Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Mol Pharmacol ; 98(2): 143-155, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32616523

RESUMO

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.


Assuntos
1-Metil-3-Isobutilxantina/farmacologia , Canais de Potássio de Domínios Poros em Tandem/química , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Arginina/genética , Sítios de Ligação/efeitos dos fármacos , Células CHO , Cricetulus , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Mutação , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/genética , Ratos , Xenopus
2.
Pflugers Arch ; 467(5): 1069-79, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25539776

RESUMO

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.


Assuntos
Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Potássio/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Inibidores de Fosfodiesterase/farmacologia , Ratos
3.
Pflugers Arch ; 467(5): 867-94, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25791628

RESUMO

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'.


Assuntos
Potenciais de Ação/fisiologia , Fenômenos Fisiológicos Celulares/fisiologia , Neurônios/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Potássio/metabolismo , Animais , Humanos , Ativação do Canal Iônico/fisiologia , Canais de Potássio de Domínios Poros em Tandem/química
4.
Pflugers Arch ; 467(5): 1105-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25559843

RESUMO

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.


Assuntos
Citoplasma/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Transporte Proteico/fisiologia , Animais , Endocitose/fisiologia , Humanos
5.
Pflugers Arch ; 466(9): 1735-45, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24297522

RESUMO

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.


Assuntos
Canais de Potássio de Domínios Poros em Tandem/metabolismo , Transporte Proteico/fisiologia , Animais , Células CHO , Células COS , Chlorocebus aethiops , Cricetulus , Células HeLa , Humanos , Oócitos , Técnicas de Patch-Clamp , Ratos , Transfecção , Xenopus laevis
6.
Pflugers Arch ; 466(8): 1559-70, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24196565

RESUMO

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.


Assuntos
Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico/genética , Sequência de Aminoácidos , Animais , Western Blotting , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido
7.
EMBO J ; 29(13): 2101-13, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20461057

RESUMO

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.


Assuntos
Ácidos Graxos Insaturados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Edição de RNA , Tetraetilamônio/farmacologia , Animais , Ácido Araquidônico/metabolismo , Sítios de Ligação , Humanos , Modelos Moleculares , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , Ratos , Xenopus laevis
8.
FASEB J ; 26(2): 513-22, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22002906

RESUMO

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.


Assuntos
Síndrome de Andersen/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Síndrome de Andersen/tratamento farmacológico , Síndrome de Andersen/genética , Animais , Feminino , Glucocorticoides/uso terapêutico , Cobaias , Células HEK293 , Células HeLa , Humanos , Proteínas Imediatamente Precoces/metabolismo , Técnicas In Vitro , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miócitos Cardíacos/metabolismo , Oócitos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Estresse Fisiológico , Xenopus laevis
9.
J Biol Chem ; 286(16): 13977-84, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21362619

RESUMO

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.


Assuntos
Benzamidas/farmacologia , Benzenoacetamidas/farmacologia , Proteínas do Tecido Nervoso/química , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/química , Potássio/química , Alanina/química , Animais , Benzamidas/química , Benzenoacetamidas/química , Sítios de Ligação , DNA Complementar/metabolismo , Desenho de Fármacos , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Mutagênese , Mutagênese Sítio-Dirigida , Oócitos/citologia , Técnicas de Patch-Clamp , Conformação Proteica , Xenopus laevis
10.
Physiology (Bethesda) ; 26(3): 181-91, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21670164

RESUMO

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.


Assuntos
Proteínas 14-3-3/metabolismo , Membrana Celular/metabolismo , Transdução de Sinais , Proteínas 14-3-3/química , Animais , Sítios de Ligação , Humanos , Fosforilação , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
EMBO J ; 27(23): 3164-74, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-18987637

RESUMO

Inactivation of voltage-gated Kv1 channels can be altered by Kvbeta subunits, which block the ion-conducting pore to induce a rapid ('N-type') inactivation. Here, we investigate the mechanisms and structural basis of Kvbeta1.3 interaction with the pore domain of Kv1.5 channels. Inactivation induced by Kvbeta1.3 was antagonized by intracellular PIP(2). Mutations of R5 or T6 in Kvbeta1.3 enhanced Kv1.5 inactivation and markedly reduced the effects of PIP(2). R5C or T6C Kvbeta1.3 also exhibited diminished binding of PIP(2) compared with wild-type channels in an in vitro lipid-binding assay. Further, scanning mutagenesis of the N terminus of Kvbeta1.3 revealed that mutations of L2 and A3 eliminated N-type inactivation. Double-mutant cycle analysis indicates that R5 interacts with A501 and T480 of Kv1.5, residues located deep within the pore of the channel. These interactions indicate that Kvbeta1.3, in contrast to Kvbeta1.1, assumes a hairpin structure to inactivate Kv1 channels. Taken together, our findings indicate that inactivation of Kv1.5 is mediated by an equilibrium binding of the N terminus of Kvbeta1.3 between phosphoinositides (PIPs) and the inner pore region of the channel.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/química , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Quaternária de Proteína
12.
Biochim Biophys Acta ; 1800(3): 385-91, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19931596

RESUMO

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.


Assuntos
Coenzima A/farmacologia , Ácidos Decanoicos/farmacologia , Hidroxiácidos/farmacologia , Canais KATP/fisiologia , Sarcolema/fisiologia , Acetato-CoA Ligase/metabolismo , Acetato-CoA Ligase/farmacologia , Animais , Ventrículos do Coração/efeitos dos fármacos , Canais KATP/antagonistas & inibidores , Canais KATP/efeitos dos fármacos , Camundongos , Células Musculares/efeitos dos fármacos , Células Musculares/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Ratos
13.
Cell Physiol Biochem ; 28(4): 663-72, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22178878

RESUMO

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.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Fosfopiruvato Hidratase/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Substituição de Aminoácidos , Animais , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/fisiologia , Células HEK293 , Humanos , Imunoprecipitação , Rim/enzimologia , Rim/metabolismo , Oócitos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Xenopus laevis/genética
14.
Cell Physiol Biochem ; 28(1): 77-86, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21865850

RESUMO

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.


Assuntos
Síndrome do QT Longo/etiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Sulfonamidas/farmacologia , ortoaminobenzoatos/farmacologia , Potenciais de Ação/fisiologia , Anestésicos Inalatórios/farmacologia , Animais , Fenômenos Eletrofisiológicos/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/fisiologia , Isoflurano/farmacologia , Metoxamina/farmacologia , Camundongos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Proteínas do Tecido Nervoso/deficiência , Canais de Potássio de Domínios Poros em Tandem/deficiência
15.
Cell Physiol Biochem ; 28(4): 613-24, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22178873

RESUMO

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.


Assuntos
Potenciais de Ação/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Idoso , Animais , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Células Cultivadas , Eletrocardiografia , Feminino , Átrios do Coração/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/genética , Oócitos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/genética , Xenopus
17.
J Physiol ; 587(Pt 5): 929-52, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19139046

RESUMO

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.


Assuntos
Proteínas 14-3-3/fisiologia , Espaço Intracelular/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Proteínas 14-3-3/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Feminino , Humanos , Espaço Intracelular/genética , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Oócitos/metabolismo , Oócitos/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Sinais Direcionadores de Proteínas/genética , Transporte Proteico/genética , Xenopus laevis
18.
Cardiovasc Res ; 79(1): 80-8, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18339646

RESUMO

AIMS: Potassium channels are essential elements of endothelial function. Recently, evidence emerged that the TWIK (tandem of P domains in a weak inwardly rectifying K+ channel)-related K+ channel (TREK-1) of the two-pore domain potassium channel gene family (K2P) may be involved in the regulation of vascular tone. However, the functional and molecular characterization of vascular TREK-1 is incomplete. In this study, we therefore analysed the functional expression of TREK-1 in the endothelium. Moreover, we hypothesized that changes in channel expression may contribute to altered endothelial vasodilator response under conditions of elevated blood pressure. METHODS AND RESULTS: Gene expression and function of endothelial TREK-1 were analysed by single-cell RT-PCR, the patch-clamp technique and pressure myography in murine carotid arteries (CA). K+ outward currents displaying the characteristics of TREK-1 were observed following various TREK-1-activating stimuli such as membrane stretch, intracellular acidosis, polyunsaturated fatty acids, isoflurane (ISOFL), riluzole, and acetylcholine (ACh). In K(Ca)3.1(-/-) mice exhibiting elevated blood pressure, endothelial TREK-1 currents and TREK-1 mRNA expression were enhanced as compared with normotensive control mice. TREK-1-mediated vasodilator responses to alpha-linolenic acid, ISOFL, or ACh were increased. A similar up-regulation of endothelial TREK-1 was observed in spontaneously hypertensive rats. CONCLUSION: We have found that TREK-1 is an endothelial K+ channel capable of producing hyperpolarization and vasodilation. A correlation between hypertension and up-regulation of TREK-1 was observed in two different animal models of elevated blood pressure. Thus, TREK-1 may play a protective role in the cardiovascular system by providing a novel type of endothelial hyperpolarization-mediated vasodilator response.


Assuntos
Artérias Carótidas/metabolismo , Endotélio Vascular/metabolismo , Hipertensão/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Pressão Sanguínea/fisiologia , Artérias Carótidas/citologia , Artérias Carótidas/patologia , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/genética , Estrutura Terciária de Proteína/genética , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Regulação para Cima/fisiologia , Vasodilatação/fisiologia
19.
Epileptic Disord ; 10(1): 13-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18367426

RESUMO

PURPOSE: Evaluation of the loci 2q36, 3q26, 5q34 and 14q23 in a German family with autosomal dominant idiopathic generalized epilepsy (IGE). METHODS: A linkage analysis was performed including 10 family members (six affected, three unaffected, one probably affected), for the loci 2q36, 3q26, 5q34 and 14q23. Subsequently, a sequence analysis of the inward rectifier potassium channel gene KCNJ13 at 2q37 was carried out. RESULTS: Suggestive linkage for IGE was found on 2q36-37 at D2S2308 and D2S2193. No mutations were identified in the coding or 5'-non-coding regions of the exons of candidate gene KCNJ13. CONCLUSIONS: Our results corroborate former linkage findings on 2q36-2q37 and warrant further investigation of additional candidate genes within this chromosomal area in IGE.


Assuntos
Epilepsia Generalizada/genética , Epilepsia Generalizada/patologia , Adulto , Idoso , Mapeamento Cromossômico , Cromossomos Humanos Par 2/genética , DNA/genética , Eletroencefalografia , Feminino , Ligação Genética/genética , Humanos , Escore Lod , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Mutação/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.1 , Proteínas do Tecido Nervoso/genética , Linhagem , Polimorfismo Genético/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Sódio/genética
20.
Cardiovasc Res ; 75(4): 748-57, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17568571

RESUMO

OBJECTIVE: Andersen syndrome (AS) is a rare genetic disease caused by mutations of the potassium channel Kir2.1 (KCNJ2). We identified two unrelated patients with mutations in the slide helix of Kir2.1 leading to AS. The functional consequences of these two mutations, Y68D and D78Y, were studied and compared with previously reported slide helix mutations. METHODS: Channel function and surface expression were studied by voltage clamp recordings and a chemiluminescence assay in Xenopus laevis oocytes and by patch clamp recordings and fluorescence microscopy in HEK293 cells. In addition, a phosphatidylinositol bisphosphate (PIP(2)) binding assay and a yeast-two-hybrid assay were used to characterize the molecular mechanisms by which slide helix mutations cause AS. RESULTS: Neither mutant channel produced any current, but both had dominant negative effects on Kir2.2, Kir2.3, and Kir2.4 channels. We show that Y68D, D78Y, and previously reported AS mutations are clustered on the hydrophilic, cytosolic side of the slide helix and traffic normally to the plasma membrane. The in vitro lipid binding assay indicated that Y68D or D78Y N-terminal peptides bind PIP(2) similar to wild-type peptides. Yeast-two-hybrid assays showed that AS-associated mutations disturb the interaction between the slide helix and the C-terminal domain of the channel protein. CONCLUSION: Our experiments indicate a new disease-causing mechanism independent of trafficking and PIP(2) binding defects. Our findings suggest that the hydrophilic side of the slide helix interacts with a specific domain of the C-terminus facing the membrane. This interaction, which may be required for normal gating both in homomeric and heteromeric Kir2 channels, is disturbed by several mutations causing AS.


Assuntos
Síndrome de Andersen/genética , Ativação do Canal Iônico/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adulto , Síndrome de Andersen/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Análise Mutacional de DNA , Feminino , Expressão Gênica , Humanos , Microscopia de Fluorescência , Oócitos/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/análise , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Técnicas do Sistema de Duplo-Híbrido , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA