Humans , Nerve Tissue Proteins/chemistry , Potassium Channels, Tandem Pore Domain/chemistry , Protein Interaction Domains and Motifs , Qa-SNARE Proteins/chemistry , Xenopus laevis
7.
Pflugers Arch ; 466(8): 1559-70, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24196565

ABSTRACT

We have identified a novel splice variant of the human and rat two-pore domain potassium (K2P) channel TREK-1. The splice variant TREK-1e results from skipping of exon 5, which causes a frame shift in exon 6. The frame shift produces a novel C-terminal amino acid sequence and a premature termination of translation, which leads to a loss of transmembrane domains M3 and M4 and of the second pore domain. RT-PCR experiments revealed a preferential expression of TREK-1e in kidney, adrenal gland, and amygdala. TREK-1e was nonfunctional when expressed in Xenopus oocytes. However, both the surface expression and the current density of full-length TREK-1 were reduced by co-expression of TREK-1e. Live cell imaging in COS-7 cells transfected with GFP-tagged TREK-1e showed that this splice variant was retained in the endoplasmic reticulum (ER). Attachment of the C-terminus of TREK-1e to two different reporter proteins (Kir2.1 and CD8) led to a strong reduction in the surface expression of these fusion proteins. Progressive truncation of the C-terminus of TREK-1e in these reporter constructs revealed a critical region (amino acids 198 to 205) responsible for the intracellular retention. Mutagenesis experiments indicated that amino acids I204 and W205 are key residues mediating the ER retention of TREK-1e. Our results suggest that the TREK-1e splice variant may interfere with the vesicular traffic of full-length TREK-1 channels from the ER to the plasma membrane. Thus, TREK-1e might modulate the copy number of functional TREK-1 channels at the cell surface, providing a novel mechanism for fine tuning of TREK-1 currents.


Subject(s)
Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/genetics , Amino Acid Sequence , Animals , Blotting, Western , Gene Expression Regulation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Rats , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
8.
Pflugers Arch ; 466(9): 1735-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24297522

ABSTRACT

THIK-2 belongs to the 'silent' channels of the two-pore-domain potassium channel family. It is highly expressed in many nuclei of the brain but has so far resisted all attempts at functional expression. THIK-2 has a highly conserved 19-amino-acid region in its N terminus (residues 6-24 in the rat orthologue) that is missing in the closely related channel THIK-1. After deletion of this region (THIK-2(Δ6-24) mutant), functional expression of the channel was observed in Xenopus oocytes and in mammalian cell lines. The resulting potassium current showed outward rectification under physiological conditions and slight inward rectification with symmetrical high-K(+) solutions and could be inhibited by application of halothane or quinidine. Another THIK-2 mutant, in which the putative retention/retrieval signal RRR at positions 14-16 was replaced by AAA, produced a similar potassium current. Both mutants showed a distinct localisation to the surface membrane when tagged with green fluorescent protein and expressed in a mammalian cell line, whereas wild-type THIK-2 was mainly localised to the endoplasmic reticulum. These findings suggest that deletion of the retention/retrieval signal RRR enabled transport of THIK-2 channels to the surface membrane. Combining the mutation THIK-2(Δ6-24) with a mutation in the pore cavity (rat THIK-2(I158G)) gave rise to a 12-fold increase in current amplitude, most likely due to an increase in open probability. In conclusion, the characteristics of THIK-2 channels can be analysed in heterologous expression systems by using trafficking and/or gating mutants. The possible mechanisms that enable THIK-2 expression at the surface membrane in vivo remain to be determined.


Subject(s)
Potassium Channels, Tandem Pore Domain/metabolism , Protein Transport/physiology , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , HeLa Cells , Humans , Oocytes , Patch-Clamp Techniques , Rats , Transfection , Xenopus laevis
9.
Chem Biol ; 20(4): 583-93, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601647

ABSTRACT

Small-molecule stabilization of protein-protein interactions is an emerging field in chemical biology. We show how fusicoccanes, originally identified as fungal toxins acting on plants, promote the interaction of 14-3-3 proteins with the human potassium channel TASK-3 and present a semisynthetic fusicoccane derivative (FC-THF) that targets the 14-3-3 recognition motif (mode 3) in TASK-3. In the presence of FC-THF, the binding of 14-3-3 proteins to TASK-3 was increased 19-fold and protein crystallography provided the atomic details of the effects of FC-THF on this interaction. We also tested the functional effects of FC-THF on TASK channels heterologously expressed in Xenopus oocytes. Incubation with 10 µM FC-THF was found to promote the transport of TASK channels to the cell membrane, leading to a significantly higher density of channels at the surface membrane and increased potassium current.


Subject(s)
Diterpenes/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biological Transport , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Kinetics , Molecular Conformation , Molecular Sequence Data , Oocytes/metabolism , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Tertiary , Xenopus laevis/growth & development , Xenopus laevis/metabolism
10.
Cardiovasc Res ; 97(1): 97-105, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22977011

ABSTRACT

AIMS: The two-pore-domain potassium channel TASK-1 is robustly inhibited by the activation of receptors coupled to the Gα(q) subgroup of G-proteins, but the signal transduction pathway is still unclear. We have studied the mechanisms by which endothelin receptors inhibit the current carried by TASK-1 channels (I(TASK)) in cardiomyocytes. METHODS AND RESULTS: Patch-clamp measurements were carried out in isolated rat cardiomyocytes. I(TASK) was identified by extracellular acidification to pH 6.0 and by the application of the TASK-1 blockers A293 and A1899. Endothelin-1 completely inhibited I(TASK) with an EC(50) of <10 nM; this effect was mainly mediated by endothelin-A receptors. Application of 20 nM endothelin-1 caused a significant increase in action potential duration under control conditions; this was significantly reduced after pre-incubation of the cardiomyocytes with 200 nM A1899. The inhibition of I(TASK) by endothelin-1 was not affected by inhibitors of protein kinase C or rho kinase, but was strongly reduced by U73122, an inhibitor of phospholipase C (PLC). The ability of endothelin-1 to activate PLC-mediated signalling pathways was examined in mammalian cells transfected with TASK-1 and the endothelin-A receptor using patch-clamp measurements and total internal reflection microscopy. U73122 prevented the inhibition of I(TASK) by endothelin-1 and blocked PLC-mediated signalling, as verified with a fluorescent probe for phosphatidylinositol-(4,5)-bisphosphate hydrolysis. CONCLUSION: Our results show that I(TASK) in rat cardiomyocytes is controlled by endothelin-1 and suggest that the inhibition of TASK-1 via endothelin receptors is mediated by the activation of PLC. The prolongation of the action potential observed with 20 nM endothelin-1 was mainly due to the inhibition of I(TASK).


Subject(s)
Endothelin-1/pharmacology , Ion Channel Gating , Myocytes, Cardiac/enzymology , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Type C Phospholipases/metabolism , Action Potentials , Animals , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Microscopy, Fluorescence , Microscopy, Interference , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Rats , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Signal Transduction/drug effects , Transfection , Type C Phospholipases/antagonists & inhibitors
11.
FASEB J ; 26(2): 513-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22002906

ABSTRACT

Inward rectifier potassium channels of the Kir2 subfamily are important determinants of the electrical activity of brain and muscle cells. Genetic mutations in Kir2.1 associate with Andersen-Tawil syndrome (ATS), a familial disorder leading to stress-triggered periodic paralysis and ventricular arrhythmia. To identify the molecular mechanisms of this stress trigger, we analyze Kir channel function and localization electrophysiologically and by time-resolved confocal microscopy. Furthermore, we employ a mathematical model of muscular membrane potential. We identify a novel corticoid signaling pathway that, when activated by glucocorticoids, leads to enrichment of Kir2 channels in the plasma membranes of mammalian cell lines and isolated cardiac and skeletal muscle cells. We further demonstrate that activation of this pathway can either partly restore (40% of cases) or further impair (20% of cases) the function of mutant ATS channels, depending on the particular Kir2.1 mutation. This means that glucocorticoid treatment might either alleviate or deteriorate symptoms of ATS depending on the patient's individual Kir2.1 genotype. Thus, our findings provide a possible explanation for the contradictory effects of glucocorticoid treatment on symptoms in patients with ATS and may open new pathways for the design of personalized medicines in ATS therapy.


Subject(s)
Andersen Syndrome/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Andersen Syndrome/drug therapy , Andersen Syndrome/genetics , Animals , Female , Glucocorticoids/therapeutic use , Guinea Pigs , HEK293 Cells , HeLa Cells , Humans , Immediate-Early Proteins/metabolism , In Vitro Techniques , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myocytes, Cardiac/metabolism , Oocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Stress, Physiological , Xenopus laevis
12.
Cell Physiol Biochem ; 28(4): 613-24, 2011.
Article in English | MEDLINE | ID: mdl-22178873

ABSTRACT

BACKGROUND/AIMS: Atrial fibrillation is the most common arrhythmia in the elderly, and potassium channels with atrium-specific expression have been discussed as targets to treat atrial fibrillation. Our aim was to characterize TASK-1 channels in human heart and to functionally describe the role of the atrial whole cell current I(TASK-1). METHODS AND RESULTS: Using quantitative PCR, we show that TASK-1 is predominantly expressed in the atria, auricles and atrio-ventricular node of the human heart. Single channel recordings show the functional expression of TASK-1 in right human auricles. In addition, we describe for the first time the whole cell current carried by TASK-1 channels (I(TASK-1)) in human atrial tissue. We show that I(TASK-1) contributes to the sustained outward current I(Ksus) and that I(TASK-1) is a major component of the background conductance in human atrial cardiomyocytes. Using patch clamp recordings and mathematical modeling of action potentials, we demonstrate that modulation of I(TASK-1) can alter human atrial action potential duration. CONCLUSION: Due to the lack of ventricular expression and the ability to alter human atrial action potential duration, TASK-1 might be a drug target for the treatment of atrial fibrillation.


Subject(s)
Action Potentials/physiology , Myocytes, Cardiac/physiology , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Aged , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Cells, Cultured , Electrocardiography , Female , Heart Atria/metabolism , Humans , Male , Middle Aged , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/genetics , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/genetics , Xenopus
13.
Cell Physiol Biochem ; 28(4): 663-72, 2011.
Article in English | MEDLINE | ID: mdl-22178878

ABSTRACT

BACKGROUND/AIMS: ROMK channels mediate potassium secretion and regulate NaCl reabsorption in the kidney. The aim was to study the functional implications of the interaction between ROMK2 (Kir1.1b) and two glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and enolase-α, which were identified as potential regulatory subunits of the channel complex. METHODS: We performed a membrane yeast-two-hybrid screen of a human kidney cDNA library with ROMK2 as a bait. Interaction of ROMK2 with GAPDH and enolase was verified using GST pull-down, co-immunoprecipitation, immunohistochemistry and co-expression in Xenopus oocytes. RESULTS: Confocal imaging showed co-localisation of enolase and GAPDH with ROMK2 in the apical membrane of the renal epithelial cells of the thick ascending limb. Over-expression of GAPDH or enolase-α in Xenopus oocytes markedly reduced the amplitude of ROMK2 currents but did not affect the surface expression of the channels. Co-expression of the glycolytically inactive GAPDH mutant C149G did not have any effect on ROMK2 current amplitude. CONCLUSION: Our results suggest that the glycolytic enzymes GAPDH and enolase are part of the ROMK2 channel supramolecular complex and may serve to couple salt reabsorption in the thick ascending limb of the loop of Henle to the metabolic status of the renal epithelial cells.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Phosphopyruvate Hydratase/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Amino Acid Substitution , Animals , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/physiology , HEK293 Cells , Humans , Immunoprecipitation , Kidney/enzymology , Kidney/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/metabolism , Two-Hybrid System Techniques , Xenopus laevis/genetics
14.
Cell Physiol Biochem ; 28(1): 77-86, 2011.
Article in English | MEDLINE | ID: mdl-21865850

ABSTRACT

BACKGROUND/AIMS: The aim of the study was to characterize the whole cell current of the two-pore domain potassium channel TASK-1 (K2P3) in mouse ventricular cardiomyocytes (I(TASK-1)) and to analyze the cardiac phenotype of the TASK-1(-/-) mice. METHODS AND RESULTS: We have quantified the ventricular I(TASK-1) current using the blocker A293 and TASK-1(-/-) mice. Surface electrocardiogram recordings of TASK-1(-/-) mice showed a prolonged QTc interval and a broadened QRS complex. The differences in electrocardiograms between wild type and TASK-1(-/-) mice disappeared during sympathetic stimulation of the animals. Quantitative RT-PCR, patch clamp recordings and measurements of hemodynamic performance of TASK-1(-/-) mice revealed no major compensatory changes in ion channel transcription. Action potential recordings of TASK-1(-/-) mouse cardiomyocytes indicated that I(TASK-1) modulates action potential duration. Our in vivo electrophysiological studies showed that isoflurane, which activates TASK-1, slowed heart rate and atrioventricular conduction of wild-type but not of TASK-1(-/-) mice. CONCLUSION: The results of an invasive electrophysiological catheter protocol in combination with the observed QRS time prolongation in the surface electrocardiogram point towards a regulatory role of TASK-1 in the cardiac conduction system.


Subject(s)
Long QT Syndrome/etiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Sulfonamides/pharmacology , ortho-Aminobenzoates/pharmacology , Action Potentials/physiology , Anesthetics, Inhalation/pharmacology , Animals , Electrophysiological Phenomena/physiology , Heart Rate/drug effects , Hemodynamics/physiology , Isoflurane/pharmacology , Methoxamine/pharmacology , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Nerve Tissue Proteins/deficiency , Potassium Channels, Tandem Pore Domain/deficiency
15.
Physiology (Bethesda) ; 26(3): 181-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21670164

ABSTRACT

14-3-3 proteins regulate the function and subcellular sorting of membrane proteins. Often, 14-3-3 binding to client proteins requires phosphorylation of the client, but the relevant kinase is unknown in most cases. We summarize current progress in identifying kinases that target membrane proteins with 14-3-3 binding sites and discuss the molecular mechanisms of 14-3-3 action. One of the kinases involved is Akt/PKB, which has recently been shown to activate the 14-3-3-dependent switch in a number of client membrane proteins.


Subject(s)
14-3-3 Proteins/metabolism , Cell Membrane/metabolism , Signal Transduction , 14-3-3 Proteins/chemistry , Animals , Binding Sites , Humans , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism
16.
J Biol Chem ; 286(16): 13977-84, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21362619

ABSTRACT

Two-pore domain potassium (K(2P)) channels play a key role in setting the membrane potential of excitable cells. Despite their role as putative targets for drugs and general anesthetics, little is known about the structure and the drug binding site of K(2P) channels. We describe A1899 as a potent and highly selective blocker of the K(2P) channel TASK-1. As A1899 acts as an open-channel blocker and binds to residues forming the wall of the central cavity, the drug was used to further our understanding of the channel pore. Using alanine mutagenesis screens, we have identified residues in both pore loops, the M2 and M4 segments, and the halothane response element to form the drug binding site of TASK-1. Our experimental data were used to validate a K(2P) open-pore homology model of TASK-1, providing structural insights for future rational design of drugs targeting K(2P) channels.


Subject(s)
Benzamides/pharmacology , Benzeneacetamides/pharmacology , Nerve Tissue Proteins/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/chemistry , Potassium/chemistry , Alanine/chemistry , Animals , Benzamides/chemistry , Benzeneacetamides/chemistry , Binding Sites , DNA, Complementary/metabolism , Drug Design , Humans , Inhibitory Concentration 50 , Models, Molecular , Mutagenesis , Mutagenesis, Site-Directed , Oocytes/cytology , Patch-Clamp Techniques , Protein Conformation , Xenopus laevis
17.
Cell Calcium ; 48(1): 19-27, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20630587

ABSTRACT

STIM1 'senses' decreases in endoplasmic reticular (ER) luminal Ca(2+) and induces store-operated Ca(2+) (SOC) entry through plasma membrane Orai channels. The Ca(2+)/calmodulin-activated K(+) channel K(Ca)3.1 (previously known as SK4) has been implicated as an 'amplifier' of the Ca(2+)-release activated Ca(2+) (CRAC) current, especially in T lymphocytes. We have previously shown that human macrophages express K(Ca)3.1, and here we used the whole-cell patch-clamp technique to investigate the activity of these channels during Ca(2+) store depletion and store-operated Ca(2+) influx. Using RT-PCR, we found that macrophages express the elementary CRAC channel components Orai1 and STIM1, as well as Orai2, Orai3 and STIM2, but not the putatively STIM1-activated channels TRPC1, TRPC3-7 or TRPV6. In whole-cell configuration, a robust Ca(2+)-induced outwardly rectifying K(+) current inhibited by clotrimazole and augmented by DC-EBIO could be detected, consistent with K(Ca)3.1 channel current (also known as intermediate-conductance IK1). Introduction of extracellular Ca(2+) following Ca(2+) store depletion via P2Y(2) receptors induced a robust charybdotoxin (CTX)- and 2-APB-sensitive outward K(+) current and hyperpolarization. We also found that SOC entry induced by thapsigargin treatment induced CTX-sensitive K(+) current in HEK293 cells transiently expressing K(Ca)3.1. Our data suggest that SOC and K(Ca)3.1 channels are tightly coupled, such that a small Ca(2+) influx current induces a much large K(Ca)3.1 channel current and hyperpolarization, providing the necessary electrochemical driving force for prolonged Ca(2+) signaling and store repletion.


Subject(s)
Calcium Channels/biosynthesis , Calcium Signaling/physiology , Calcium/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Macrophages/metabolism , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Cell Adhesion Molecules/biosynthesis , Charybdotoxin/pharmacology , Clotrimazole/pharmacology , HEK293 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , ORAI1 Protein , ORAI2 Protein , Patch-Clamp Techniques , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , Uridine Triphosphate/pharmacology
18.
EMBO J ; 29(13): 2101-13, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20461057

ABSTRACT

The time course of inactivation of voltage-activated potassium (Kv) channels is an important determinant of the firing rate of neurons. In many Kv channels highly unsaturated lipids as arachidonic acid, docosahexaenoic acid and anandamide can induce fast inactivation. We found that these lipids interact with hydrophobic residues lining the inner cavity of the pore. We analysed the effects of these lipids on Kv1.1 current kinetics and their competition with intracellular tetraethylammonium and Kvbeta subunits. Our data suggest that inactivation most likely represents occlusion of the permeation pathway, similar to drugs that produce 'open-channel block'. Open-channel block by drugs and lipids was strongly reduced in Kv1.1 channels whose amino acid sequence was altered by RNA editing in the pore cavity, and in Kv1.x heteromeric channels containing edited Kv1.1 subunits. We show that differential editing of Kv1.1 channels in different regions of the brain can profoundly alter the pharmacology of Kv1.x channels. Our findings provide a mechanistic understanding of lipid-induced inactivation and establish RNA editing as a mechanism to induce drug and lipid resistance in Kv channels.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/metabolism , RNA Editing , Tetraethylammonium/pharmacology , Animals , Arachidonic Acid/metabolism , Binding Sites , Humans , Models, Molecular , Mutation , Neurons/drug effects , Neurons/metabolism , Oocytes/drug effects , Oocytes/metabolism , Potassium Channels, Voltage-Gated/genetics , Protein Binding , Rats , Xenopus laevis
19.
Biochim Biophys Acta ; 1800(3): 385-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19931596

ABSTRACT

BACKGROUND: 5-Hydroxydecanoate (5-HD) inhibits preconditioning, and it is assumed to be a selective inhibitor of mitochondrial ATP-sensitive K(+) (mitoK(ATP)) channels. However, 5-HD is a substrate for mitochondrial outer membrane acyl-CoA synthetase, which catalyzes the reaction: 5HD + CoA + ATP --> 5-HD-CoA (5-hydroxydecanoyl-CoA) + AMP + pyrophosphate. We aimed to determine whether the reactants or principal product of this reaction modulate sarcolemmal K(ATP) (sarcK(ATP)) channel activity. METHODS: Single sarcK(ATP) channel currents were measured in inside-out patches excised from rat ventricular myocytes. In addition, sarcK(ATP) channel activity was recorded in whole-cell configuration or in giant inside-out patches excised from oocytes expressing Kir6.2/SUR2A. RESULTS: 5-HD inhibited (IC(50) approximately 30 microM) K(ATP) channel activity, albeit only in the presence of (non-inhibitory) concentrations of ATP. Similarly, when the inhibitory effect of 0.2 mM ATP was reversed by 1 microM oleoyl-CoA, subsequent application of 5-HD blocked channel activity, but no effect was seen in the absence of ATP. Furthermore, we found that 1 microM coenzyme A (CoA) inhibited sarcK(ATP) channels. Using giant inside-out patches, which are weakly sensitive to "contaminating" CoA, we found that Kir6.2/SUR2A channels were insensitive to 5-HD-CoA. In intact myocytes, 5-HD failed to reverse sarcK(ATP) channel activation by either metabolic inhibition or rilmakalim. GENERAL SIGNIFICANCE: SarcK(ATP) channels are inhibited by 5-HD (provided that ATP is present) and CoA but insensitive to 5-HD-CoA. 5-HD is equally potent at "directly" inhibiting sarcK(ATP) and mitoK(ATP) channels. However, in intact cells, 5-HD fails to inhibit sarcK(ATP) channels, suggesting that mitochondria are the preconditioning-relevant targets of 5-HD.


Subject(s)
Coenzyme A/pharmacology , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , KATP Channels/physiology , Sarcolemma/physiology , Acetate-CoA Ligase/metabolism , Acetate-CoA Ligase/pharmacology , Animals , Heart Ventricles/drug effects , KATP Channels/antagonists & inhibitors , KATP Channels/drug effects , Mice , Muscle Cells/drug effects , Muscle Cells/physiology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Rats
20.
J Physiol ; 587(Pt 5): 929-52, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19139046

ABSTRACT

The two-pore-domain potassium channels TASK-1 (KCNK3) and TASK-3 (KCNK9) modulate the electrical activity of neurons and many other cell types. We expressed TASK-1, TASK-3 and related reporter constructs in Xenopus oocytes, mammalian cell lines and various yeast strains to study the mechanisms controlling their transport to the surface membrane and the role of 14-3-3 proteins. We measured potassium currents with the voltage-clamp technique and fused N- and C-terminal fragments of the channels to various reporter proteins to study changes in subcellular localisation and surface expression. Mutational analysis showed that binding of 14-3-3 proteins to the extreme C-terminus of TASK-1 and TASK-3 masks a tri-basic motif, KRR, which differs in several important aspects from canonical arginine-based (RxR) or lysine-based (KKxx) retention signals. Pulldown experiments with GST fusion proteins showed that the KRR motif in the C-terminus of TASK-3 channels was able to bind to COPI coatomer. Disabling the binding of 14-3-3, which exposes the KRR motif, caused localisation of the GFP-tagged channel protein mainly to the Golgi complex. TASK-1 and TASK-3 also possess a di-basic N-terminal retention signal, KR, whose function was found to be independent of the binding of 14-3-3. Suppression of channel surface expression with dominant-negative channel mutants revealed that interaction with 14-3-3 has no significant effect on the dimeric assembly of the channels. Our results give a comprehensive description of the mechanisms by which 14-3-3 proteins, together with N- and C-terminal sorting signals, control the intracellular traffic of TASK-1 and TASK-3.


Subject(s)
14-3-3 Proteins/physiology , Intracellular Space/physiology , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Protein Sorting Signals/physiology , 14-3-3 Proteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Female , Humans , Intracellular Space/genetics , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Oocytes/metabolism , Oocytes/physiology , Potassium Channels, Tandem Pore Domain/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...