Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
1.
Commun Biol ; 6(1): 651, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37336943

ABSTRACT

The voltage-gated K+ channel plays a key role in atrial excitability, conducting the ultra-rapid rectifier K+ current (IKur) and contributing to the repolarization of the atrial action potential. In this study, we examine its regulation by hydrogen sulfide (H2S) in HL-1 cardiomyocytes and in HEK293 cells expressing human Kv1.5. Pacing induced remodeling resulted in shorting action potential duration, enhanced both Kv1.5 channel and H2S producing enzymes protein expression in HL-1 cardiomyocytes. H2S supplementation reduced these remodeling changes and restored action potential duration through inhibition of Kv1.5 channel. H2S also inhibited recombinant hKv1.5, lead to nitric oxide (NO) mediated S-nitrosylation and activated endothelial nitric oxide synthase (eNOS) by increased phosphorylation of Ser1177, prevention of NO formation precluded these effects. Regulation of Ikur by H2S has important cardiovascular implications and represents a novel and potential therapeutic target.


Subject(s)
Atrial Fibrillation , Hydrogen Sulfide , Potassium Channels, Voltage-Gated , Humans , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Atrial Fibrillation/metabolism , HEK293 Cells , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism
2.
Int J Mol Sci ; 24(12)2023 Jun 17.
Article in English | MEDLINE | ID: mdl-37373419

ABSTRACT

In this paper, the influence of external terahertz electromagnetic fields with different frequencies of 4 THz, 10 THz, 15 THz, and 20 THz on the permeability of the Kv1.2 voltage-gated potassium ion channel on the nerve cell membrane was studied using the combined model of the "Constant Electric Field-Ion Imbalance" method by molecular dynamics. We found that although the applied terahertz electric field does not produce strong resonance with the -C=O groups of the conservative sequence T-V-G-Y-G amino acid residue of the selective filter (SF) of the channel, it would affect the stability of the electrostatic bond between potassium ions and the carbonyl group of T-V-G-Y-G of SF, and it would affect the stability of the hydrogen bond between water molecules and oxygen atoms of the hydroxyl group of the 374THR side chain at the SF entrance, changing the potential and occupied states of ions in the SF and the occurrence probability of the permeation mode of ions and resulting in the change in the permeability of the channel. Compared with no external electric field, when the external electric field with 15 THz frequency is applied, the lifetime of the hydrogen bond is reduced by 29%, the probability of the "soft knock on" mode is decreased by 46.9%, and the ion flux of the channel is activated by 67.7%. Our research results support the view that compared to "direct knock-on", "soft knock-on" is a slower permeation mode.


Subject(s)
Electromagnetic Fields , Potassium Channels, Voltage-Gated , Potassium Channels, Voltage-Gated/metabolism , Molecular Dynamics Simulation , Ions/metabolism , Permeability , Potassium/metabolism , Kv1.2 Potassium Channel/chemistry , Kv1.5 Potassium Channel/metabolism
3.
Am J Respir Cell Mol Biol ; 69(2): 147-158, 2023 08.
Article in English | MEDLINE | ID: mdl-36917789

ABSTRACT

Reduced expression and/or activity of Kv1.5 channels (encoded by KCNA5) is a common hallmark in human or experimental pulmonary arterial hypertension (PAH). Likewise, genetic variants in KCNA5 have been found in patients with PAH, but their functional consequences and potential impact on the disease are largely unknown. Herein, this study aimed to characterize the functional consequences of seven KCNA5 variants found in a cohort of patients with PAH. Potassium currents were recorded by patch-clamp technique in HEK293 cells transfected with wild-type or mutant Kv1.5 cDNA. Flow cytometry, Western blot, and confocal microscopy techniques were used for measuring protein expression and cell apoptosis in HEK293 and human pulmonary artery smooth muscle cells. KCNA5 variants (namely, Arg184Pro and Gly384Arg) found in patients with PAH resulted in a clear loss of potassium channel function as assessed by electrophysiological and molecular modeling analyses. The Arg184Pro variant also resulted in a pronounced reduction of Kv1.5 expression. Transfection with Arg184Pro or Gly384Arg variants decreased apoptosis of human pulmonary artery smooth muscle cells compared with the wild-type cells, demonstrating that KCNA5 dysfunction in both variants affects cell viability. Thus, in addition to affecting channel activity, both variants were associated with impaired apoptosis, a crucial process linked to the disease. The estimated prevalence of dysfunctional KCNA5 variants in the PAH population analyzed was around 1%. The data indicate that some KCNA5 variants found in patients with PAH have critical consequences for channel function, supporting the idea that KCNA5 pathogenic variants may be a causative or contributing factor for PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/metabolism , HEK293 Cells , Hypertension, Pulmonary/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Pulmonary Artery/pathology
4.
J Biol Chem ; 298(11): 102467, 2022 11.
Article in English | MEDLINE | ID: mdl-36087839

ABSTRACT

Among voltage-gated potassium channel (KV) isoforms, KV1.6 is one of the most widespread in the nervous system. However, there are little data concerning its physiological significance, in part due to the scarcity of specific ligands. The known high-affinity ligands of KV1.6 lack selectivity, and conversely, its selective ligands show low affinity. Here, we present a designer peptide with both high affinity and selectivity to KV1.6. Previously, we have demonstrated that KV isoform-selective peptides can be constructed based on the simplistic α-hairpinin scaffold, and we obtained a number of artificial Tk-hefu peptides showing selective blockage of KV1.3 in the submicromolar range. We have now proposed amino acid substitutions to enhance their activity. As a result, we have been able to produce Tk-hefu-11 that shows an EC50 of ≈70 nM against KV1.3. Quite surprisingly, Tk-hefu-11 turns out to block KV1.6 with even higher potency, presenting an EC50 of ≈10 nM. Furthermore, we have solved the peptide structure and used molecular dynamics to investigate the determinants of selective interactions between artificial α-hairpinins and KV channels to explain the dramatic increase in KV1.6 affinity. Since KV1.3 is not highly expressed in the nervous system, we hope that Tk-hefu-11 will be useful in studies of KV1.6 and its functions.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Amino Acid Sequence , Potassium Channel Blockers/chemistry , Peptides/chemistry , Ligands , Protein Isoforms/genetics , Protein Isoforms/metabolism , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism
5.
Mol Biol Rep ; 49(7): 5939-5952, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35368226

ABSTRACT

BACKGROUND: Gout is usually found in patients with atrial fibrillation (AF). K+ efflux is a common trigger of NLRP3 inflammasome activation which is involved in the pathogenesis of AF. We investigated the role of the K+ channel Kv1.5 in monosodium urate crystal (MSU)-induced activation of the NLRP3 inflammasome and electrical remodeling in mouse and human macrophages J774.1 and THP-1, and mouse atrial myocytes HL-1. METHODS AND RESULTS: Macrophages, primed with lipopolysaccharide (LPS), were stimulated by MSU. HL-1 cells were incubated with the conditioned medium (CM) from MSU-stimulated macrophages. Western blot, ELISA and patch clamp were used. MSU induced caspase-1 expression in LPS-primed J774.1 cells and IL-1ß secretion, suggesting NLRP3 inflammasome activation. A selective Kv1.5 inhibitor, diphenyl phosphine oxide-1 (DPO-1), and siRNAs against Kv1.5 suppressed the levels of caspase-1 and IL-1ß. MSU reduced intracellular K+ concentration which was prevented by DPO-1 and siRNAs against Kv1.5. MSU increased expression of Hsp70, and Kv1.5 on the plasma membrane. siRNAs against Hsp70 were suppressed but heat shock increased the expression of Hsp70, caspase-1, IL-1ß, and Kv1.5 in MSU-stimulated J774.1 cells. The CM from MSU-stimulated macrophages enhanced the expression of caspase-1, IL-1ß and Kv1.5 with increased Kv1.5-mediated currents that shortened action potential duration in HL-1 cells. These responses were abolished by DPO-1 and a siRNA against Kv1.5. CONCLUSIONS: Kv1.5 regulates MSU-induced activation of NLRP3 inflammasome in macrophages. MSUrelated activation of NLRP3 inflammasome and electrical remodeling in HL-1 cells are via macrophages. Kv1.5 may have therapeutic value for diseases related to gout-induced activation of the NLRP3 inflammsome, including AF.


Subject(s)
Atrial Remodeling , Gout , Kv1.5 Potassium Channel/metabolism , Animals , Caspase 1/metabolism , Gout/drug therapy , Gout/metabolism , Gout/pathology , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/metabolism , Uric Acid/pharmacology
6.
J Enzyme Inhib Med Chem ; 37(1): 462-471, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35012386

ABSTRACT

Kv1.5 potassium channel, encoded by KCNA5, is a promising target for the treatment of atrial fibrillation, one of the common arrhythmia. A new series of arylmethylpiperidines derivatives based on DDO-02001 were synthesised and evaluated for their ability to inhibit Kv1.5 channel. Among them, compound DDO-02005 showed good inhibitory activity (IC50 = 0.72 µM), preferable anti-arrhythmic effects and favoured safety. These results indicate that DDO-02005 can be a promising Kv1.5 inhibitor for further studies.


Subject(s)
Drug Design , Kv1.5 Potassium Channel/antagonists & inhibitors , Piperidines/pharmacology , Potassium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Humans , Kv1.5 Potassium Channel/metabolism , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Structure-Activity Relationship
7.
Cells ; 10(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34572065

ABSTRACT

Both inherited and acquired cardiac arrhythmias are often associated with the abnormal functional expression of ion channels at the cellular level. The complex machinery that continuously traffics, anchors, organizes, and recycles ion channels at the plasma membrane of a cardiomyocyte appears to be a major source of channel dysfunction during cardiac arrhythmias. This has been well established with the discovery of mutations in the genes encoding several ion channels and ion channel partners during inherited cardiac arrhythmias. Fibrosis, altered myocyte contacts, and post-transcriptional protein changes are common factors that disorganize normal channel trafficking during acquired cardiac arrhythmias. Channel availability, described notably for hERG and KV1.5 channels, could be another potent arrhythmogenic mechanism. From this molecular knowledge on cardiac arrhythmias will emerge novel antiarrhythmic strategies.


Subject(s)
Arrhythmias, Cardiac/pathology , Cell Membrane/physiology , ERG1 Potassium Channel/metabolism , Ion Channels/physiology , Kv1.5 Potassium Channel/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Biological Transport , Humans
8.
Eur J Pharmacol ; 904: 174158, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33971179

ABSTRACT

Voltage-dependent K+ (Kv) channels play the role of returning the membrane potential to the resting state, thereby maintaining the vascular tone. Here, we used native smooth-muscle cells from rabbit coronary arteries to investigate the inhibitory effect of lorcainide, a class Ic antiarrhythmic agent, on Kv channels. Lorcainide inhibited Kv channels in a concentration-dependent manner with an IC50 of 4.46 ± 0.15 µM and a Hill coefficient of 0.95 ± 0.01. Although application of lorcainide did not change the activation curve, it shifted the inactivation curve toward a more negative potential, implying that lorcainide inhibits Kv channels by changing the channels' voltage sensors. The recovery time constant from channel inactivation increased in the presence of lorcainide. Furthermore, application of train steps (of 1 or 2 Hz) in the presence of lorcainide progressively augmented the inhibition of Kv currents, implying that lorcainide-induced inhibition of Kv channels is use (state)-dependent. Pretreatment with Kv1.5 or Kv2.1/2.2 inhibitors effectively reduced the amplitude of the Kv current but did not affect the inhibitory effect of lorcainide. Based on these results, we conclude that lorcainide inhibits vascular Kv channels in a concentration and use (state)-dependent manner by changing their inactivation gating properties. Considering the clinical efficacy of lorcainide, and the pathophysiological significance of vascular Kv channels, our findings should be considered when prescribing lorcainide to patients with arrhythmia and vascular disease.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzeneacetamides/pharmacology , Coronary Vessels/metabolism , Muscle, Smooth, Vascular/metabolism , Piperidines/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Animals , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Kinetics , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/metabolism , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Rabbits , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/metabolism
9.
J Biol Chem ; 296: 100514, 2021.
Article in English | MEDLINE | ID: mdl-33676894

ABSTRACT

The voltage-gated potassium channel Kv1.5 plays important roles in the repolarization of atrial action potentials and regulation of the vascular tone. While the modulation of Kv1.5 function has been well studied, less is known about how the protein levels of Kv1.5 on the cell membrane are regulated. Here, through electrophysiological and biochemical analyses of Kv1.5 channels heterologously expressed in HEK293 cells and neonatal rat ventricular myocytes, as well as native Kv1.5 in human induced pluripotent stem cell (iPSC)-derived atrial cardiomyocytes, we found that activation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate (PMA, 10 nM) diminished Kv1.5 current (IKv1.5) and protein levels of Kv1.5 in the plasma membrane. Mechanistically, PKC activation led to monoubiquitination and degradation of the mature Kv1.5 proteins. Overexpression of Vps24, a protein that sorts transmembrane proteins into lysosomes via the multivesicular body (MVB) pathway, accelerated, whereas the lysosome inhibitor bafilomycin A1 completely prevented PKC-mediated Kv1.5 degradation. Kv1.5, but not Kv1.1, Kv1.2, Kv1.3, or Kv1.4, was uniquely sensitive to PMA treatment. Sequence alignments suggested that residues within the N terminus of Kv1.5 are essential for PKC-mediated Kv1.5 reduction. Using N-terminal truncation as well as site-directed mutagenesis, we identified that Thr15 is the target site for PKC that mediates endocytic degradation of Kv1.5 channels. These findings indicate that alteration of protein levels in the plasma membrane represents an important regulatory mechanism of Kv1.5 channel function under PKC activation conditions.


Subject(s)
Endocytosis , Induced Pluripotent Stem Cells/metabolism , Kv1.5 Potassium Channel/metabolism , Protein Kinase C/metabolism , Ubiquitination , Animals , Animals, Newborn , Humans , Induced Pluripotent Stem Cells/cytology , Kv1.5 Potassium Channel/genetics , Phosphorylation , Protein Kinase C/genetics , Rats , Signal Transduction
10.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669857

ABSTRACT

(1) Background: As membrane channels contribute to different cell functions, understanding the underlying mechanisms becomes extremely important. A large number of neuronal channels have been investigated, however, less studied are the channels expressed in the glia population, particularly in microglia. In the present study, we focused on the function of the Kv1.3, Kv1.5 and Kir2.1 potassium channels expressed in both BV2 cells and primary microglia cultures, which may impact the cellular migration process. (2) Methods: Using an immunocytochemical approach, we were able to show the presence of the investigated channels in BV2 microglial cells, record their currents using a patch clamp and their role in cell migration using the scratch assay. The migration of the primary microglial cells in culture was assessed using cell culture inserts. (3) Results: By blocking each potassium channel, we showed that Kv1.3 and Kir2.1 but not Kv1.5 are essential for BV2 cell migration. Further, primary microglial cultures were obtained from a line of transgenic CX3CR1-eGFP mice that express fluorescent labeled microglia. The mice were subjected to a spared nerve injury model of pain and we found that microglia motility in an 8 µm insert was reduced 2 days after spared nerve injury (SNI) compared with sham conditions. Additional investigations showed a further impact on cell motility by specifically blocking Kv1.3 and Kir2.1 but not Kv1.5; (4) Conclusions: Our study highlights the importance of the Kv1.3 and Kir2.1 but not Kv1.5 potassium channels on microglia migration both in BV2 and primary cell cultures.


Subject(s)
Cell Movement , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Microglia/cytology , Microglia/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Cell Line , Electrophysiological Phenomena , Mice, Transgenic , Nerve Tissue/injuries , Nerve Tissue/pathology
11.
Acta Biochim Pol ; 68(1): 83-89, 2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33533580

ABSTRACT

Abnormal proliferation of granulosa cells is implicated in ovarian dysfunction and dysregulated folliculogenesis in the polycystic ovary syndrome (PCOS). Aberrant microRNA (miRNA) expression might contribute to disordered folliculogenesis and granulosa cell proliferation in PCOS. This study aimed to investigate the roles of miR-3188 in ovarian dysfunction, as well as the mechanism involved in granulosa cell proliferation in PCOS. Firstly, peripheral blood samples were isolated from PCOS patients and healthy controls, and qRT-PCR analysis demonstrated a dramatic increase in miR-3188 in PCOS patients when compared to the healthy controls. Secondly, miR-3188 overexpression increased cell viability of the granulosa-like tumor cell line (KGN). However, cell viability of KGN was repressed by interference with miR-3188. MiR-3188 promoted cell cycle of KGN through increasing cyclinD1 and decreasing p21 levels. Moreover, cell apoptosis was suppressed by miR-3188 in KGN, indicated by enhanced Bcl-2, and reduced Bax and cleaved caspase-3 levels, whereas knockdown of miR-3188 resulted in opposite effects. Lastly, potassium voltage-gated channel subfamily A member 5 (KCNA5) was verified as a target of miR-3188. KCNA5 expression was decreased and displayed negative correlation with miR-3188 levels in PCOS patients. Overexpression of KCNA5 attenuated the promotive effects of miR-3188 on cell viability and cell cycle in KGN. In conclusion, miR-3188, a key miRNA enhanced in PCOS, promoted granulosa cell proliferation through down-regulation of KCNA5, providing a new therapeutic target for PCOS.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Granulosa Cells/metabolism , Kv1.5 Potassium Channel/metabolism , MicroRNAs/metabolism , Polycystic Ovary Syndrome/blood , Signal Transduction/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/genetics , Female , Gene Knockdown Techniques , Humans , Kv1.5 Potassium Channel/genetics , MicroRNAs/genetics , Polycystic Ovary Syndrome/pathology , Transfection
12.
Cardiovasc Res ; 117(7): 1746-1759, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33523143

ABSTRACT

AIMS: Obesity, an established risk factor of atrial fibrillation (AF), is frequently associated with enhanced inflammatory response. However, whether inflammatory signaling is causally linked to AF pathogenesis in obesity remains elusive. We recently demonstrated that the constitutive activation of the 'NACHT, LRR, and PYD Domains-containing Protein 3' (NLRP3) inflammasome promotes AF susceptibility. In this study, we hypothesized that the NLRP3 inflammasome is a key driver of obesity-induced AF. METHODS AND RESULTS: Western blotting was performed to determine the level of NLRP3 inflammasome activation in atrial tissues of obese patients, sheep, and diet-induced obese (DIO) mice. The increased body weight in patients, sheep, and mice was associated with enhanced NLRP3-inflammasome activation. To determine whether NLRP3 contributes to the obesity-induced atrial arrhythmogenesis, wild-type (WT) and NLRP3 homozygous knockout (NLRP3-/-) mice were subjected to high-fat-diet (HFD) or normal chow (NC) for 10 weeks. Relative to NC-fed WT mice, HFD-fed WT mice were more susceptible to pacing-induced AF with longer AF duration. In contrast, HFD-fed NLRP3-/- mice were resistant to pacing-induced AF. Optical mapping in DIO mice revealed an arrhythmogenic substrate characterized by abbreviated refractoriness and action potential duration (APD), two key determinants of reentry-promoting electrical remodeling. Upregulation of ultra-rapid delayed-rectifier K+-channel (Kv1.5) contributed to the shortening of atrial refractoriness. Increased profibrotic signaling and fibrosis along with abnormal Ca2+ release from sarcoplasmic reticulum (SR) accompanied atrial arrhythmogenesis in DIO mice. Conversely, genetic ablation of Nlrp3 (NLRP3-/-) in HFD-fed mice prevented the increases in Kv1.5 and the evolution of electrical remodeling, the upregulation of profibrotic genes, and abnormal SR Ca2+ release in DIO mice. CONCLUSION: These results demonstrate that the atrial NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmogenesis and establishes a mechanistic link between obesity-induced AF and NLRP3-inflammasome activation.


Subject(s)
Atrial Fibrillation/etiology , Heart Atria/metabolism , Heart Rate , Inflammasomes/metabolism , Inflammation/etiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/complications , Action Potentials , Aged , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Calcium Signaling , Case-Control Studies , Disease Models, Animal , Female , Heart Atria/physiopathology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Obesity/metabolism , Obesity/physiopathology , Sheep, Domestic
13.
Int J Mol Sci ; 22(3)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572906

ABSTRACT

KV1.5 channel function is modified by different regulatory subunits. KVß1.3 subunits assemble with KV1.5 channels and induce a fast and incomplete inactivation. Inhibition of PKC abolishes the KVß1.3-induced fast inactivation, decreases the amplitude of the current KV1.5-KVß1.3 and modifies their pharmacology likely due to changes in the traffic of KV1.5-KVß1.3 channels in a PKC-dependent manner. In order to analyze this hypothesis, HEK293 cells were transfected with KV1.5-KVß1.3 channels, and currents were recorded by whole-cell configuration of the patch-clamp technique. The presence of KV1.5 in the membrane was analyzed by biotinylation techniques, live cell imaging and confocal microscopy approaches. PKC inhibition resulted in a decrease of 33 ± 7% of channels in the cell surface due to reduced recycling to the plasma membrane, as was confirmed by confocal microscopy. Live cell imaging indicated that PKC inhibition almost abolished the recycling of the KV1.5-KVß1.3 channels, generating an accumulation of channels into the cytoplasm. All these results suggest that the trafficking regulation of KV1.5-KVß1.3 channels is dependent on phosphorylation by PKC and, therefore, they could represent a clinically relevant issue, mainly in those diseases that exhibit modifications in PKC activity.


Subject(s)
Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Protein Kinase C/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , Phosphorylation , Protein Kinase C/antagonists & inhibitors
14.
Toxicol Appl Pharmacol ; 411: 115365, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33316272

ABSTRACT

Polychlorinated biphenyls (PCBs) are persistent and serious organic pollutants and can theoretically form 209 congeners. PCBs can be divided into two categories: dioxin-like (DL) and non-DL (NDL). NDL-PCBs, which lack aryl hydrocarbon receptor affinity, have been shown to perturb the functions of Jurkat T cells, cerebellar granule cells, and uterine cells. Kv1.3 and Kv1.5 channels are important in immune and heart functions, respectively. We investigated the acute effects of 2,2',6-trichlorinated biphenyl (PCB19), an NDL-PCB, on the currents of human Kv1.3 and Kv1.5 channels. PCB19 acutely blocked the Kv1.3 peak currents concentration-dependently with an IC50 of ~2 µM, without changing the steady-state current. The PCB19-induced inhibition of the Kv1.3 peak current occurred rapidly and voltage-independently, and the effect was irreversible, excluding the possibility of genomic regulation. PCB19 increased the time constants of both activation and inactivation of Kv1.3 channels, resulting in the slowing down of both ultra-rapid activation and intrinsic inactivation. However, PCB19 failed to alter the steady-state curves of activation and inactivation. Regarding the Kv1.5 channel, PCB19 affected neither the peak current nor the steady-state current at the same concentrations tested in the Kv1.3 experiments, showing selective inhibition of PCB19 on the Kv1.3 than the Kv1.5. The presented data indicate that PCB19 could acutely affect the human Kv1.3 channel through a non-genomic mechanism, possibly causing toxic effects on various human physiological functions related to the Kv1.3 channel, such as immune and neural systems.


Subject(s)
Environmental Pollutants/toxicity , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/drug effects , Polychlorinated Biphenyls/toxicity , Potassium Channel Blockers/toxicity , Animals , Dose-Response Relationship, Drug , Female , Humans , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Membrane Potentials , Oocytes , Time Factors , Xenopus laevis
15.
Eur J Pharmacol ; 891: 173707, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33137332

ABSTRACT

Darifenacin, an anticholinergic agent, has been used to treat overactive bladder syndrome. Despite its extensive clinical use, there is little information about the effect of darifenacin on vascular ion channels, specifically K+ channels. This study aimed to investigate the effect of the anti-muscarinic drug darifenacin on voltage-gated K+ (Kv) channels, vascular contractility, and coronary blood flow in rabbit coronary arteries. We used the whole-cell patch-clamp technique to evaluate the effect of darifenacin on Kv channels. Darifenacin inhibited the Kv current in a concentration-dependent manner. Applying 1 µM darifenacin shifted the activation and inactivation curves toward a more positive and negative potential, respectively. Darifenacin slowed the time constants of recovery from inactivation. Furthermore, blockade of the Kv current with darifenacin was increased gradually by applying a train of pulses, indicating that darifenacin inhibited Kv currents in a use- (state)-dependent manner. The darifenacin-mediated inhibition of Kv currents was associated with the Kv1.5 subtype, not the Kv2.1 or Kv7 subtype. Applying another anti-muscarinic drug atropine or ipratropium did not affect the Kv current or change the inhibitory effect of darifenacin. Isometric organ bath experiments using isolated coronary arteries were applied to evaluate whether darifenacin-induced inhibition of the Kv channel causes vasocontraction. Darifenacin substantially induced vasocontraction. Furthermore, darifenacin caused membrane depolarization and decreased coronary blood flow. From these results, we concluded that darifenacin inhibits the Kv currents in concentration- and use- (state)-dependent manners. Inhibition of the Kv current with darifenacin occurred by shifting the steady-state activation and inactivation curves regardless of its anti-muscarinic effect.


Subject(s)
Benzofurans/pharmacology , Coronary Vessels/drug effects , Kv1.5 Potassium Channel/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Potassium Channel Blockers/pharmacology , Pyrrolidines/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Kinetics , Kv1.5 Potassium Channel/metabolism , Male , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rabbits
16.
BMC Cancer ; 20(1): 1179, 2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33267786

ABSTRACT

BACKGROUND: Gastric and colorectal cancers are the most common malignant tumours, leading to a significant number of cancer-related deaths worldwide. Recently, increasing evidence has demonstrated that cancer cells exhibit a differential expression of potassium channels and this can contribute to cancer progression. However, their expression and localisation at the somatic level remains uncertain. In this study, we have investigated the expression levels of KCNB1 and KCNA5 genes encoding ubiquitous Kv2.1 and Kv1.5 potassium channels in gastric and colorectal tumours. METHODS: Gastric and colorectal tumoral and peritumoral tissues were collected to evaluate the expression of KCNB1 and KCNA5 mRNA by quantitative PCR. Moreover, the immunohistochemical staining profile of Kv2.1 and Kv1.5 was assessed on 40 Formalin-Fixed and Paraffin-Embedded (FFPE) gastric carcinoma tissues. Differences in gene expression between tumoral and peritumoral tissues were compared statistically with the Mann-Whitney U test. The association between the clinicopathological features of the GC patients and the expression of both Kv proteins was investigated with χ2 and Fisher's exact tests. RESULTS: The mRNA fold expression of KCNB1 and KCNA5 genes showed a lower mean in the tumoral tissues (0.06 ± 0.17, 0.006 ± 0.009) compared to peritumoral tissues (0.08 ± 0.16, 0.16 ± 0.48, respectively) without reaching the significance rate (p = 0.861, p = 0.152, respectively). Interestingly, Kv2.1 and Kv1.5 immunostaining was detectable and characterised by a large distribution in peritumoral and tumoral epithelial cells. More interestingly, inflammatory cells were also stained. Surprisingly, Kv2.1 and Kv1.5 staining was undoubtedly and predominantly detected in the cytoplasm compartment of tumour cells. Indeed, the expression of Kv2.1 in tumour cells revealed a significant association with the early gastric cancer clinical stage (p = 0.026). CONCLUSION: The data highlight, for the first time, the potential role of Kv1.5 and Kv2.1 in gastrointestinal-related cancers and suggests they may be promising prognostic markers for these tumours.


Subject(s)
Colorectal Neoplasms/genetics , Kv1.5 Potassium Channel/metabolism , Shab Potassium Channels/metabolism , Stomach Neoplasms/genetics , Female , Humans , Male , Middle Aged , Retrospective Studies
17.
Proc Natl Acad Sci U S A ; 117(43): 27016-27021, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33051293

ABSTRACT

The opening and closing of voltage-gated ion channels are regulated by voltage sensors coupled to a gate that controls the ion flux across the cellular membrane. Modulation of any part of gating constitutes an entry point for pharmacologically regulating channel function. Here, we report on the discovery of a large family of warfarin-like compounds that open the two voltage-gated type 1 potassium (KV1) channels KV1.5 and Shaker, but not the related KV2-, KV4-, or KV7-type channels. These negatively charged compounds bind in the open state to positively charged arginines and lysines between the intracellular ends of the voltage-sensor domains and the pore domain. This mechanism of action resembles that of endogenous channel-opening lipids and opens up an avenue for the development of ion-channel modulators.


Subject(s)
Ion Channel Gating , Kv1.5 Potassium Channel/agonists , Shaker Superfamily of Potassium Channels/agonists , Animals , High-Throughput Screening Assays , Kv1.5 Potassium Channel/metabolism , Molecular Docking Simulation , Patch-Clamp Techniques , Shaker Superfamily of Potassium Channels/metabolism , Xenopus laevis
18.
Biomolecules ; 10(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32882918

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare and severe cardiopulmonary disease without curative treatments. PAH is a multifactorial disease that involves genetic predisposition, epigenetic factors, and environmental factors (drugs, toxins, viruses, hypoxia, and inflammation), which contribute to the initiation or development of irreversible remodeling of the pulmonary vessels. The recent identification of loss-of-function mutations in KCNK3 (KCNK3 or TASK-1) and ABCC8 (SUR1), or gain-of-function mutations in ABCC9 (SUR2), as well as polymorphisms in KCNA5 (Kv1.5), which encode two potassium (K+) channels and two K+ channel regulatory subunits, has revived the interest of ion channels in PAH. This review focuses on KCNK3, SUR1, SUR2, and Kv1.5 channels in pulmonary vasculature and discusses their pathophysiological contribution to and therapeutic potential in PAH.


Subject(s)
Potassium Channels/metabolism , Pulmonary Arterial Hypertension/etiology , Animals , Drug Delivery Systems , Humans , Kv1.5 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Pulmonary Arterial Hypertension/drug therapy , Sulfonylurea Receptors/metabolism
19.
Mar Drugs ; 18(8)2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32823677

ABSTRACT

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Subject(s)
Mollusk Venoms/pharmacology , Potassium Channel Blockers/pharmacology , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Animals , Conus Snail , Ion Channel Gating , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/genetics , Kv1.4 Potassium Channel/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Kv1.6 Potassium Channel/antagonists & inhibitors , Kv1.6 Potassium Channel/genetics , Kv1.6 Potassium Channel/metabolism , Membrane Potentials , Oocytes , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism , Xenopus laevis
20.
BMC Cardiovasc Disord ; 20(1): 337, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32664860

ABSTRACT

BACKGROUND: High blood glucose impairs voltage-gated K+ (Kv) channel-mediated vasodilation in rat coronary artery smooth muscle cells (CSMCs) via oxidative stress. Advanced glycation end product (AGE) and receptor for AGE (RAGE) axis has been found to impair coronary dilation by reducing Kv channel activity in diabetic rat small coronary arteries (RSCAs). However, its underlying mechanism remain unclear. Here, we used isolated arteries and primary CSMCs to investigate the effect of AGE incubation on Kv channel-mediated coronary dilation and the possible involvement of peroxisome proliferators-activated receptor (PPAR) -γ pathway. METHODS: The RSCAs and primary CSMCs were isolated, cultured, and treated with bovine serum albumin (BSA), AGE-BSA, alagrebrium (ALA, AGE cross-linking breaker), pioglitazone (PIO, PPAR-γ activator) and/or GW9662 (PPAR-γ inhibitor). The groups were accordingly divided as control, BSA, AGE, AGE + ALA, AGE + PIO, or AGE + PIO + GW9662. Kv channel-mediated dilation was analyzed using wire myograph. Histology and immunohistochemistry of RSCAs were performed. Western blot was used to detect the protein expression of RAGE, major Kv channel subunits expressed in CSMCs (Kv1.2 and Kv1.5), PPAR-γ, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-2 (NOX-2). RESULTS: AGE markedly reduced Forskolin-induced Kv channel-mediated dilation of RSCAs by engaging with RAGE, and ALA or PIO significantly reversed the functional loss of Kv channel. In both RSCAs and CSMCs, AGE reduced Kv1.2/1.5 expression, increased RAGE and NOX-2 expression, and inhibited PPAR-γ expression, while ALA or PIO treatment partially reversed the inhibiting effects of AGE on Kv1.2/1.5 expression, accompanied by the downregulation of RAGE and decreased oxidative stress. Meanwhile, silencing of RAGE with siRNA remarkably alleviated the AGE-induced downregulation of Kv1.2/1.5 expression in CSMCs. CONCLUSION: AGE reduces the Kv channel expression in CSMCs and further impairs the Kv channel-mediated dilation in RSCAs. The AGE/RAGE axis may enhance oxidative stress by inhibiting the downstream PPAR-γ pathway, thus playing a critical role in the dysfunction of Kv channels.


Subject(s)
Glycation End Products, Advanced/pharmacology , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , PPAR gamma/metabolism , Serum Albumin, Bovine/pharmacology , Vasodilation/drug effects , Anilides/pharmacology , Animals , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Kv1.2 Potassium Channel/genetics , Kv1.5 Potassium Channel/genetics , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , PPAR gamma/drug effects , Pioglitazone/pharmacology , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...