Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Korean J Physiol Pharmacol ; 28(4): 323-333, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38926840

RESUMO

Polychlorinated biphenyls (PCBs) were once used throughout various industries; however, because of their persistence in the environment, exposure remains a global threat to the environment and human health. The Kv1.3 and Kv1.5 channels have been implicated in the immunotoxicity and cardiotoxicity of PCBs, respectively. We determined whether 3,3',4,4'-tetrachlorobiphenyl (PCB77), a dioxin-like PCB, alters human Kv1.3 and Kv1.5 currents using the Xenopus oocyte expression system. Exposure to 10 nM PCB77 for 15 min enhanced the Kv1.3 current by approximately 30.6%, whereas PCB77 did not affect the Kv1.5 current at concentrations up to 10 nM. This increase in the Kv1.3 current was associated with slower activation and inactivation kinetics as well as right-shifting of the steady-state activation curve. Pretreatment with PCB77 significantly suppressed tumor necrosis factor-α and interleukin-10 production in lipopolysaccharide-stimulated Raw264.7 macrophages. Overall, these data suggest that acute exposure to trace concentrations of PCB77 impairs immune function, possibly by enhancing Kv1.3 currents.

2.
Biol Pharm Bull ; 46(10): 1394-1402, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37779040

RESUMO

Dimenhydrinate, an H1 receptor antagonist, is generally used for the prevention and treatment of nausea and vomiting. However, cardiac arrhythmias have been reported to be associated with the overdose of histamine H1 receptor antagonists, indicating the probable effect of antihistamines on ion channels. By using a two-microelectrode voltage clamp, we have herein studied the electrophysiological effects of dimenhydrinate on the human Kv1.5 channel in the Xenopus oocyte expression system. Dimenhydrinate acutely and reversibly suppressed the amplitudes of the peak and the steady-state current, within 6 min. The inhibitory effect of dimenhydrinate on the peak and the steady-state Kv1.5 currents increased progressively from -10 to +50 mV. At each test voltage, the drug suppressed both the peak and the steady-state currents to a similar extent. When the oocytes were stimulated at the rates of 5- and 30-s intervals, dimenhydrinate-induced a use-dependent blockade of the human Kv1.5 channel. Dimenhydrinate expedited the timecourse of the Kv1.5 channel activation more effectively than the timecourse of its inactivation. However, the activation and inactivation curves of the channel were not altered by the H1 receptor antagonist. In conclusion, we found that dimenhydrinate inhibits the human Kv1.5 channel by changing the channel's activation mode, thereby possibly increasing the possibility of triggering cardiac arrhythmias and affecting atrial fibrillation.


Assuntos
Dimenidrinato , Humanos , Dimenidrinato/metabolismo , Dimenidrinato/farmacologia , Fenômenos Eletrofisiológicos , Antagonistas dos Receptores Histamínicos H1/farmacologia , Oócitos/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia
3.
Toxicol Appl Pharmacol ; 411: 115365, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33316272

RESUMO

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.


Assuntos
Poluentes Ambientais/toxicidade , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.5/efeitos dos fármacos , Bifenilos Policlorados/toxicidade , Bloqueadores dos Canais de Potássio/toxicidade , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Potenciais da Membrana , Oócitos , Fatores de Tempo , Xenopus laevis
4.
Nature ; 519(7544): 472-6, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25799991

RESUMO

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Cardiomegalia/enzimologia , Cardiomegalia/metabolismo , GMP Cíclico/metabolismo , Óxido Nítrico , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/deficiência , 3',5'-AMP Cíclico Fosfodiesterases/genética , Animais , Estenose da Valva Aórtica/complicações , Cardiomegalia/tratamento farmacológico , Cardiomegalia/etiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/enzimologia , Miocárdio/enzimologia , Peptídeos Natriuréticos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfodiesterase/uso terapêutico , Pressão , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico , Regulação para Cima
5.
Eur J Oral Sci ; 125(1): 18-27, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28032657

RESUMO

Bradykinin is an important peptide modulator that affects the function of neurons and immune cells. However, there is no evidence of the bradykinin receptors and their functions in human salivary glands. Here we have identified and characterized bradykinin receptors on human submandibular gland cells. Both bradykinin B1 and B2 receptors are expressed on human submandibular gland cells, A253 cells, and HSG cells. Bradykinin increased the intracellular Ca2+ concentration ([Ca2+ ]i ) in a concentration-dependent manner. Interestingly, a specific agonist of the B1 receptor did not have any effect on [Ca2+ ]i in HSG cells, whereas specific agonists of the B2 receptor had a Ca2+ mobilizing effect. Furthermore, application of the B1 receptor antagonist, R715, did not alter the bradykinin-mediated increase in cytosolic Ca2+ , whereas the B2 receptor antagonist, HOE140, showed a strong inhibitory effect, which implies that bradykinin B2 receptors are functional in modulating the concentration of cytosolic Ca2+ . Bradykinin did not affect a carbachol-induced rise of [Ca2+ ]i and did not modulate translocation of aquaporin-5. However, bradykinin did promote the expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), implying the role of bradykinin in salivary gland inflammation. These data suggest that bradykinin receptors are involved in Ca2+ signaling in human submandibular gland cells and serve a unique role, which is separate from that of other salivary gland G protein-coupled receptors.


Assuntos
Citocinas/metabolismo , Receptores da Bradicinina/metabolismo , Glândulas Salivares/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Aquaporina 5/metabolismo , Western Blotting , Bradicinina/análogos & derivados , Bradicinina/farmacologia , Cálcio/metabolismo , Carbacol/farmacologia , Linhagem Celular , Células Cultivadas , AMP Cíclico/metabolismo , Feminino , Imunofluorescência , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Glândulas Salivares/citologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(4): 1551-6, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24453217

RESUMO

Chronic neurohormonal and mechanical stresses are central features of heart disease. Increasing evidence supports a role for the transient receptor potential canonical channels TRPC3 and TRPC6 in this pathophysiology. Channel expression for both is normally very low but is increased by cardiac disease, and genetic gain- or loss-of-function studies support contributions to hypertrophy and dysfunction. Selective small-molecule inhibitors remain scarce, and none target both channels, which may be useful given the high homology among them and evidence of redundant signaling. Here we tested selective TRPC3/6 antagonists (GSK2332255B and GSK2833503A; IC50, 3-21 nM against TRPC3 and TRPC6) and found dose-dependent blockade of cell hypertrophy signaling triggered by angiotensin II or endothelin-1 in HEK293T cells as well as in neonatal and adult cardiac myocytes. In vivo efficacy in mice and rats was greatly limited by rapid metabolism and high protein binding, although antifibrotic effects with pressure overload were observed. Intriguingly, although gene deletion of TRPC3 or TRPC6 alone did not protect against hypertrophy or dysfunction from pressure overload, combined deletion was protective, supporting the value of dual inhibition. Further development of this pharmaceutical class may yield a useful therapeutic agent for heart disease management.


Assuntos
Cardiomegalia/genética , Canais de Cátion TRPC/antagonistas & inibidores , Animais , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Ratos , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6
7.
J Neurochem ; 132(3): 276-85, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25418874

RESUMO

Methyl-ß-cyclodextrin (MßCD) is a reagent that depletes cholesterol and disrupts lipid rafts, a type of cholesterol-enriched cell membrane microdomain. Lipid rafts are essential for neuronal functions such as synaptic transmission and plasticity, which are sensitive to even low doses of MßCD. However, how MßCD changes synaptic function, such as N-methyl-d-aspartate receptor (NMDA-R) activity, remains unclear. We monitored changes in synaptic transmission and plasticity after disrupting lipid rafts with MßCD. At low concentrations (0.5 mg/mL), MßCD decreased basal synaptic transmission and miniature excitatory post-synaptic current without changing NMDA-R-mediated synaptic transmission and the paired-pulse facilitation ratio. Interestingly, low doses of MßCD failed to deplete cholesterol or affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-R) and NMDA-R levels, while clearly reducing GluA1 levels selectively in the synaptosomal fraction. Low doses of MßCD decreased the inhibitory effects of NASPM, an inhibitor for GluA2-lacking AMPA-R. MßCD successfully decreased NMDA-R-mediated long-term potentiation but did not affect the formation of either NMDA-R-mediated or group I metabotropic glutamate receptor-dependent long-term depression. MßCD inhibited de-depression without affecting de-potentiation. These results suggest that MßCD regulates GluA1-dependent synaptic potentiation but not synaptic depression in a cholesterol-independent manner.


Assuntos
Receptores de AMPA/fisiologia , Sinapses/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia , Animais , Colesterol/metabolismo , Técnicas In Vitro , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos
8.
Biochem Biophys Res Commun ; 459(4): 604-9, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25753203

RESUMO

Calcineurin is a calcium/calmodulin-dependent phosphatase that has been implicated in T cell activation through the induction of nuclear factors of activated T cells (NFAT). We have previously suggested that endogenous regulator of calcineurin (RCAN1, also known as DSCR1) is targeted by protein kinase A (PKA) for the control of calcineurin activity. In the present study, we characterized the PKA-mediated phosphorylation site in RCAN1 by mass spectrometric analysis and revealed that PKA directly phosphorylated RCAN1 at the Ser 93. PKA-induced phosphorylation and the increase in the half-life of the RCAN1 protein were prevented by the substitution of Ser 93 with Ala (S93A). Furthermore, the PKA-mediated phosphorylation of RCAN1 at Ser 93 potentiated the inhibition of calcineurin-dependent pro-inflammatory cytokine gene expression by RCAN1. Our results suggest the presence of a novel phosphorylation site in RCAN1 and that its phosphorylation influences calcineurin-dependent inflammatory target gene expression.


Assuntos
Calcineurina/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , Sequência de Aminoácidos , Proteínas de Ligação a DNA , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Dados de Sequência Molecular , Proteínas Musculares/química , Fosforilação
9.
Biol Pharm Bull ; 37(9): 1495-504, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25177033

RESUMO

The effects of paroxetine, a selective serotonin reuptake inhibitor, on human ether-a-go-go-related gene (HERG) channels were investigated using the whole-cell patch-clamp technique. The HERG channels were stably expressed in human embryonic kidney cells. Paroxetine inhibited the peak tail currents of the HERG channel in a concentration-dependent manner, with an IC50 value of 0.45 µM and a Hill coefficient of 0.85. These effects were reversible after wash-out of the drug. The paroxetine-induced inhibition of the HERG channels was voltage-dependent. There was a steep increase in inhibition over the voltage range of the channel opening. Also, a shallow voltage-dependent inhibition was detected over the voltage range in which the channels were fully activated. The fractional electrical distance was estimated to be 0.11. Paroxetine induced a leftward shift in the voltage-dependence of the steady-state activation of the HERG channels. Before and after application of the 1 µM paroxetine, the half-maximum activation was -14.21 mV and -27.04 mV, respectively, with no shift in the slope value. The HERG channel block was not use-dependent. The characteristics of the block were dependent on open and inactivated channel states rather than closed state. Paroxetine had no effect on activation and deactivation kinetics, steady-state inactivation. These results suggest that paroxetine blocks the HERG channels by binding to these channels in the open and inactivated states.


Assuntos
Antidepressivos/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Paroxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/fisiologia , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Modelos Moleculares
10.
J Pharmacol Sci ; 123(1): 67-77, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-24005046

RESUMO

Azelastine is a second generation histamine H1-receptor antagonist used as an anti-asthmatic and anti-allergic drug that can induce QT prolongation and torsades de pointes. We investigated the acute effects of azelastine on human ether-a-go-go-related gene (hERG) channels, action potential duration (APD), and L-type (I(Ca,L)) and T-type Ca²âº current (I(Ca,T)) to determine the electrophysiological basis for its proarrhythmic potential. Azelastine increased the APD at 90% of repolarization concentration dependently, with an IC50 of 1.08 nM in guinea-pig ventricular myocytes. We examined the effects of azelastine on the hERG channels expressed in Xenopus oocytes and HEK293 cells using two-microelectrode voltage-clamp and patch-clamp techniques. Azelastine induced a concentration-dependent decrease of the hERG current amplitude at the end of the voltage steps and tail currents. The IC50 for the azelastine-induced block of the hERG currents expressed in HEK293 cells was 11.43 nM, while the drug inhibited I(Ca,L) and I(Ca,T) with IC50 values of 7.60 and 26.21 µM, respectively. The S6 domain mutations, Y652A partially attenuated and F656A abolished hERG current block. These results suggest that azelastine is a potent blocker of hERG channels rather than I(Ca,L) or I(Ca,T), providing molecular mechanisms for the arrhythmogenic side effects during the clinical administration of azelastine.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo T/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Antagonistas dos Receptores Histamínicos H1/efeitos adversos , Antagonistas dos Receptores Histamínicos H1/farmacologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ftalazinas/efeitos adversos , Ftalazinas/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Cobaias , Células HEK293 , Ventrículos do Coração/citologia , Humanos , Oócitos , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Xenopus laevis
11.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3149-3161, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37166464

RESUMO

Ifenprodil has been known to reduce cardiac contractility and cerebral vasodilation by antagonizing α1-adrenergic and N-methyl D-aspartate receptor-mediated intracellular signals. This study aimed to investigate the direct effect of ifenprodil on the human voltage-gated Kv1.5 channel (hKv1.5) by using a Xenopus oocyte expression system and a two-microelectrode voltage clamp technique. The amplitudes of hKv1.5 currents, including peak and steady state, were suppressed in a concentration-dependent manner (IC50; 43.1 and 35.5 µM, respectively) after 6 min of ifenprodil treatment. However, these effects were ~ 80% reversed by washout, suggesting that ifenprodil directly inhibited the hKv1.5 independent of membrane receptors or intracellular signals. The inhibition rate of steady state showed voltage dependence, wherein the rates increased according to test voltage depolarization. Ifenprodil reduced the time constants of hKv1.5 inactivation but has higher effects on activation. hKv1.5 inhibition by ifenprodil showed use dependency because the drug more rapidly reduced the current at the higher activation frequencies, and subsequent reduction in frequency after high activation frequency caused a partial channel block relief. Therefore, ifenprodil directly blocked the hKv1.5 in an open state and accelerated the time course of the channel inactivation, which provided a biophysical mechanism for the hKv1.5 blocking effects of ifenprodil.


Assuntos
N-Metilaspartato , Piperidinas , Humanos , Piperidinas/farmacologia , Receptores de N-Metil-D-Aspartato , Antagonistas de Receptores Adrenérgicos alfa 1 , Canal de Potássio Kv1.5 , Bloqueadores dos Canais de Potássio/farmacologia
12.
Toxicol Appl Pharmacol ; 262(1): 60-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22676973

RESUMO

Polychlorinated biphenyls (PCBs) have been known as serious persistent organic pollutants (POPs), causing developmental delays and motor dysfunction. We have investigated the effects of two PCB congeners, 3,3',4,4'-tetrachlorobiphenyl (PCB 77) and 3,3',4,4',5-pentachlorobiphenyl (PCB 126) on ECG, action potential, and the rapidly activating delayed rectifier K+ current (I(Kr)) of guinea pigs' hearts, and hERG K+ current expressed in Xenopus oocytes. PCB 126 shortened the corrected QT interval (QTc) of ECG and decreased the action potential duration at 90% (APD(90)), and 50% of repolarization (APD50) (P<0.05) without changing the action potential duration at 20% (APD20). PCB 77 decreased APD20 (P<0.05) without affecting QTc, APD90, and APD50. The PCB 126 increased the I(Kr) in guinea-pig ventricular myocytes held at 36°C and hERG K+ current amplitude at the end of the voltage steps in voltage-dependent mode (P<0.05); however, PCB 77 did not change the hERG K+ current amplitude. The PCB 77 increased the diastolic Ca²âº and decreased Ca²âº transient amplitude (P<0.05), however PCB 126 did not change. The results suggest that PCB 126 shortened the QTc and decreased the APD90 possibly by increasing I(Kr), while PCB 77 decreased the APD20 possibly by other modulation related with intracellular Ca²âº. The present data indicate that the environmental toxicants, PCBs, can acutely affect cardiac electrophysiology including ECG, action potential, intracellular Ca²âº, and channel activity, resulting in toxic effects on the cardiac function in view of the possible accumulation of the PCBs in human body.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Bifenilos Policlorados/toxicidade , Animais , Cálcio/metabolismo , Canais de Potássio de Retificação Tardia/metabolismo , Canal de Potássio ERG1 , Eletrocardiografia , Poluentes Ambientais/toxicidade , Canais de Potássio Éter-A-Go-Go/metabolismo , Cobaias , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oócitos , Xenopus laevis
13.
Acta Pharmacol Sin ; 32(9): 1128-37, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21892192

RESUMO

AIM: To investigate the effects of hydroxyzine on human ether-a-go-go-related gene (hERG) channels to determine the electrolphysiological basis for its proarrhythmic effects. METHODS: hERG channels were expressed in Xenopus oocytes and HEK293 cells, and the effects of hydroxyzine on the channels were examined using two-microelectrode voltage-clamp and patch-clamp techniques, respectively. The effects of hydroxyzine on action potential duration were examined in guinea pig ventricular myocytes using current clamp. RESULTS: Hydroxyzine (0.2 and 2 µmol/L) significantly increased the action potential duration at 90% repolarization (APD(90)) in both concentration- and time-dependent manners. Hydroxyzine (0.03-3 µmol/L) blocked both the steady-state and tail hERG currents. The block was voltage-dependent, and the values of IC(50) for blocking the steady-state and tail currents at +20 mV was 0.18±0.02 µmol/L and 0.16±0.01 µmol/L, respectively, in HEK293 cells. Hydroxyzine (5 µmol/L) affected both the activated and the inactivated states of the channels, but not the closed state. The S6 domain mutation Y652A attenuated the blocking of hERG current by ~6-fold. CONCLUSION: The results suggest that hydroxyzine could block hERG channels and prolong APD. The tyrosine at position 652 in the channel may be responsible for the proarrhythmic effects of hydroxyzine.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/metabolismo , Antagonistas dos Receptores Histamínicos H1/farmacologia , Hidroxizina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/genética , Expressão Gênica , Cobaias , Humanos , Técnicas de Patch-Clamp , Xenopus laevis
14.
Eur J Pharmacol ; 912: 174567, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34662565

RESUMO

Paroxetine is one of the most effective selective serotonin reuptake inhibitors used to treat depressive and panic disorders that reduce the viability of human T lymphocytes, in which Kv1.3 channels are highly expressed. We examined whether paroxetine could modulate human Kv1.3 channels acutely and directly with the aim of understanding the biophysical effects and the underlying mechanisms of the drug. Kv1.3 channel proteins were expressed in Xenopus oocytes. Paroxetine rapidly inhibited the steady-state current and peak current of these channels within 6 min in a concentration-dependent manner; IC50s were 26.3 µM and 53.9 µM, respectively, and these effects were partially reversed by washout, which excluded the possibility of genomic regulation. At the same test voltage, paroxetine blockade of the steady-state currents was higher than that of the peak currents, and the inhibition of the steady-state current increased relative to the degree of depolarization. Paroxetine decreased the inactivation time constant in a concentration-dependent manner, but it did not affect the activation time constant, which resulted in the acceleration of intrinsic inactivation without changing ultrarapid activation. Blockade of Kv1.3 channels by paroxetine exhibited more rapid inhibition at higher activation frequencies showing the use-dependency of the blockade. Overall, these results show that paroxetine directly suppresses human Kv1.3 channels in an open state and accelerates the process of steady-state inactivation; thus, we have revealed a biophysical mechanism for possible acute immunosuppressive effects of paroxetine.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Paroxetina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Humanos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Xenopus laevis
15.
Biochem Biophys Res Commun ; 394(3): 536-41, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20211602

RESUMO

Desipramine is a tricyclic antidepressant for psychiatric disorders that can induce QT prolongation, which may lead to torsades de pointes. Since blockade of cardiac human ether-a-go-go-related gene (hERG) channels is an important cause of acquired long QT syndrome, we investigated the acute effects of desipramine on hERG channels to determine the electrophysiological basis for its pro-arrhythmic potential. We examined the effects of desipramine on the hERG channels expressed in Xenopus oocytes using two-microelectrode voltage-clamp techniques. Desipramine-induced concentration-dependent decreases in the current amplitude at the end of the voltage steps and hERG tail currents. The IC(50) for desipramine needed to block the hERG current in Xenopus oocytes decreased progressively relative to the degree of depolarization. Desipramine affected the channels in the activated and inactivated states but not in the closed states. The S6 domain mutations, Tyr-652 located in the S6 domain of the hERG channel reduced the potency of the channel block by desipramine more than a mutation of Phe-656 in the same region. These results suggest that desipramine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of desipramine.


Assuntos
Antidepressivos Tricíclicos/efeitos adversos , Desipramina/efeitos adversos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Síndrome do QT Longo/induzido quimicamente , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Concentração Inibidora 50 , Mutação , Oócitos , Fenilalanina/genética , Estrutura Terciária de Proteína/genética , Tirosina/genética , Xenopus
16.
J Pharmacol Exp Ther ; 330(2): 403-12, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19443731

RESUMO

One of the common side effects of antihistamine medicines is xerostomia (dry mouth). The current consensus is that antihistamine-induced xerostomia comes from an antimuscarinic effect. Although the effect of antihistamines on salivary secretion is both obvious and significant, the cellular mechanism whereby this happens is still unclear because of the lack of knowledge of histamine signaling in human salivary glands. Here, we have studied histamine receptors and the effect of antihistamines on human submandibular acinar cells. In primary cultured human submandibular gland and a HSG cell line, histamine increased the intracellular Ca(2+) concentration. The histamine-induced cytosolic free Ca(2+) concentration ([Ca(2+)](i)) increase was inhibited by histamine H1 receptor-specific antagonists, and the expression of the functional histamine H1 receptor was confirmed by reverse transcription-polymerase chain reaction. Interestingly, histamine pretreatment did not inhibit a subsequent carbachol-induced [Ca(2+)](i) rise without "heterologous desensitization." Chlorpheniramine inhibited a carbachol-induced [Ca(2+)](i) increase at a 100-fold greater concentration than histamine receptor antagonism, whereas astemizole and cetrizine showed more than 1000-fold difference, which in part explains the xerostomia-inducing potency among the antihistamines. Notably, histamine resulted in translocation of aquaporin-5 to the plasma membrane in human submandibular gland cells and green fluorescent protein-tagged aquaporin-5 expressing HSG cells. We found that histidine decarboxylase and the histamine H1 receptor are broadly distributed in submandibular gland cells, whereas choline acetyltransferase is localized only at the parasympathetic terminals. Our results suggest that human salivary gland cells express histamine H1 receptors and histamine-synthesizing enzymes, revealing the cellular mechanism of antihistamine-induced xerostomia.


Assuntos
Aquaporina 5/metabolismo , Cálcio/metabolismo , Citosol/metabolismo , Receptores Histamínicos H1/fisiologia , Glândula Submandibular/metabolismo , Adulto , Idoso , Células Cultivadas , Citosol/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transporte Proteico/fisiologia , Glândula Submandibular/química , Glândula Submandibular/citologia , Regulação para Cima/fisiologia
17.
Pharmacol Res ; 60(5): 429-37, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19497368

RESUMO

Promethazine is a phenothiazine derivative with antihistaminic (H(1)), sedative, antiemetic, anticholinergic, and antimotion sickness properties that can induce QT prolongation, which may lead to torsades de pointes. Since block of cardiac human ether-a-go-go-related gene (hERG) channels is one of the leading causes of acquired long QT syndrome, we investigated the acute effects of promethazine on hERG channels to determine the electrophysiological basis for its proarrhythmic potential. Promethazine increased the action potential duration at 90% of repolarization (APD(90)) in a concentration-dependent manner, with an IC(50) of 0.73microM when action potentials were elicited under current clamp in guinea pig ventricular myocytes. We examined the effects of promethazine on the hERG channels expressed in Xenopus oocytes and HEK293 cells using two-microelectrode voltage-clamp and patch-clamp techniques. Promethazine induced a concentration-dependent decrease of the current amplitude at the end of the voltage steps and hERG tail currents. The IC(50) of promethazine dependent hERG block in Xenopus oocytes decreased progressively relative to the degree of depolarization. The IC(50) for the promethazine-induced block of the hERG currents in HEK293 cells at 36 degrees C was 1.46microM at +20mV. Promethazine affected the channels in the activated and inactivated states but not in the closed states. The S6 domain mutations, Y652A and F656A partially attenuated (Y652A) or abolished (F656A) the hERG current block. These results suggest that promethazine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of promethazine.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Antagonistas dos Receptores Histamínicos H1/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Prometazina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular , Canais de Potássio Éter-A-Go-Go/genética , Feminino , Cobaias , Humanos , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Oócitos/metabolismo , Xenopus laevis/metabolismo
18.
J Korean Med Sci ; 24(6): 1089-98, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19949665

RESUMO

Lindera erythrocarpa Makino (Lauraceae) is used as a traditional medicine for analgesic, antidote, and antibacterial purposes and shows anti-tumor activity. We studied the effects of Lindera erythrocarpa on the human ether-a-go-go-related gene (HERG) channel, which appears of importance in favoring cancer progression in vivo and determining cardiac action potential duration. Application of MeOH extract of Lindera erythrocarpa showed a dose-dependent decrease in the amplitudes of the outward currents measured at the end of the pulse (I(HERG)) and the tail currents of HERG (I(tail)). When the BuOH fraction and H(2)O fraction of Lindera erythrocarpa were added to the perfusate, both I(HERG) and I(tail) were suppressed, while the hexane fraction, CHCl(3) fraction, and EtOAc fraction did not inhibit either I(HERG) or I(tail). The potential required for half-maximal activation caused by EtOAc fraction, BuOH fraction, and H(2)O fraction shifted significantly. The BuOH fraction and H(2)O fraction (100 microg/mL) decreased g(max) by 59.6% and 52.9%, respectively. The H(2)O fraction- and BuOH fraction-induced blockades of I(tail) progressively decreased with increasing depolarization, showing the voltage-dependent block. Our findings suggest that Lindera erythrocarpa, a traditional medicine, blocks HERG channel, which could contribute to its anticancer and cardiac arrhythmogenic effect.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Lindera/química , Extratos Vegetais/metabolismo , Bloqueadores dos Canais de Potássio/metabolismo , Animais , Butanóis/química , Butanóis/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Humanos , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Xenopus laevis
19.
Mol Cell Endocrinol ; 494: 110488, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31207272

RESUMO

Hydrocortisone exerts adverse effects on various organs, including the heart. This study investigated the still unclear effects of hydrocortisone on electrophysiological and biochemical aspects of cardiac excitation-contraction coupling. In guinea pigs' hearts, hydrocortisone administration reduced the QT interval of ECG and the action potential duration (APD). In guinea pig ventricular myocytes, hydrocortisone reduced contraction and Ca2+ transient amplitudes. These reductions and the effects on APD were prevented by pretreatment with the protein kinase C (PKC) inhibitor staurosporine. In an overexpression system of Xenopus oocytes, hydrocortisone increased hERG K+ currents and reduced Kv1.5 K+ currents; these effects were negated by pretreatment with staurosporine. Western blot analysis revealed dose- and time-dependent changes in PKCα/ßII, PKCε, and PKCγ phosphorylation by hydrocortisone in guinea pig ventricular myocytes. Therefore, hydrocortisone can acutely affect cardiac excitation-contraction coupling, including ion channel activity, APD, ECG, Ca2+ transients, and contraction, possibly via biochemical changes in PKC.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Cálcio/metabolismo , Eletrocardiografia , Coração/fisiologia , Hidrocortisona/farmacologia , Espaço Intracelular/metabolismo , Contração Miocárdica/efeitos dos fármacos , Proteína Quinase C/metabolismo , Animais , Diástole/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/metabolismo , Cobaias , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Ventrículos do Coração/citologia , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Estaurosporina/farmacologia , Fatores de Tempo , Xenopus laevis
20.
Eur J Pharmacol ; 592(1-3): 19-25, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18634780

RESUMO

Clomipramine is a tricyclic antidepressant for psychiatric disorders that can induce QT prolongation, which may lead to torsades de pointes. Since blockade of cardiac human ether-a-go-go-related gene (hERG) channels is an important cause of acquired long QT syndrome, we investigated the acute effects of clomipramine on hERG channels to determine the electrophysiological basis for its proarrhythmic potential. We examined the effects of clomipramine on the hERG channels expressed in Xenopus oocytes and HEK293 cells using two-microelectrode voltage-clamp and patch-clamp techniques. Clomipramine induced a concentration-dependent decrease in the current amplitude at the end of the voltage steps and hERG tail currents. The IC50 for clomipramine needed to block the hERG current in Xenopus oocytes decreased progressively relative to the degree of depolarization. The fractional electrical distance was estimated to be delta=0.83. The IC50 for the clomipramine-induced blockade of the hERG currents in HEK293 cells at 36 degrees C was 0.13 microM at +20 mV. Clomipramine affected the channels in the activated and inactivated states but not in the closed states. The clomipramine-induced blockade of hERG was found to be use-dependent, exhibiting a more rapid onset and a greater steady-state block at the higher frequencies of activation. The S6 domain mutations, Y652A and F656A partially attenuated (Y652A) or abolished (F656A) the hERG-current blockade. These results suggest that clomipramine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of clomipramine.


Assuntos
Clomipramina/farmacologia , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Bloqueadores dos Canais de Potássio , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Algoritmos , Animais , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Síndrome do QT Longo/induzido quimicamente , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Mutação Puntual/genética , Mutação Puntual/fisiologia , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA