Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Adv Exp Med Biol ; 1422: 279-304, 2023.
Article in English | MEDLINE | ID: mdl-36988885

ABSTRACT

Chloride fluxes through homo-dimeric calcium-activated channels TMEM16A and TMEM16B are critical to blood pressure, gastrointestinal motility, hormone, fluid and electrolyte secretion, pain sensation, sensory transduction, and neuronal and muscle excitability. Their gating depends on the voltage-dependent binding of two intracellular calcium ions to a high-affinity site formed by acidic residues from α-helices 6-8 in each monomer. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a low-abundant lipid of the inner leaflet, supports TMEM16A function; it allows TMEM16A to evade the down-regulation induced by calcium, poly-L-lysine, or PI(4,5)P2 5-phosphatase. In stark contrast, adding or removing PI(4,5)P2 diminishes or increases TMEM16B function, respectively. PI(4,5)P2-binding sites on TMEM16A, and presumably on TMEM16B, are on the cytosolic side of α-helices 3-5, opposite the calcium-binding sites. This modular structure suggested that PI(4,5)P2 and calcium cooperate to maintain the conductive state in TMEM16A. Cholesterol, the second-largest constituent of the plasma membrane, also regulates TMEM16A though the mechanism, functional outcomes, binding site(s), and effects on TMEM16A and TMEM16B remain unknown.


Subject(s)
Chloride Channels , Phosphatidylinositols , Humans , Chloride Channels/genetics , Chloride Channels/chemistry , Chloride Channels/metabolism , Anoctamin-1/metabolism , Calcium/metabolism , Cholesterol , Calcium Channels , HEK293 Cells
2.
Biophys J ; 120(11): 2287-2295, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33864789

ABSTRACT

Fluorescence resonance energy transfer (FRET) is a high-resolution technique that allows the characterization of spatial and temporal properties of biological structures and mechanisms. In this work, we developed an in silico single-molecule FRET methodology to study the dynamics of fluorophores inside lipid rafts. We monitored the fluorescence of a single acceptor molecule in the presence of several donor molecules. By looking at the average fluorescence, we selected events with single acceptor and donor molecules, and we used them to determine the raft size in the range of 5-16 nm. We conclude that our method is robust and insensitive to variations in the diffusion coefficient, donor density, or selected fluorescence threshold.


Subject(s)
Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Computer Simulation , Membrane Microdomains , Nanotechnology
3.
Eur J Pharmacol ; 899: 174026, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33722592

ABSTRACT

Riluzole is an anticonvulsant drug also used to treat the amyotrophic lateral sclerosis and major depressive disorder. This compound has antiglutamatergic activity and is an important multichannel blocker. However, little is known about its actions on the Kv4.2 channels, the molecular correlate of the A-type K+ current (IA) and the fast transient outward current (Itof). Here, we investigated the effects of riluzole on Kv4.2 channels transiently expressed in HEK-293 cells. Riluzole inhibited Kv4.2 channels with an IC50 of 190 ± 14 µM and the effect was voltage- and frequency-independent. The activation rate of the current (at +50 mV) was not affected by the drug, nor the voltage dependence of channel activation, but the inactivation rate was accelerated by 100 and 300 µM riluzole. When Kv4.2 channels were maintained at the closed state, riluzole incubation induced a tonic current inhibition. In addition, riluzole significantly shifted the voltage dependence of inactivation to hyperpolarized potentials without affecting the recovery from inactivation. In the presence of the drug, the closed-state inactivation was significantly accelerated, and the percentage of inactivated channels was increased. Altogether, our findings indicate that riluzole inhibits Kv4.2 channels mainly affecting the closed and closed-inactivated states.


Subject(s)
Potassium Channel Blockers/pharmacology , Riluzole/pharmacology , Shal Potassium Channels/antagonists & inhibitors , HEK293 Cells , Humans , Ion Channel Gating , Membrane Potentials , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Time Factors
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.
Biochem Pharmacol ; 177: 113961, 2020 07.
Article in English | MEDLINE | ID: mdl-32272111

ABSTRACT

It has been reported that muscarinic type-2 receptors (M2R) are voltage sensitive in an agonist-specific manner. In this work, we studied the effects of membrane potential on the interaction of M2R with the superagonist iperoxo (IXO), both functionally (using the activation of the ACh-gated K+ current (IKACh) in cardiomyocytes) and by molecular dynamics (MD) simulations. We found that IXO activated IKACh with remarkable high potency and clear voltage dependence, displaying a larger effect at the hyperpolarized potential. This result is consistent with a greater affinity, as validated by a slower (τ = 14.8 ± 2.3 s) deactivation kinetics of the IXO-evoked IKACh than that at the positive voltage (τ = 6.7 ± 1.2 s). The voltage-dependent M2R-IXO interaction induced IKACh to exhibit voltage-dependent features of this current, such as the 'relaxation gating' and the modulation of rectification. MD simulations revealed that membrane potential evoked specific conformational changes both at the external access and orthosteric site of M2R that underlie the agonist affinity change provoked by voltage on M2R. Moreover, our experimental data suggest that the 'tyrosine lid' (Y104, Y403, and Y426) is not the previously proposed voltage sensor of M2R. These findings provide an insight into the structural and functional framework of the biased signaling induced by voltage on GPCRs.


Subject(s)
Ion Channel Gating/drug effects , Isoxazoles/pharmacology , Molecular Dynamics Simulation , Quaternary Ammonium Compounds/pharmacology , Receptor, Muscarinic M2/physiology , Acetylcholine/pharmacology , Animals , Cats , Cells, Cultured , Electric Stimulation , Female , Ion Channel Gating/physiology , Male , Membrane Potentials/drug effects , Models, Molecular , Muscarinic Agonists/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Protein Conformation , Receptor, Muscarinic M2/chemistry , Receptor, Muscarinic M2/metabolism , Xenopus laevis
6.
Eur J Pharmacol ; 855: 262-266, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31082369

ABSTRACT

Chloroquine (CQ) is an old antimalarial drug currently being investigated for its anti-tumor properties. As chloroquine has been shown to inhibits several potassium channels, we decided to study its effect on the tumor-related Kv10.1 channel by using patch-clamp electrophysiology and cell migration assays. We found that chloroquine inhibited Kv10.1 channels transiently expressed in HEK-293 cells in a concentration- and voltage-dependent manner acting from the cytoplasmic side of the plasma membrane. Chloroquine also inhibited the outward potassium currents from MDA-MB-231 cells, which are mainly carried through Kv10.1 channels as was confirmed using astemizole. Additionally, chloroquine decreased MDA-MB-231 cell migration in the in vitro scratch wound healing assay. In conclusion, our data suggest that chloroquine decreases MDA-MB-231 cell migration by inhibiting Kv10.1 channels. The inhibition of Kv10.1 channels could represent another mechanism of the antitumoral action of chloroquine, besides autophagy inhibition and tumor vessel normalization.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/drug effects , Chloroquine/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , HEK293 Cells , Humans
7.
J Physiol Sci ; 69(3): 513-521, 2019 May.
Article in English | MEDLINE | ID: mdl-30900190

ABSTRACT

Voltage-gated potassium channels are expressed in a wide variety of excitable and non-excitable cells and regulate numerous cellular functions. The activity of ion channels can be modulated by direct interaction or/and functional coupling with other proteins including auxiliary subunits, scaffold proteins and the cytoskeleton. Here, we evaluated the influence of the actin-based cytoskeleton on the Kv2.1 channel using pharmacological and electrophysiological methods. We found that disruption of the actin-based cytoskeleton by latrunculin B resulted in the regulation of the Kv2.1 inactivation mechanism; it shifted the voltage of half-maximal inactivation toward negative potentials by approximately 15 mV, accelerated the rate of closed-state inactivation, and delayed the recovery rate from inactivation. The actin cytoskeleton stabilizing agent phalloidin prevented the hyperpolarizing shift in the half-maximal inactivation potential when co-applied with latrunculin B. Additionally, PIP2 depletion (a strategy that regulates Kv2.1 inactivation) after cytoskeleton disruption does not regulate further the inactivation of Kv2.1, which suggests that both factors could be regulating the Kv2.1 channel by a common mechanism. In summary, our results suggest a role for the actin-based cytoskeleton in regulating Kv2.1 channels.


Subject(s)
Cytoskeleton/drug effects , Cytoskeleton/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Shab Potassium Channels/metabolism , Actins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Potassium/metabolism , Potassium Channels, Voltage-Gated/metabolism , Thiazolidines/pharmacology
8.
Biochem Biophys Res Commun ; 503(4): 2531-2535, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30208521

ABSTRACT

The ether-à-go-go1 (EAG1, Kv10.1) K+ channel is a member of the voltage-gated K+ channel family mainly expressed in the central nervous system and cancer cells. Membrane lipids regulate several voltage-gated K+ channels but their influence on EAG1 channels has been poorly explored. Here we have studied the regulation of hEAG1 channels by phosphatidylinositol 4,5-bisfofate (PIP2) by using different strategies to manipulate the levels of this lipid, and the patch clamp technique. We found that depletion of endogenous PIP2 by activation of the voltage-sensing phosphatase from Danio rerio (Dr-VSP) or the human muscarinic type-1 receptor (hM1R) inhibits hEAG1 currents; however, the application of exogenous PIP2 to increase the level of this lipid on the plasma membrane, also induced an inhibition of hEAG1. In summary, our results indicate that PIP2 have dual effects on hEAG1 channels and its action as activator or inhibitor depends on its initial level on the plasma membrane.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Animals , Humans , Patch-Clamp Techniques , Phosphoric Monoester Hydrolases , Receptors, Muscarinic , Zebrafish
9.
Biochim Biophys Acta Biomembr ; 1860(5): 1242-1251, 2018 May.
Article in English | MEDLINE | ID: mdl-29474891

ABSTRACT

Kv7.2/Kv7.3 channels are the molecular correlate of the M-current, which stabilizes the membrane potential and controls neuronal excitability. Previous studies have shown the relevance of plasma membrane lipids on both M-currents and Kv7.2/Kv7.3 channels. Here, we report the sensitive modulation of Kv7.2/Kv7.3 channels by membrane cholesterol level. Kv7.2/Kv7.3 channels transiently expressed in HEK-293 cells were significantly inhibited by decreasing the cholesterol level in the plasma membrane by three different pharmacological strategies: methyl-ß-cyclodextrin (MßCD), Filipin III, and cholesterol oxidase treatment. Surprisingly, Kv7.2/Kv7.3 channels were also inhibited by membrane cholesterol loading with the MßCD/cholesterol complex. Depletion or enrichment of plasma membrane cholesterol differentially affected the biophysical parameters of the macroscopic Kv7.2/Kv7.3 currents. These results indicate a complex mechanism of Kv7.2/Kv7.3 channels modulation by membrane cholesterol. We propose that inhibition of Kv7.2/Kv7.3 channels by membrane cholesterol depletion involves a loss of a direct cholesterol-channel interaction. However, the inhibition of Kv7.2/Kv7.3 channels by membrane cholesterol enrichment could include an additional direct cholesterol-channel interaction, or changes in the physical properties of the plasma membrane. In summary, our results indicate that an optimum cholesterol level in the plasma membrane is required for the proper functioning of Kv7.2/Kv7.3 channels.


Subject(s)
Cholesterol/physiology , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/chemistry , Cholesterol/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , beta-Cyclodextrins/pharmacology
10.
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
11.
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
12.
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
13.
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
14.
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
15.
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
16.
J Pharmacol Toxicol Methods ; 69(3): 237-44, 2014.
Article in English | MEDLINE | ID: mdl-24412489

ABSTRACT

INTRODUCTION: Voltage- and state-dependent blocks are important mechanisms by which drugs affect voltage-gated ionic channels. However, spontaneous (i.e. drug-free) time-dependent changes in the activation and inactivation of hERG and Na(+) channels have been reported when using conventional whole-cell patch-clamp in HEK-293 cells. METHODS: hERG channels were heterologously expressed in HEK-293 cells and in Xenopus laevis oocytes. hERG current (IhERG) was recorded using both conventional and perforated whole-cell patch-clamp (HEK-293 cells), and two microelectrode voltage-clamp (Xenopus oocytes) in drug-free solution, and in the presence of the drug trazodone. RESULTS: In conventional whole-cell setup, we observed a spontaneous time-dependent hyperpolarizing shift in the activation curve of IhERG. Conversely, in perforated patch whole-cell (HEK-293 cells) or in two microelectrode voltage-clamp (Xenopus oocytes) activation curves of IhERG were very stable for periods ~50min. Voltage-dependent inactivation of IhERG was not significantly altered in the three voltage clamp configurations tested. When comparing voltage- and state-dependent effects of the antidepressant drug trazodone on IhERG, similar changes between the three voltage clamp configurations were observed as under drug-free conditions. DISCUSSION: The comparative analysis performed in this work showed that only under conventional whole-cell voltage-clamp conditions, a leftward shift in the activation curve of IhERG occurred, both in the presence and absence of drugs. These spontaneous time-dependent changes in the voltage activation gate of IhERG are a potential confounder in pharmacological studies on hERG channels expressed in HEK-293 cells.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Patch-Clamp Techniques/methods , Trazodone/pharmacology , Animals , Antidepressive Agents, Second-Generation , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Oocytes , Time Factors , Xenopus laevis
17.
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
18.
Proc Natl Acad Sci U S A ; 105(4): 1364-8, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18216262

ABSTRACT

Although chloroquine remains an important therapeutic agent for treatment of malaria in many parts of the world, its safety margin is very narrow. Chloroquine inhibits the cardiac inward rectifier K(+) current I(K1) and can induce lethal ventricular arrhythmias. In this study, we characterized the biophysical and molecular basis of chloroquine block of Kir2.1 channels that underlie cardiac I(K1). The voltage- and K(+)-dependence of chloroquine block implied that the binding site was located within the ion-conduction pathway. Site-directed mutagenesis revealed the location of the chloroquine-binding site within the cytoplasmic pore domain rather than within the transmembrane pore. Molecular modeling suggested that chloroquine blocks Kir2.1 channels by plugging the cytoplasmic conduction pathway, stabilized by negatively charged and aromatic amino acids within a central pocket. Unlike most ion-channel blockers, chloroquine does not bind within the transmembrane pore and thus can reach its binding site, even while polyamines remain deeper within the channel vestibule. These findings explain how a relatively low-affinity blocker like chloroquine can effectively block I(K1) even in the presence of high-affinity endogenous blockers. Moreover, our findings provide the structural framework for the design of safer, alternative compounds that are devoid of Kir2.1-blocking properties.


Subject(s)
Chloroquine/metabolism , Chloroquine/pharmacology , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Antimalarials/chemical synthesis , Antimalarials/metabolism , Antimalarials/pharmacology , Binding Sites/genetics , Cell Line , Cytoplasm/drug effects , Cytoplasm/genetics , Cytoplasm/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channel Blockers/chemical synthesis , Potassium Channels, Inwardly Rectifying/genetics , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Surface Properties , Transfection
19.
Eur J Pharmacol ; 531(1-3): 1-8, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16423345

ABSTRACT

Many drugs block delayed rectifier K+ channels and prolong the cardiac action potential duration. Here we investigate the molecular mechanisms of voltage-dependent block of human ether-a-go-go-related gene (HERG) K+ channels expressed in cells HEK-293 and Xenopus oocytes by maprotiline. The IC50 determined at 0 mV on HERG expressed HEK-293 cell and oocytes was 5.2 and 23.7 microM, respectively. Block of HERG expressed in oocytes by maprotiline was enhanced by progressive membrane depolarization and accompanied by a negative shift in the voltage dependence of channel activation. The potency of maprotiline was reduced 7-fold by point mutation of a key aromatic residue (F656T) and 3-fold for Y652A, both located in the S6 domain. The mutation Y652A inverted the voltage dependence of HERG channel block by maprotiline. Together, these results suggest that voltage-dependent block of HERG results from gating dependent changes in the accessibility of Y652, a critical component of the drug binding site.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Maprotiline/pharmacology , Animals , Binding Sites , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Electric Stimulation , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/physiology , Female , Gene Expression , Humans , Membrane Potentials/drug effects , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Point Mutation , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL