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










Publication year range
1.
J Gen Physiol ; 154(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35687042

ABSTRACT

Numerous essential physiological processes depend on the TMEM16A-mediated Ca2+-activated chloride fluxes. Extensive structure-function studies have helped to elucidate the Ca2+ gating mechanism of TMEM16A, revealing a Ca2+-sensing element close to the anion pore that alters conduction. However, substrate selection and the substrate-gating relationship in TMEM16A remain less explored. Here, we study the gating-permeant anion relationship on mouse TMEM16A expressed in HEK 293 cells using electrophysiological recordings coupled with site-directed mutagenesis. We show that the apparent Ca2+ sensitivity of TMEM16A increased with highly permeant anions and SCN- mole fractions, likely by stabilizing bound Ca2+. Conversely, mutations at crucial gating elements, including the Ca2+-binding site 1, the transmembrane helix 6 (TM6), and the hydrophobic gate, impaired the anion permeability and selectivity of TMEM16A. Finally, we found that, unlike anion-selective wild-type channels, the voltage dependence of unselective TMEM16A mutant channels was less sensitive to SCN-. Therefore, our work identifies structural determinants of selectivity at the Ca2+ site, TM6, and hydrophobic gate and reveals a reciprocal regulation of gating and selectivity. We suggest that this regulation is essential to set ionic selectivity and the Ca2+ and voltage sensitivities in TMEM16A.


Subject(s)
Calcium , Chloride Channels , Animals , Anions/metabolism , Anoctamin-1/genetics , Calcium/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , HEK293 Cells , Humans , Ion Channel Gating , Mice , Neoplasm Proteins/metabolism
2.
PLoS One ; 17(1): e0261960, 2022.
Article in English | MEDLINE | ID: mdl-35030226

ABSTRACT

Inhibitory regulation of the heart is determined by both cholinergic M2 receptors (M2R) and adenosine A1 receptors (A1R) that activate the same signaling pathway, the ACh-gated inward rectifier K+ (KACh) channels via Gi/o proteins. Previously, we have shown that the agonist-specific voltage sensitivity of M2R underlies several voltage-dependent features of IKACh, including the 'relaxation' property, which is characterized by a gradual increase or decrease of the current when cardiomyocytes are stepped to hyperpolarized or depolarized voltages, respectively. However, it is unknown whether membrane potential also affects A1R and how this could impact IKACh. Upon recording whole-cell currents of guinea-pig cardiomyocytes, we found that stimulation of the A1R-Gi/o-IKACh pathway with adenosine only caused a very slight voltage dependence in concentration-response relationships (~1.2-fold EC50 increase with depolarization) that was not manifested in the relative affinity, as estimated by the current deactivation kinetics (τ = 4074 ± 214 ms at -100 mV and τ = 4331 ± 341 ms at +30 mV; P = 0.31). Moreover, IKACh did not exhibit relaxation. Contrarily, activation of the M2R-Gi/o-IKACh pathway with acetylcholine induced the typical relaxation of the current, which correlated with the clear voltage-dependent effect observed in the concentration-response curves (~2.8-fold EC50 increase with depolarization) and in the IKACh deactivation kinetics (τ = 1762 ± 119 ms at -100 mV and τ = 1503 ± 160 ms at +30 mV; P = 0.01). Our findings further substantiate the hypothesis of the agonist-specific voltage dependence of GPCRs and that the IKACh relaxation is consequence of this property.


Subject(s)
Acetylcholine/pharmacology , Adenosine A1 Receptor Agonists/pharmacology , Adenosine/pharmacology , Ion Channel Gating/drug effects , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Receptor, Adenosine A1/metabolism , Animals , Female , Guinea Pigs , Male , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/metabolism
3.
J Gen Physiol ; 153(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33822868

ABSTRACT

Inwardly rectifying potassium (Kir) channels are broadly expressed in both excitable and nonexcitable tissues, where they contribute to a wide variety of cellular functions. Numerous studies have established that rectification of Kir channels is not an inherent property of the channel protein itself, but rather reflects strong voltage dependence of channel block by intracellular cations, such as polyamines and Mg2+. Here, we identify a previously unknown mechanism of inward rectification in Kir4.1/Kir5.1 channels in the absence of these endogenous blockers. This novel intrinsic rectification originates from the voltage-dependent behavior of Kir4.1/Kir5.1, which is generated by the flux of potassium ions through the channel pore; the inward K+-flux induces the opening of the gate, whereas the outward flux is unable to maintain the gate open. This gating mechanism powered by the K+-flux is convergent with the gating of PIP2 because, at a saturating concentration, PIP2 greatly reduces the inward rectification. Our findings provide evidence of the coexistence of two rectification mechanisms in Kir4.1/Kir5.1 channels: the classical inward rectification induced by blocking cations and an intrinsic voltage-dependent mechanism generated by the K+-flux gating.


Subject(s)
Potassium Channels, Inwardly Rectifying , Ions , Potassium , Potassium Channel Blockers
4.
Br J Pharmacol ; 177(19): 4548-4560, 2020 10.
Article in English | MEDLINE | ID: mdl-32726456

ABSTRACT

BACKGROUND AND PURPOSE: Aminoglycoside antibiotics are positively charged molecules that are known to inhibit several ion channels. In this study, we have shown that aminoglycosides also inhibit the activity of Kir4.1 channels. Aminoglycosides inhibit Kir4.1 channels by a pore-blocking mechanism, plugging the central vestibule of the channel. EXPERIMENTAL APPROACH: Patch-clamp recordings were made in HEK-293 cells transiently expressing Kir4.1 channels to analyse the effects of gentamicin, neomycin and kanamycin. In silico modelling followed by mutagenesis were realized to identify the residues critical for aminoglycosides binding to Kir4.1. KEY RESULTS: Aminoglycoside antibiotics block Kir4.1 channels in a concentration- and voltage-dependent manner, getting access to the protein from the intracellular side of the plasma membrane. Aminoglycosides block Ki4.1 with a rank order of potency as follows: gentamicin ˃ neomycin ˃ kanamycin. The residues T128 and principally E158, facing the central cavity of Kir4.1, are important structural determinants for aminoglycosides binding to the channel, as determined by our in silico modelling and confirmed by mutagenesis experiments. CONCLUSION AND IMPLICATIONS: Kir4.1 channels are also target of aminoglycoside antibiotics, which could affect potassium transport in several tissues.


Subject(s)
Potassium Channels, Inwardly Rectifying , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Computer Simulation , HEK293 Cells , Humans , Potassium Channels, Inwardly Rectifying/genetics
5.
Cell Calcium ; 89: 102211, 2020 07.
Article in English | MEDLINE | ID: mdl-32422433

ABSTRACT

Chloride fluxes through the calcium-gated chloride channel Anoctamin-1 (TMEM16A) control blood pressure, secretion of saliva, mucin, insulin, and melatonin, gastrointestinal motility, sperm capacitation and motility, and pain sensation. Calcium activates a myriad of regulatory proteins but how these proteins affect TMEM16A activity is unresolved. Here we show by co-immunoprecipitation that increasing intracellular calcium with ionomycin or by activating sphingosine-1-phosphate receptors, induces coupling of calcium/calmodulin-dependent phosphatase calcineurin and prolyl isomerase FK506-binding protein 12 (FKBP12) to TMEM16A in HEK-293 cells. Application of drugs that target either calcineurin (cyclosporine A) or FKBP12 (tacrolimus known as FK506 and sirolimus known as rapamycin) caused a decrease in TMEM16A activity. In addition, FK506 and BAPTA-AM prevented co-immunoprecipitation between FKBP12 and TMEM16A. FK506 rendered the channel insensitive to cyclosporine A without altering its apparent calcium sensitivity whereas zero intracellular calcium blocked the effect of FK506. Rapamycin decreased TMEM16A activity in cells pre-treated with cyclosporine A or FK506. These results suggest the formation of a TMEM16A-FKBP12-calcineurin complex that regulates channel function. We conclude that upon a cytosolic calcium increase the TMEM16A-FKPB12-calcineurin trimers are assembled. Such hetero-oligomerization enhances TMEM16A channel activity but is not mandatory for activation by calcium.


Subject(s)
Anoctamin-1/metabolism , Calcineurin/metabolism , Calcium/pharmacology , Tacrolimus Binding Protein 1A/metabolism , Cyclosporine/pharmacology , HEK293 Cells , Humans , Protein Binding/drug effects , Protein Multimerization , Sirolimus/pharmacology , Tacrolimus/pharmacology
6.
Sci Rep ; 10(1): 6644, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313203

ABSTRACT

Anoctamin-1 (ANO1 or TMEM16A) is a homo-dimeric Ca2+-activated Cl- channel responsible for essential physiological processes. Each monomer harbours a pore and a Ca2+-binding pocket; the voltage-dependent binding of two intracellular Ca2+ ions to the pocket gates the pore. However, in the absence of intracellular Ca2+ voltage activates TMEM16A by an unknown mechanism. Here we show voltage-activated anion currents that are outwardly rectifying, time-independent with fast or absent tail currents that are inhibited by tannic and anthracene-9-carboxylic acids. Since intracellular protons compete with Ca2+ for binding sites in the pocket, we hypothesized that voltage-dependent titration of these sites would induce gating. Indeed intracellular acidification enabled activation of TMEM16A by voltage-dependent protonation, which enhanced the open probability of the channel. Mutating Glu/Asp residues in the Ca2+-binding pocket to glutamine (to resemble a permanent protonated Glu) yielded channels that were easier to activate at physiological pH. Notably, the response of these mutants to intracellular acidification was diminished and became voltage-independent. Thus, voltage-dependent protonation of glutamate/aspartate residues (Glu/Asp) located in the Ca2+-binding pocket underlines TMEM16A activation in the absence of intracellular Ca2+.


Subject(s)
Anoctamin-1/metabolism , Calcium/metabolism , Chlorides/metabolism , Recombinant Fusion Proteins/genetics , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anoctamin-1/antagonists & inhibitors , Anoctamin-1/genetics , Anthracenes/pharmacology , Cations, Divalent , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Transport/drug effects , Mice , Mutation , Patch-Clamp Techniques , Plasmids/chemistry , Plasmids/metabolism , Protons , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Tannins/pharmacology , Transfection
7.
Pflugers Arch ; 470(12): 1765-1776, 2018 12.
Article in English | MEDLINE | ID: mdl-30155776

ABSTRACT

The acetylcholine (ACh)-gated inwardly rectifying K+ current (IKACh) plays a vital role in cardiac excitability by regulating heart rate variability and vulnerability to atrial arrhythmias. These crucial physiological contributions are determined principally by the inwardly rectifying nature of IKACh. Here, we investigated the relative contribution of two distinct mechanisms of IKACh inward rectification measured in atrial myocytes: a rapid component due to KACh channel block by intracellular Mg2+ and polyamines; and a time- and concentration-dependent mechanism. The time- and ACh concentration-dependent inward rectification component was eliminated when IKACh was activated by GTPγS, a compound that bypasses the muscarinic-2 receptor (M2R) and directly stimulates trimeric G proteins to open KACh channels. Moreover, the time-dependent component of IKACh inward rectification was also eliminated at ACh concentrations that saturate the receptor. These observations indicate that the time- and concentration-dependent rectification mechanism is an intrinsic property of the receptor, M2R; consistent with our previous work demonstrating that voltage-dependent conformational changes in the M2R alter the receptor affinity for ACh. Our analysis of the initial and time-dependent components of IKACh indicate that rapid Mg2+-polyamine block accounts for 60-70% of inward rectification, with M2R voltage sensitivity contributing 30-40% at sub-saturating ACh concentrations. Thus, while both inward rectification mechanisms are extrinsic to the KACh channel, to our knowledge, this is the first description of extrinsic inward rectification of ionic current attributable to an intrinsic voltage-sensitive property of a G protein-coupled receptor.


Subject(s)
Action Potentials , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Myocytes, Cardiac/metabolism , Receptor, Muscarinic M2/metabolism , Acetylcholine/metabolism , Animals , Cats , Cells, Cultured , Female , Heart Atria/cytology , Magnesium/metabolism , Male , Myocytes, Cardiac/physiology , Polyamines/metabolism
8.
Sci Rep ; 8(1): 1769, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29379118

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) is a membrane phospholipid that regulates the function of multiple ion channels, including some members of the voltage-gated potassium (Kv) channel superfamily. The PIP2 sensitivity of Kv channels is well established for all five members of the Kv7 family and for Kv1.2 channels; however, regulation of other Kv channels by PIP2 remains unclear. Here, we investigate the effects of PIP2 on Kv2.1 channels by applying exogenous PIP2 to the cytoplasmic face of excised membrane patches, activating muscarinic receptors (M1R), or depleting endogenous PIP2 using a rapamycin-translocated 5-phosphatase (FKBP-Inp54p). Exogenous PIP2 rescued Kv2.1 channels from rundown and partially prevented the shift in the voltage-dependence of inactivation observed in inside-out patch recordings. Native PIP2 depletion by the recruitment of FKBP-Insp54P or M1R activation in whole-cell experiments, induced a shift in the voltage-dependence of inactivation, an acceleration of the closed-state inactivation, and a delayed recovery of channels from inactivation. No significant effects were observed on the activation mechanism by any of these treatments. Our data can be modeled by a 13-state allosteric model that takes into account that PIP2 depletion facilitates inactivation of Kv2.1. We propose that PIP2 regulates Kv2.1 channels by interfering with the inactivation mechanism.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/metabolism , Shab Potassium Channels/metabolism , HEK293 Cells , Humans , Ion Channel Gating/physiology , Patch-Clamp Techniques/methods , Potassium Channels, Voltage-Gated/metabolism , Receptors, Muscarinic/metabolism
9.
Pharmacol Rep ; 69(6): 1145-1153, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29128793

ABSTRACT

BACKGROUND: Phytochemicals are a large group of plant-derived compounds that have a broad range of pharmacological effects. Some of these effects are derived from their action on transport proteins, including ion channels. The present study investigates the effects of the phytochemicals genistein and capsaicin on voltage-gated potassium Kv2.1 channels. METHODS: The whole-cell patch clamp technique was used to explore the regulation of Kv2.1 channels expressed in HEK293 cells by genistein and capsaicin. RESULTS: Both phytochemicals had a profound effect on the gating properties of Kv2.1 channels; the voltage dependence of activation and inactivation was shifted to hyperpolarized potentials, the closed-state inactivation was accelerated, and the recovery from inactivation was delayed. Moreover, genistein and capsaicin inhibited Kv2.1 currents in a concentration dependent manner. CONCLUSION: This study effectively demonstrated the inhibitory effects of genistein and capsaicin on Kv2.1 channels. As Kv2.1 channels play a prominent role in glucose-stimulated insulin secretion, our findings contribute to our understanding of the putative mechanism by which these phytochemicals exert their reported hypoglycemic effects.


Subject(s)
Capsaicin/pharmacology , Genistein/pharmacology , Shab Potassium Channels/antagonists & inhibitors , Capsaicin/administration & dosage , Dose-Response Relationship, Drug , Genistein/administration & dosage , HEK293 Cells , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/pharmacology , Shab Potassium Channels/metabolism
10.
Eur J Pharmacol ; 815: 56-63, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28993158

ABSTRACT

Inward rectifier potassium (Kir) channels are expressed in almost all mammalian tissues and contribute to a wide range of physiological processes. Kir4.1 channel expression is found in the brain, inner ear, eye, and kidney. Loss-of-function mutations in the pore-forming Kir4.1 subunit cause an autosomal recessive disorder characterized by epilepsy, ataxia, sensorineural deafness and tubulopathy (SeSAME/EST syndrome). Despite its importance in physiological and pathological conditions, pharmacological research of Kir4.1 is limited. Here, we characterized the effect of pentamidine on Kir4.1 channels using electrophysiology, mutagenesis and computational methods. Pentamidine potently inhibited Kir4.1 channels when applied to the cytoplasmic side under inside-out patch clamp configuration (IC50 = 97nM). The block was voltage dependent. Molecular modeling predicted the binding of pentamidine to the transmembrane pore region of Kir4.1 at aminoacids T127, T128 and E158. Mutation of each of these residues reduced the potency of pentamidine to block Kir4.1 channels. A pentamidine analog (PA-6) inhibited Kir4.1 with similar potency (IC50 = 132nM). Overall, this study shows that pentamidine blocks Kir4.1 channels interacting with threonine and glutamate residues in the transmembrane pore region. These results can be useful to design novel compounds with major potency and specificity over Kir4.1 channels.


Subject(s)
Pentamidine/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Binding Sites , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Pentamidine/metabolism , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Protein Conformation
11.
Brain Res ; 1663: 87-94, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28288868

ABSTRACT

Inwardly rectifying potassium (Kir) channels are expressed in many cell types and contribute to a wide range of physiological processes. Particularly, Kir4.1 channels are involved in the astroglial spatial potassium buffering. In this work, we examined the effects of the cationic amphiphilic drug quinacrine on Kir4.1 channels heterologously expressed in HEK293 cells, employing the patch clamp technique. Quinacrine inhibited the currents of Kir4.1 channels in a concentration and voltage dependent manner. In inside-out patches, quinacrine inhibited Kir4.1 channels with an IC50 value of 1.8±0.3µM and with extremely slow blocking and unblocking kinetics. Molecular modeling combined with mutagenesis studies suggested that quinacrine blocks Kir4.1 by plugging the central cavity of the channels, stabilized by the residues E158 and T128. Overall, this study shows that quinacrine blocks Kir4.1 channels, which would be expected to impact the potassium transport in several tissues.


Subject(s)
Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Quinacrine/pharmacology , Animals , Astrocytes/metabolism , HEK293 Cells , Humans , Ion Channel Gating/physiology , Patch-Clamp Techniques/methods , Potassium/metabolism , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Quinacrine/metabolism , Rats
12.
Eur J Pharmacol ; 800: 40-47, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28216048

ABSTRACT

Kir4.1 channels have been implicated in various physiological processes, mainly in the K+ homeostasis of the central nervous system and in the control of glial function and neuronal excitability. Even though, pharmacological research of these channels is very limited. Chloroquine (CQ) is an amino quinolone derivative known to inhibit Kir2.1 and Kir6.2 channels with different action mechanism and binding site. Here, we employed patch-clamp methods, mutagenesis analysis, and molecular modeling to characterize the molecular pharmacology of Kir4.1 inhibition by CQ. We found that this drug inhibits Kir4.1 channels heterologously expressed in HEK-293 cells. CQ produced a fast-onset voltage-dependent pore-blocking effect on these channels. In inside-out patches, CQ showed notable higher potency (IC50 ≈0.5µM at +50mV) and faster onset of block when compared to whole-cell configuration (IC50 ≈7µM at +60mV). Also, CQ showed a voltage-dependent unblock with repolarization. These results suggest that the drug directly blocks Kir4.1 channels by a pore-plugging mechanism. Moreover, we found that two residues (Thr128 and Glu158), facing the central cavity and located within the transmembrane pore, are particularly important structural determinants of CQ block. This evidence was similar to what was previously reported with Kir6.2, but distinct from the interaction site (cytoplasmic pore) CQ-Kir2.1. Thus, our findings highlight the diversity of interaction sites and mechanisms that underlie amino quinolone inhibition of Kir channels.


Subject(s)
Chloroquine/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/chemistry , Binding Sites , Chloroquine/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Kinetics , Molecular Docking Simulation , Porosity , Potassium Channel Blockers/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Protein Conformation
13.
J Physiol ; 595(5): 1515-1531, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27859335

ABSTRACT

KEY POINTS: The calcium-activated chloride channel TMEM16A provides a pathway for chloride ion movements that are key in preventing polyspermy, allowing fluid secretion, controlling blood pressure, and enabling gastrointestinal activity. TMEM16A is opened by voltage-dependent calcium binding and regulated by permeant anions and intracellular protons. Here we show that a low proton concentration reduces TMEM16A activity while maximum activation is obtained when the external proton concentration is high. In addition, protonation conditions determine the open probability of TMEM16A without changing its calcium sensitivity. External glutamic acid 623 (E623) is key for TMEM16A's ability to respond to external protons. At physiological pH, E623 is un-protonated and TMEM16A is activated when intracellular calcium increases; however, under acidic conditions E623 is partially protonated and works synergistically with intracellular calcium to activate the channel. These findings are critical for understanding physiological and pathological processes that involve changes in pH and chloride flux via TMEM16A. ABSTRACT: Transmembrane protein 16A (TMEM16A), also known as ANO1, the pore-forming subunit of a Ca2+ -dependent Cl- channel (CaCC), is activated by direct, voltage-dependent, binding of intracellular Ca2+ . Endogenous CaCCs are regulated by extracellular protons; however, the molecular basis of such regulation remains unidentified. Here, we evaluated the effects of different extracellular proton concentrations ([H+ ]o ) on mouse TMEM16A expressed in HEK-293 cells using whole-cell and inside-out patch-clamp recordings. We found that increasing the [H+ ]o from 10-10 to 10-5.5  m caused a progressive increase in the chloride current (ICl ) that is described by titration of a protonatable site with pK = 7.3. Protons regulate TMEM16A in a voltage-independent manner, regardless of channel state (open or closed), and without altering its apparent Ca2+ sensitivity. Noise analysis showed that protons regulate TMEM16A by tuning its open probability without modifying the single channel current. We found a robust reduction of the proton effect at high [Ca2+ ]i . To identify protonation targets we mutated all extracellular glutamate and histidine residues and 4 of 11 aspartates. Most mutants were sensitive to protons. However, mutation that substituted glutamic acid (E) for glutamine (Q) at amino acid position 623 (E623Q) displayed a titration curve shifted to the left relative to wild type channels and the ICl was nearly insensitive to proton concentrations between 10-5.5 and 10-9.0  m. Additionally, ICl of the mutant containing an aspartic acid (D) to asparagine (N) substitution at position 405 (D405N) mutant was partially inhibited by a proton concentration of 10-5.5  m, but 10-9.0  m produced the same effect as in wild type. Based on our findings we propose that external protons titrate glutamic acid 623, which enables voltage activation of TMEM16A at non-saturating [Ca2+ ]i .


Subject(s)
Chloride Channels/physiology , Anoctamin-1 , Calcium/physiology , Chloride Channels/genetics , HEK293 Cells , Humans , Models, Molecular , Protons
14.
Cell Physiol Biochem ; 40(6): 1261-1273, 2016.
Article in English | MEDLINE | ID: mdl-27997898

ABSTRACT

BACKGROUND: The rapid delayed rectifier K+ current (IKr), carried by the hERG protein, is one of the main repolarising currents in the human heart and a reduction of this current increases the risk of ventricular fibrillation. α1-adrenoceptors (α1-AR) activation reduces IKr but, despite the clear relationship between an increase in the sympathetic tone and arrhythmias, the mechanisms underlying the α1-AR regulation of the hERG channel are controversial. Thus, we aimed to investigate the mechanisms by which α1-AR stimulation regulates IKr. METHODS: α1-adrenoceptors, hERG channels, auxiliary subunits minK and MIRP1, the non PIP2-interacting mutant D-hERG (with a deletion of the 883-894 amino acids) in the C-terminal and the non PKC-phosphorylable mutant N-terminal truncated-hERG (NTK-hERG) were transfected in HEK293 cells. Cell membranes were extracted by centrifugation and the different proteins were visualized by Western blot. Potassium currents were recorded by the patch-clamp technique. IKr was recorded in isolated feline cardiac myocytes. RESULTS: Activation of the α1-AR reduces the amplitude of IhERG and IKr through a positive shift in the activation half voltage, which reduces the channel availability at physiological membrane potentials. The intracellular pathway connecting the α1-AR to the hERG channel in HEK293 cells includes activation of the Gαq protein, PLC activation and PIP2 hydrolysis, activation of PKC and direct phosphorylation of the hERG channel N-terminal. The PKC-mediated IKr channel phosphorylation and subsequent IKr reduction after α1-AR stimulation was corroborated in feline cardiac myocytes. CONCLUSIONS: These findings clarify the link between sympathetic nervous system hyperactivity and IKr reduction, one of the best characterized causes of torsades de pointes and ventricular fibrillation.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Ion Channel Gating , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Animals , Cats , Enzyme Activation/drug effects , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phenylephrine/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation/drug effects , Potassium Channels, Voltage-Gated/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism
15.
Pharmacol Rep ; 68(2): 383-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26922543

ABSTRACT

BACKGROUND: Inwardly rectifying potassium (Kir) channels are expressed in many cell types and contribute to a wide range of physiological processes. Kir channels dysfunction cause several diseases in brain, ear, heart, muscle, kidney and pancreas, and developmental abnormalities. Therefore, a better understanding of Kir channels pharmacology is desirable. In this study we characterized the electrophysiological and molecular basis of the inhibition produced by the α-adrenergic agonist/antagonist chloroethylclonidine of the currents generated by wild type and mutant Kir2.1 and Kir4.1 channels heterologously expressed in HEK293 cells. METHODS: Macroscopic currents were recorded using the patch clamp technique in the inside out configuration. RESULTS: We found that chloroethylclonidine inhibits the Kir2.1 and Kir4.1 channels in a voltage-dependent manner by interacting with pore facing residues in the cytoplasmic and transmembrane domains, respectively. Site-directed mutagenesis experiments demonstrate that chloroethylclonidine interact with Kir2.1 channels in the cytoplasmic pore involving the E224, E299, D255 and D259 residues, whereas in Kir4.1channels T128 and E158 residues located in the transmembrane pore are important for the chloroethylclonidine effect. CONCLUSIONS: Overall, our results suggest that differences in the cavity of Kir channels are determinants in its interactions with chloroethylclonidine.


Subject(s)
Clonidine/analogs & derivatives , Potassium Channels, Inwardly Rectifying/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Clonidine/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , HEK293 Cells , Humans , Membrane Proteins , Mutagenesis, Site-Directed/methods , Potassium/metabolism
16.
Pharmacol Rep ; 68(2): 457-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26922553

ABSTRACT

BACKGROUND: The aim of the present study was to assess the effects of perifosine-a third generation alkylphospholipid analog with anti-tumor properties-on the activity of Kv2.1 channels. METHODS: The whole-cell patch clamp technique was applied to follow the modulatory effect of perifosine on Kv2.1 channels expressed in HEK293 cells. RESULTS: Obtained data provide evidence that perifosine application decreases the whole cell Kv2.1 currents in a concentration-independent manner. Perifosine induces a hyperpolarizing shift in the voltage dependence of Kv2.1 channels inactivation without altering the voltage dependence of channels activation. The kinetics of Kv2.1 closed-state inactivation was accelerated by perifosine, with no significant effects on the recovery rate from inactivation. CONCLUSIONS: Taken together, these results show that perifosine modified the Kv2.1 inactivation gating resulting in a decrease of the current amplitude. These data will help to elucidate the mechanism of action of this promising anti-cancer drug on ion channels and their possible implications.


Subject(s)
Antineoplastic Agents/pharmacology , Ion Channel Gating/drug effects , Phosphorylcholine/analogs & derivatives , Potassium Channels, Voltage-Gated/metabolism , Shab Potassium Channels/metabolism , Cell Line , HEK293 Cells , Humans , Kinetics , Membrane Potentials/drug effects , Patch-Clamp Techniques/methods , Phosphorylcholine/pharmacology , Potassium/metabolism
17.
Pharmacol Rep ; 67(6): 1273-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26481552

ABSTRACT

BACKGROUND: The aim of the present study was to assess the effects of curcumin on the voltage-dependent Kv2.1 potassium channel. METHODS: The whole-cell patch-clamp technique was used to explore the regulation of Kv2.1 channels expressed in HEK293 cells by curcumin. RESULTS: Curcumin reduced the Kv2.1 currents; the inhibition occurred with a slow time course and was partially reversible. Curcumin did not alter the kinetics and voltage dependence of activation; however, the kinetics of open- and closed-state inactivation was accelerated by curcumin along with a hyperpolarizing shift in the voltage dependence of inactivation. Curcumin inhibition of Kv2.1 current was not use-dependent. CONCLUSIONS: Overall, our data suggest that curcumin inhibits Kv2.1 channels by modulating the inactivation gating, which would be expected to impact cellular physiology.


Subject(s)
Curcumin/pharmacology , Shab Potassium Channels/antagonists & inhibitors , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects
18.
PLoS One ; 8(9): e76085, 2013.
Article in English | MEDLINE | ID: mdl-24086693

ABSTRACT

KCNQ genes encode five Kv7 K(+) channel subunits (Kv7.1-Kv7.5). Four of these (Kv7.2-Kv7.5) are expressed in the nervous system. Kv7.2 and Kv7.3 are the principal molecular components of the slow voltage-gated M-channel, which regulates neuronal excitability. In this study, we demonstrate that tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits Kv7.2/Kv7.3 currents heterologously expressed in human embryonic kidney HEK-293 cells. Current inhibition by tamoxifen was voltage independent but concentration-dependent. The IC50 for current inhibition was 1.68 ± 0.44 µM. The voltage-dependent activation of the channel was not modified. Tamoxifen inhibited Kv7.2 homomeric channels with a higher potency (IC50 = 0.74 ± 0.16 µM). The mutation Kv7.2 R463E increases phosphatidylinositol- 4,5-bisphosphate (PIP2) - channel interaction and diminished dramatically the inhibitory effect of tamoxifen compared with that for wild type Kv7.2. Conversely, the mutation Kv7.2 R463Q, which decreases PIP2 -channel interaction, increased tamoxifen potency. Similar results were obtained on the heteromeric Kv7.2 R463Q/Kv7.3 and Kv7.2 R463E/Kv7.3 channels, compared to Kv7.2/Kv7.3 WT. Overexpression of type 2A PI(4)P5-kinase (PIP5K 2A) significantly reduced tamoxifen inhibition of Kv7.2/Kv7.3 and Kv7.2 R463Q channels. Our results suggest that tamoxifen inhibited Kv7.2/Kv7.3 channels by interfering with PIP2-channel interaction because of its documented interaction with PIP2 and the similar effect of tamoxifen on various PIP2 sensitive channels.


Subject(s)
KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/antagonists & inhibitors , KCNQ3 Potassium Channel/metabolism , Tamoxifen/pharmacology , HEK293 Cells , Humans , Inhibitory Concentration 50 , KCNQ2 Potassium Channel/genetics , Mutation, Missense/genetics , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism
19.
J Physiol ; 591(17): 4273-86, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23652593

ABSTRACT

Choline (Ch) is a precursor and metabolite of the neurotransmitter acetylcholine (ACh). In canine and guinea pig atrial myocytes, Ch was shown to activate an outward K(+) current in a delayed rectifier fashion. This current has been suggested to modulate cardiac electrical activity and to play a role in atrial fibrillation pathophysiology. However, the exact nature and identity of this current has not been convincingly established. We recently described the unique ligand- and voltage-dependent properties of muscarinic activation of ACh-activated K(+) current (IKACh) and showed that, in contrast to ACh, pilocarpine induces a current with delayed rectifier-like properties with membrane depolarization. Here, we tested the hypothesis that Ch activates IKACh in feline atrial myocytes in a voltage-dependent manner similar to pilocarpine. Single-channel recordings, biophysical profiles, specific pharmacological inhibition and computational data indicate that the current activated by Ch is IKACh. Moreover, we show that membrane depolarization increases the potency and efficacy of IKACh activation by Ch and thus gives the appearance of a delayed rectifier activating K(+) current at depolarized potentials. Our findings support the emerging concept that IKACh modulation is both voltage- and ligand-specific and reinforce the importance of these properties in understanding cardiac physiology.


Subject(s)
Action Potentials , Delayed Rectifier Potassium Channels/metabolism , Heart Atria/metabolism , Myocytes, Cardiac/metabolism , Receptor, Muscarinic M2/metabolism , Animals , Cats , Choline/pharmacology , Female , Heart Atria/cytology , Male , Membrane Potentials , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pilocarpine/pharmacology
20.
Pflugers Arch ; 462(4): 505-17, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21779761

ABSTRACT

Cardiac inward rectifier potassium currents determine the resting membrane potential and contribute repolarization capacity during phase 3 repolarization. Quinacrine is a cationic amphiphilic drug. In this work, the effects of quinacrine were studied on cardiac Kir channels expressed in HEK 293 cells and on the inward rectifier potassium currents, I(K1) and I(KATP), in cardiac myocytes. We found that quinacrine differentially inhibited Kir channels, Kir6.2 ∼ Kir2.3 > Kir2.1. In addition, we found in cardiac myocytes that quinacrine inhibited I(KATP) > I(K1). We presented evidence that quinacrine displays a double action towards strong inward rectifier Kir2.x channels, i.e., direct pore block and interference in phosphatidylinositol 4,5-bisphosphate, PIP(2)-Kir channel interaction. Pore block is evident in Kir2.1 and 2.3 channels as rapid block; channel block involves residues E224 and E299 facing the cytoplasmic pore of Kir2.1. The interference of the drug with the interaction of Kir2.x and Kir6.2/SUR2A channels and PIP(2) is suggested from four sources of evidence: (1) Slow onset of current block when quinacrine is applied from either the inside or the outside of the channel. (2) Mutation of Kir2.3(I213L) and mutation of Kir6.2(C166S) increase their affinity for PIP(2) and lowers its sensitivity for quinacrine. (3) Mutations of Kir2.1(L222I and K182Q) which decreased its affinity for PIP(2) increased its sensitivity for quinacrine. (4) Co-application of quinacrine with PIP(2) lowers quinacrine-mediated current inhibition. In conclusion, our data demonstrate how an old drug provides insight into a dual a blocking mechanism of Kir carried inward rectifier channels.


Subject(s)
Myocytes, Cardiac/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , HEK293 Cells , Humans , Quinacrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...