Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 186
Filtrer
1.
J Biol Chem ; 300(7): 107483, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38897569

RÉSUMÉ

The voltage-gated Kv1.5 potassium channel, conducting the ultra-rapid delayed rectifier K+ current (IKur) in human cells, plays important roles in the repolarization of atrial action potentials and regulation of the vascular tone. We previously reported that activation of protein kinase C (PKC) by phorbol 12-myristate 13-acetate (PMA) induces endocytic degradation of cell-surface Kv1.5 channels, and a point mutation removing the phosphorylation site, T15A, in the N terminus of Kv1.5 abolished the PMA-effect. In the present study, using mutagenesis, patch clamp recording, Western blot analysis, and immunocytochemical staining, we demonstrate that ubiquitination is involved in the PMA-mediated degradation of mature Kv1.5 channels. Since the expression of the Kv1.4 channel is unaffected by PMA treatment, we swapped the N- and/or C-termini between Kv1.5 and Kv1.4. We found that the N-terminus alone did not but both N- and C-termini of Kv1.5 did confer PMA sensitivity to mature Kv1.4 channels, suggesting the involvement of Kv1.5 C-terminus in the channel ubiquitination. Removal of each of the potential ubiquitination residue Lysine at position 536, 565, and 591 by Arginine substitution (K536R, K565R, and K591R) had little effect, but removal of all three Lysine residues with Arginine substitution (3K-R) partially reduced PMA-mediated Kv1.5 degradation. Furthermore, removing the cysteine residue at position 604 by Serine substitution (C604S) drastically reduced PMA-induced channel degradation. Removal of the three Lysines and Cys604 with a quadruple mutation (3K-R/C604S) or a truncation mutation (Δ536) completely abolished the PKC activation-mediated degradation of Kv1.5 channels. These results provide mechanistic insight into PKC activation-mediated Kv1.5 degradation.


Sujet(s)
Canal potassique Kv1.5 , Protéine kinase C , Protéolyse , 12-Myristate-13-acétate de phorbol , Ubiquitination , Canal potassique Kv1.5/métabolisme , Canal potassique Kv1.5/génétique , Humains , Protéine kinase C/métabolisme , Protéine kinase C/génétique , 12-Myristate-13-acétate de phorbol/pharmacologie , Cellules HEK293 , Animaux , Phosphorylation , Membrane cellulaire/métabolisme , Canal potassique Kv1.4/métabolisme , Canal potassique Kv1.4/génétique
2.
Commun Biol ; 6(1): 651, 2023 06 19.
Article de Anglais | MEDLINE | ID: mdl-37336943

RÉSUMÉ

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.


Sujet(s)
Fibrillation auriculaire , Sulfure d'hydrogène , Canaux potassiques voltage-dépendants , Humains , Sulfure d'hydrogène/pharmacologie , Sulfure d'hydrogène/métabolisme , Fibrillation auriculaire/métabolisme , Cellules HEK293 , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Myocytes cardiaques/métabolisme
3.
Am J Respir Cell Mol Biol ; 69(2): 147-158, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-36917789

RÉSUMÉ

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.


Sujet(s)
Hypertension pulmonaire , Hypertension artérielle pulmonaire , Humains , Hypertension artérielle pulmonaire/métabolisme , Cellules HEK293 , Hypertension pulmonaire/métabolisme , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Hypertension artérielle pulmonaire primitive familiale/métabolisme , Artère pulmonaire/anatomopathologie
4.
Cell Biol Int ; 46(11): 1834-1840, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-35870168

RÉSUMÉ

The objective of the study was to investigate the expression levels of potassium voltage-gated channel subfamily A member 5 (KCNA5), connexin 43 (Cx43), and connexin 40 (Cx40) in the left atrial appendage of patients with atrial fibrillation (AF) and the interactions between them. We gathered tissue samples from patients with persistent AF and sinus rhythm and used fluorescence quantitative polymerase chain reaction to evaluate messenger RNA (mRNA) changes of KCNA5, Cx43, and Cx40. Then, we studied the protein levels of KCNA5, Cx43, and Cx40 by immunofluorescence and western blot analysis and the interactions between these proteins were identified by immunoprecipitation and immunofluorescence colocation, respectively. Compared with the control group, the mRNA and protein levels of KCNA5, Cx43, and Cx40 in the AF group were decreased and the positive expression of KCNA5, Cx43, and Cx40 protein was also decreased by immunofluorescence staining in the AF group. In addition, immunoprecipitation and immunofluorescence colocation revealed that KCNA5 was coexpressed with Cx43 and Cx40 proteins. The expressions of KCNA5, Cx43, and Cx40 were substantially downregulated in the myocardium of patients with AF and KCNA5 interacted with Cx43 and Cx40 proteins, respectively.


Sujet(s)
Fibrillation auriculaire , Connexine 43 , Connexines/métabolisme , Fibrillation auriculaire/génétique , Fibrillation auriculaire/métabolisme , Connexine 43/génétique , Connexine 43/métabolisme , Humains , Canal potassique Kv1.5/génétique , Myocarde/métabolisme , Potassium/métabolisme , ARN messager/génétique
5.
Sci Rep ; 11(1): 15829, 2021 08 04.
Article de Anglais | MEDLINE | ID: mdl-34349187

RÉSUMÉ

The dysregulation of K+ channels is a hallmark of pulmonary arterial hypertension (PAH). Herein, the channelome was analyzed in lungs of patients with PAH in a public transcriptomic database. Sixty six (46%) mRNA encoding cationic channels were dysregulated in PAH with most of them downregulated (83%). The principal component analysis indicated that dysregulated cationic channel expression is a signature of the disease. Changes were very similar in idiopathic, connective tissue disease and congenital heart disease associated PAH. This analysis 1) is in agreement with the widely recognized pathophysiological role of TASK1 and KV1.5, 2) supports previous preliminary reports pointing to the dysregulation of several K+ channels including the downregulation of KV1.1, KV1.4, KV1.6, KV7.1, KV7.4, KV9.3 and TWIK2 and the upregulation of KCa1.1 and 3) points to other cationic channels dysregulated such as Kv7.3, TALK2, CaV1 and TRPV4 which might play a pathophysiological role in PAH. The significance of other changes found in Na+ and TRP channels remains to be investigated.


Sujet(s)
Cations/métabolisme , Régulation de l'expression des gènes , Canaux ioniques/génétique , Hypertension artérielle pulmonaire/génétique , Transcriptome , Analyse de profil d'expression de gènes , Humains , Canal potassique Kv1.5/génétique , Hypertension artérielle pulmonaire/anatomopathologie
6.
Methodist Debakey Cardiovasc J ; 17(1): 65-67, 2021 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-34104323

RÉSUMÉ

Loperamide, a µ-opioid receptor agonist, can cause cardiotoxicity by inhibiting the potassium ion channel and slowing cardiomyocyte repolarization. This, in turn, can lead to frequent early afterdepolarizations, the most common mechanism of drug-induced long QT syndrome and torsades de pointes. Apical hypertrophic cardiomyopathy (AHCM) is a nonobstructive hypertrophic cardiomyopathy rarely associated with malignant arrhythmias. We present a case of loperamide-induced malignant ventricular arrhythmia revealing underlying AHCM in a 25-year-old woman with a history of sudden cardiac arrest (SCA) and opioid use. It is important to evaluate for structural heart disease in all patients presenting with SCA, regardless of presumed etiology such as drug-induced cardiotoxicity, to prevent missed opportunities for adequate treatment. Furthermore, the diagnosis of AHCM in SCA warrants further genetic evaluation for variances with a predilection for malignant arrhythmias.


Sujet(s)
Antidiarrhéiques/effets indésirables , Cardiomyopathie hypertrophique/génétique , Canal potassique Kv1.5/génétique , Lopéramide/effets indésirables , Mutation , Troubles liés aux opiacés/complications , Tachycardie ventriculaire/étiologie , Adulte , Cardiomyopathie hypertrophique/complications , Cardiomyopathie hypertrophique/diagnostic , Cardiomyopathie hypertrophique/physiopathologie , Cardiotoxicité , Analyse de mutations d'ADN , Électrocardiographie , Femelle , Prédisposition génétique à une maladie , Humains , Imagerie par résonance magnétique , Troubles liés aux opiacés/diagnostic , Facteurs de risque , Tachycardie ventriculaire/diagnostic , Tachycardie ventriculaire/physiopathologie
7.
J Ethnopharmacol ; 276: 114168, 2021 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-33932511

RÉSUMÉ

ETHNOPHARMACOLOGY RELEVANCE: In folkloric medicine of many cultures, one of the medical uses of Valeriana officinalis Linn is to treat heart-related disease. Recently, it was shown that the ethanol extracts from V. officinalis could effectively prevent auricular fibrillation, and 8-hydroxypinoresinol-4-O-ß-D-glucoside (HPG) from the extracts is one of the two active compounds showing antiarrhythmia activities. AIM OF THE STUDY: The human Kv1.5 channel (hKv1.5) has potential antiarrhythmia activities, and this study arms at investigating the current blocking effects of HPG on hKv1.5 channel. MATERIAL AND METHODS: HPG was obtained from V. officinalis extracts, and hKv1.5 channels were expressed in HEK 293 cells. HPG was perfused while recording the current through hKv1.5 channels. Patch-clamp recording techniques were used to study the effects of HPG at various concentrations (10 µM, 30 µM, and 50 µM) on hKv1.5 channels. RESULTS: The present study demonstrated that HPG inhibited hKv1.5 channel current in a concentration-dependent manner; the higher the concentration, the greater is the inhibition at each depolarization potential. During washout, the channels did not full recover indicating that the un-coupling between HPG and hKv1.5 channels is a slow process. CONCLUSION: HPG may be an effective and safe active ingredient for AF having translational potential.


Sujet(s)
Antiarythmiques/pharmacologie , Canal potassique Kv1.5/antagonistes et inhibiteurs , Extraits de plantes/pharmacologie , Inhibiteurs des canaux potassiques/pharmacologie , Valeriana/composition chimique , Potentiels d'action/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Cellules HEK293 , Humains , Canal potassique Kv1.5/génétique , Techniques de patch-clamp , Facteurs temps , Vérapamil/pharmacologie
8.
J Biol Chem ; 296: 100514, 2021.
Article de Anglais | MEDLINE | ID: mdl-33676894

RÉSUMÉ

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.


Sujet(s)
Endocytose , Cellules souches pluripotentes induites/métabolisme , Canal potassique Kv1.5/métabolisme , Protéine kinase C/métabolisme , Ubiquitination , Animaux , Animaux nouveau-nés , Humains , Cellules souches pluripotentes induites/cytologie , Canal potassique Kv1.5/génétique , Phosphorylation , Protéine kinase C/génétique , Rats , Transduction du signal
9.
Cardiovasc Res ; 117(7): 1746-1759, 2021 06 16.
Article de Anglais | MEDLINE | ID: mdl-33523143

RÉSUMÉ

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.


Sujet(s)
Fibrillation auriculaire/étiologie , Atrium du coeur/métabolisme , Rythme cardiaque , Inflammasomes/métabolisme , Inflammation/étiologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Obésité/complications , Potentiels d'action , Sujet âgé , Animaux , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/physiopathologie , Signalisation calcique , Études cas-témoins , Modèles animaux de maladie humaine , Femelle , Atrium du coeur/physiopathologie , Humains , Inflammation/métabolisme , Inflammation/physiopathologie , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Mâle , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Obésité/métabolisme , Obésité/physiopathologie , Ovis aries
10.
Acta Biochim Pol ; 68(1): 83-89, 2021 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-33533580

RÉSUMÉ

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.


Sujet(s)
Apoptose/génétique , Prolifération cellulaire/génétique , Cellules de la granulosa/métabolisme , Canal potassique Kv1.5/métabolisme , microARN/métabolisme , Syndrome des ovaires polykystiques/sang , Transduction du signal/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Études cas-témoins , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Survie cellulaire/génétique , Femelle , Techniques de knock-down de gènes , Humains , Canal potassique Kv1.5/génétique , microARN/génétique , Syndrome des ovaires polykystiques/anatomopathologie , Transfection
11.
Toxicol Appl Pharmacol ; 411: 115365, 2021 01 15.
Article de Anglais | MEDLINE | ID: mdl-33316272

RÉSUMÉ

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.


Sujet(s)
Polluants environnementaux/toxicité , Canal potassique Kv1.3/antagonistes et inhibiteurs , Canal potassique Kv1.5/effets des médicaments et des substances chimiques , Polychlorobiphényles/toxicité , Inhibiteurs des canaux potassiques/toxicité , Animaux , Relation dose-effet des médicaments , Femelle , Humains , Canal potassique Kv1.3/génétique , Canal potassique Kv1.3/métabolisme , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Potentiels de membrane , Ovocytes , Facteurs temps , Xenopus laevis
12.
Toxins (Basel) ; 12(11)2020 11 23.
Article de Anglais | MEDLINE | ID: mdl-33238397

RÉSUMÉ

Since 1970s, aplysiatoxins (ATXs), a class of biologically active dermatoxins, were identified from the marine mollusk Stylocheilus longicauda, whilst further research indicated that ATXs were originally metabolized by cyanobacteria. So far, there have been 45 aplysiatoxin derivatives discovered from marine cyanobacteria with various geographies. Recently, we isolated two neo-debromoaplysiatoxins, neo-debromoaplysiatoxin G (1) and neo-debromoaplysiatoxin H (2) from the cyanobacterium Lyngbya sp. collected from the South China Sea. The freeze-dried cyanobacterium was extracted with liquid-liquid extraction of organic solvents, and then was subjected to multiple chromatographies to yield neo-debromoaplysiatoxin G (1) (3.6 mg) and neo-debromoaplysiatoxin H (2) (4.3 mg). They were elucidated with spectroscopic methods. Moreover, the brine shrimp toxicity of the aplysiatoxin derivatives representing differential structural classifications indicated that the debromoaplysiatoxin was the most toxic compound (half inhibitory concentration (IC50) value = 0.34 ± 0.036 µM). While neo-aplysiatoxins (neo-ATXs) did not exhibit apparent brine shrimp toxicity, but showed potent blocking action against potassium channel Kv1.5, likewise, compounds 1 and 2 with IC50 values of 1.79 ± 0.22 µM and 1.46 ± 0.14 µM, respectively. Therefore, much of the current knowledge suggests the ATXs with different structure modifications may modulate multiple cellular signaling processes in animal systems leading to the harmful effects on public health.


Sujet(s)
Toxines de Lyngbya/composition chimique , Toxines de Lyngbya/toxicité , Lyngbya , Inhibiteurs des canaux potassiques/composition chimique , Inhibiteurs des canaux potassiques/toxicité , Animaux , Artemia/effets des médicaments et des substances chimiques , Cellules CHO , Cricetulus , Canal potassique Kv1.5/antagonistes et inhibiteurs , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/physiologie
13.
Mar Drugs ; 18(8)2020 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-32823677

RÉSUMÉ

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.


Sujet(s)
Venins de mollusque/pharmacologie , Inhibiteurs des canaux potassiques/pharmacologie , Canaux potassiques de la superfamille Shaker/antagonistes et inhibiteurs , Animaux , Conus , Ouverture et fermeture des portes des canaux ioniques , Canal potassique Kv1.3/antagonistes et inhibiteurs , Canal potassique Kv1.3/génétique , Canal potassique Kv1.3/métabolisme , Canal potassique Kv1.4/antagonistes et inhibiteurs , Canal potassique Kv1.4/génétique , Canal potassique Kv1.4/métabolisme , Canal potassique Kv1.5/antagonistes et inhibiteurs , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Canal potassique Kv1.6/antagonistes et inhibiteurs , Canal potassique Kv1.6/génétique , Canal potassique Kv1.6/métabolisme , Potentiels de membrane , Ovocytes , Canaux potassiques de la superfamille Shaker/génétique , Canaux potassiques de la superfamille Shaker/métabolisme , Xenopus laevis
14.
Circ Arrhythm Electrophysiol ; 13(9): e008093, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32706628

RÉSUMÉ

BACKGROUND: Cardiac gene expression and arrhythmia occurrence have time-of-day variation; however, daily changes in cardiac electrophysiology, arrhythmia susceptibility, and Ca2+ handling have not been characterized. Furthermore, how these patterns change with age is unknown. METHODS: Hearts were isolated during the light (zeitgeber time [ZT] 4 and ZT9) and dark cycle (ZT14 and ZT21) from adult (12-18 weeks) male mice. Hearts from aged (18-20 months) male mice were isolated at ZT4 and ZT14. All hearts were Langendorff-perfused for optical mapping with voltage- and Ca2+-sensitive dyes (n=4-7/group). Cardiac gene and protein expression were assessed with real-time polymerase chain reaction (n=4-6/group) and Western blot (n=3-4/group). RESULTS: Adult hearts had the shortest action potential duration (APD) and Ca2+ transient duration (CaTD) at ZT14 (APD80: ZT4: 45.4±4.1 ms; ZT9: 45.1±8.6 ms; ZT14: 34.7±4.2 ms; ZT21: 49.2±7.6 ms, P<0.05 versus ZT4 and ZT21; and CaTD80: ZT4: 70.1±3.3 ms; ZT9: 72.7±2.7 ms; ZT14: 64.3±3.3 ms; ZT21: 74.4±1.2 ms, P<0.05 versus other time points). The pacing frequency at which CaT alternans emerged was faster, and average CaT alternans magnitude was significantly reduced at ZT14 compared with the other time points. There was a trend for decreased spontaneous premature ventricular complexes and pacing-induced ventricular arrhythmias at ZT14, and the hearts at ZT14 had diminished responses to isoproterenol compared with ZT4 (ZT4: 49.5.0±5.6% versus ZT14: 22.7±9.5% decrease in APD, P<0.01). In contrast, aged hearts exhibited no difference between ZT14 and ZT4 in nearly every parameter assessed (except APD80: ZT4: 39.7±1.9 ms versus ZT14: 33.8±3.1 ms, P<0.01). Gene expression of KCNA5 (potassium voltage-gated channel subfamily A member 5; encoding Kv1.5) was increased, whereas gene expression of ADRB1 (encoding ß1-adrenergic receptors) was decreased at ZT14 versus ZT4 in adult hearts. No time-of-day changes in expression or phosphorylation of Ca2+ handling proteins (SERCA2 [sarco/endoplasmic reticulum Ca2+-ATPase], RyR2 [ryanodine receptor 2], and PLB [phospholamban]) was found in ex vivo perfused adult isolated hearts. CONCLUSIONS: Isolated adult hearts have strong time-of-day variation in cardiac electrophysiology, Ca2+ handling, and adrenergic responsiveness, which is disrupted with age.


Sujet(s)
Potentiels d'action , Vieillissement , Signalisation calcique , Rythme circadien , Rythme cardiaque , Myocarde/métabolisme , Potentiels d'action/effets des médicaments et des substances chimiques , Agonistes bêta-adrénergiques/pharmacologie , Facteurs âges , Animaux , Troubles du rythme cardiaque/étiologie , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/physiopathologie , Signalisation calcique/effets des médicaments et des substances chimiques , Entraînement électrosystolique , Régulation de l'expression des gènes , Rythme cardiaque/effets des médicaments et des substances chimiques , Préparation de coeur isolé , Isoprénaline/pharmacologie , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Mâle , Souris de lignée C57BL , Récepteurs bêta-1 adrénergiques/génétique , Récepteurs bêta-1 adrénergiques/métabolisme , Facteurs temps
15.
BMC Cardiovasc Disord ; 20(1): 337, 2020 07 14.
Article de Anglais | MEDLINE | ID: mdl-32664860

RÉSUMÉ

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.


Sujet(s)
Produits terminaux de glycation avancée/pharmacologie , Canal potassique Kv1.2/métabolisme , Canal potassique Kv1.5/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/métabolisme , Sérumalbumine bovine/pharmacologie , Vasodilatation/effets des médicaments et des substances chimiques , Anilides/pharmacologie , Animaux , Cellules cultivées , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Vaisseaux coronaires/métabolisme , Canal potassique Kv1.2/génétique , Canal potassique Kv1.5/génétique , Mâle , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/effets des médicaments et des substances chimiques , Pioglitazone/pharmacologie , Rat Sprague-Dawley , Transduction du signal
16.
Transl Res ; 224: 40-54, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32522668

RÉSUMÉ

The modulation of voltage-gated K+ (Kv) channels, involved in cell proliferation, arises as a potential therapeutic approach for the prevention of intimal hyperplasia present in in-stent restenosis (ISR) and allograft vasculopathy (AV). We studied the effect of PAP-1, a selective blocker of Kv1.3 channels, on development of intimal hyperplasia in vitro and in vivo in 2 porcine models of vascular injury. In vitro phenotypic modulation of VSMCs was associated to an increased functional expression of Kv1.3 channels, and only selective Kv1.3 channel blockers were able to inhibit porcine VSMC proliferation. The therapeutic potential of PAP-1 was then evaluated in vivo in swine models of ISR and AV. At 15-days follow-up, morphometric analysis demonstrated a substantial reduction of luminal stenosis in the allografts treated with PAP-1 (autograft 2.72 ± 1.79 vs allograft 10.32 ± 1.92 vs allograft + polymer 13.54 ± 8.59 vs allograft + polymer + PAP-1 3.06 ± 1.08 % of luminal stenosis; P = 0.006) in the swine model of femoral artery transplant. In the pig model of coronary ISR, using a prototype of PAP-1-eluting stent, no differences were observed regarding % of stenosis compared to control stents (31 ± 13 % vs 37 ± 18%, respectively; P = 0.372) at 28-days follow-up. PAP-1 treatment was safe and did not impair vascular healing in terms of delayed endothelialization, inflammation or thrombosis. However, an incomplete release of PAP-1 from stents was documented. We conclude that the use of selective Kv1.3 blockers represents a promising therapeutic approach for the prevention of intimal hyperplasia in AV, although further studies to improve their delivery method are needed to elucidate its potential in ISR.


Sujet(s)
Canal potassique Kv1.3/antagonistes et inhibiteurs , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Inhibiteurs des canaux potassiques/pharmacologie , Tunique intime/anatomopathologie , Allogreffes/effets des médicaments et des substances chimiques , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Resténose coronaire/anatomopathologie , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Vaisseaux coronaires/traumatismes , Vaisseaux coronaires/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Artère fémorale/effets des médicaments et des substances chimiques , Artère fémorale/anatomopathologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hyperplasie , Canal potassique Kv1.3/génétique , Canal potassique Kv1.3/métabolisme , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Modèles biologiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , ARN messager/génétique , ARN messager/métabolisme , Endoprothèses , Suidae , Tunique intime/effets des médicaments et des substances chimiques
17.
J Mol Cell Cardiol ; 144: 127-139, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32445844

RÉSUMÉ

Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.


Sujet(s)
Clathrine/métabolisme , Microtubules/métabolisme , Canaux potassiques voltage-dépendants/métabolisme , Multimérisation de protéines , Animaux , Fibrillation auriculaire/étiologie , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/physiopathologie , Remodelage auriculaire/génétique , Clathrine/composition chimique , Vésicules tapissées de clathrine , Cytosquelette/composition chimique , Cytosquelette/métabolisme , Phénomènes électrophysiologiques , Atrium du coeur/métabolisme , Humains , Canal potassique Kv1.5/génétique , Canal potassique Kv1.5/métabolisme , Microtubules/composition chimique , Microtubules/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/ultrastructure , Canaux potassiques voltage-dépendants/composition chimique , Rats , Sarcolemme/métabolisme , Transduction du signal
18.
Am J Physiol Cell Physiol ; 318(5): C954-C968, 2020 05 01.
Article de Anglais | MEDLINE | ID: mdl-32186932

RÉSUMÉ

The increase in cytosolic Ca2+ concentration ([Ca2+]cyt) and upregulation of calcium-sensing receptor (CaSR) and stromal interaction molecule 2 (STIM2) along with inhibition of voltage-gated K+ (KV) channels in pulmonary arterial smooth muscle cells (PASMC) have been implicated in the development of pulmonary arterial hypertension; however, the precise upstream mechanisms remain elusive. Activation of CaSR, a G protein-coupled receptor (GPCR), results in Ca2+ release from the endoplasmic/sarcoplasmic reticulum (ER/SR) and Ca2+ influx through receptor-operated and store-operated Ca2+ channels (SOC). Upon Ca2+ depletion from the SR, STIM forms clusters to mediate store-operated Ca2+ entry. Activity of KV channels, like KCNA5/KV1.5 and KCNA2/KV1.2, contributes to regulating membrane potential, and inhibition of KV channels results in membrane depolarization that increases [Ca2+]cyt by opening voltage-dependent Ca2+ channels. In this study, we show that activation of Notch by its ligand Jag-1 promotes the clustering of STIM2, and clustered STIM2 subsequently enhances the CaSR-induced Ca2+ influx through SOC channels. Extracellular Ca2+-mediated activation of CaSR increases [Ca2+]cyt in CASR-transfected HEK293 cells. Treatment of CASR-transfected cells with Jag-1 further enhances CaSR-mediated increase in [Ca2+]cyt. Moreover, CaSR-mediated increase in [Ca2+]cyt was significantly augmented in cells co-transfected with CASR and STIM2. CaSR activation results in STIM2 clustering in CASR/STIM2-cotransfected cells. Notch activation also induces significant clustering of STIM2. Furthermore, activation of Notch attenuates whole cell K+ currents in KCNA5- and KCNA2-transfected cells. Together, these results suggest that Notch activation enhances CaSR-mediated increases in [Ca2+]cyt by enhancing store-operated Ca2+ entry and inhibits KCNA5/KV1.5 and KCNA2/KV1.2, ultimately leading to voltage-activated Ca2+ entry.


Sujet(s)
Canal potassique Kv1.2/génétique , Canal potassique Kv1.5/génétique , Hypertension artérielle pulmonaire/génétique , Récepteurs-détecteurs du calcium/génétique , Molécule-2 d'interaction stromale/génétique , Canaux calciques/effets des médicaments et des substances chimiques , Canaux calciques/génétique , Signalisation calcique/génétique , Réticulum endoplasmique/génétique , Réticulum endoplasmique/métabolisme , Oestrènes/pharmacologie , Cellules HEK293 , Humains , Indoles/pharmacologie , Protéine jagged-1/génétique , Potentiels de membrane/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Pyrrolidones/pharmacologie , Récepteurs-détecteurs du calcium/effets des médicaments et des substances chimiques , Récepteurs Notch/génétique , Analyse sur cellule unique
19.
J Biol Chem ; 295(14): 4723-4732, 2020 04 03.
Article de Anglais | MEDLINE | ID: mdl-32122972

RÉSUMÉ

The voltage-gated potassium channel Kv1.5 plays important roles in atrial repolarization and regulation of vascular tone. In the present study, we investigated the effects of mechanical stretch on Kv1.5 channels. We induced mechanical stretch by centrifuging or culturing Kv1.5-expressing HEK 293 cells and neonatal rat ventricular myocytes in low osmolarity (LO) medium and then recorded Kv1.5 current (IKv1.5) in a normal, isotonic solution. We observed that mechanical stretch increased IKv1.5, and this increase required the intact, long, proline-rich extracellular S1-S2 linker of the Kv1.5 channel. The low osmolarity-induced IKv1.5 increase also required an intact intracellular N terminus, which contains the binding motif for endogenous Src tyrosine kinase that constitutively inhibits IKv1.5 Disrupting the Src-binding motif of Kv1.5 through N-terminal truncation or mutagenesis abolished the mechanical stretch-mediated increase in IKv1.5 Our results further showed that the extracellular S1-S2 linker of Kv1.5 communicates with the intracellular N terminus. Although the S1-S2 linker of WT Kv1.5 could be cleaved by extracellularly applied proteinase K (PK), an N-terminal truncation up to amino acid residue 209 altered the conformation of the S1-S2 linker and made it no longer susceptible to proteinase K-mediated cleavage. In summary, the findings of our study indicate that the S1-S2 linker of Kv1.5 represents a mechanosensor that regulates the activity of this channel. By targeting the S1-S2 linker, mechanical stretch may induce a change in the N-terminal conformation of Kv1.5 that relieves Src-mediated tonic channel inhibition and results in an increase in IKv1.5.


Sujet(s)
Canal potassique Kv1.5/métabolisme , Potentiels de membrane/physiologie , Contrainte mécanique , Séquence d'acides aminés , Animaux , Sites de fixation , Cellules cultivées , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glycosylation , Cellules HEK293 , Humains , Canal potassique Kv1.5/composition chimique , Canal potassique Kv1.5/génétique , Cellules musculaires/cytologie , Cellules musculaires/métabolisme , Pression osmotique , Domaines protéiques , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Rats , Rat Sprague-Dawley , src-Family kinases/antagonistes et inhibiteurs , src-Family kinases/métabolisme
20.
Biochem Biophys Res Commun ; 524(4): 791-797, 2020 04 16.
Article de Anglais | MEDLINE | ID: mdl-32019676

RÉSUMÉ

Increased granulosa cell (GC) proliferation may contribute to abnormal folliculogenesis in patients with polycystic ovary syndrome (PCOS), which affects approximately 10% reproductive aged women. However, the mechanisms underlying increased GC proliferation in PCOS remain incompletely understood. In this study, we identified miR-3940-5p as a hub miRNA in GC from PCOS using weighted gene co-expression network analysis (WGCNA), and real-time polymerase chain reaction (RT-PCR) analysis confirmed that miR-3940-5p was significantly increased in GC from PCOS. Enrichment analysis of predicted target genes of miR-3940-5p indicated potential roles of miR-3940-5p in follicular development and cell proliferation regulation. Consistently, functional study confirmed that miR-3940-5p overexpression promoted granulosa cell proliferation. Integrated analysis of mRNA expression profiling data and predicted target genes of miR-3940-5p identified potassium voltage-gated channel subfamily A member 5 (KCNA5) as a potential target of miR-3940-5p, and was validated by luciferase reporter assay. Finally, functional analysis suggested that miR-3940-5p promoted GC proliferation in a KCNA5 dependent manner. In conclusion, miR-3940-5p was a hub miRNA upregulated in GC from PCOS, and promoted GC proliferation by targeting KCNA5.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Cellules de la granulosa/métabolisme , Canal potassique Kv1.5/génétique , microARN/génétique , Protéines tumorales/génétique , Syndrome des ovaires polykystiques/génétique , Adulte , Antagomirs/génétique , Antagomirs/métabolisme , Prolifération cellulaire , Femelle , Analyse de profil d'expression de gènes , Gene Ontology , Réseaux de régulation génique , Gènes rapporteurs , Cellules de la granulosa/anatomopathologie , Humains , Canal potassique Kv1.5/antagonistes et inhibiteurs , Canal potassique Kv1.5/métabolisme , Luciferases/génétique , Luciferases/métabolisme , microARN/antagonistes et inhibiteurs , microARN/métabolisme , Annotation de séquence moléculaire , Protéines tumorales/métabolisme , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/anatomopathologie , Culture de cellules primaires , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE