Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.582
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Immunity ; 49(1): 56-65.e4, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29958799

RESUMO

Potassium (K+) efflux across the plasma membrane is thought to be an essential mechanism for ATP-induced NLRP3 inflammasome activation, yet the identity of the efflux channel has remained elusive. Here we identified the two-pore domain K+ channel (K2P) TWIK2 as the K+ efflux channel triggering NLRP3 inflammasome activation. Deletion of Kcnk6 (encoding TWIK2) prevented NLRP3 activation in macrophages and suppressed sepsis-induced lung inflammation. Adoptive transfer of Kcnk6-/- macrophages into mouse airways after macrophage depletion also prevented inflammatory lung injury. The K+ efflux channel TWIK2 in macrophages has a fundamental role in activating the NLRP3 inflammasome and consequently mediates inflammation, pointing to TWIK2 as a potential target for anti-inflammatory therapies.


Assuntos
Inflamassomos/metabolismo , Inflamação/fisiopatologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Caspase 1/deficiência , Caspase 1/metabolismo , Linhagem Celular , Inflamassomos/efeitos dos fármacos , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/fisiopatologia , Macrófagos/transplante , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/deficiência , Quinina/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores Purinérgicos P2X7/deficiência , Receptores Purinérgicos P2X7/metabolismo , Sepse/metabolismo , Sepse/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
2.
Immunity ; 38(6): 1142-53, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23809161

RESUMO

The NLRP3 inflammasome is an important component of the innate immune system. However, its mechanism of activation remains largely unknown. We show that NLRP3 activators including bacterial pore-forming toxins, nigericin, ATP, and particulate matter caused mitochondrial perturbation or the opening of a large membrane pore, but this was not required for NLRP3 activation. Furthermore, reactive oxygen species generation or a change in cell volume was not necessary for NLRP3 activation. Instead, the only common activity induced by all NLRP3 agonists was the permeation of the cell membrane to K⁺ and Na⁺. Notably, reduction of the intracellular K⁺ concentration was sufficient to activate NLRP3, whereas an increase in intracellular Na⁺ modulated but was not strictly required for inflammasome activation. These results provide a unifying model for the activation of the NLRP3 inflammasome in which a drop in cytosolic K⁺ is the common step that is necessary and sufficient for caspase-1 activation.


Assuntos
Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Macrófagos/imunologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Caspase 1/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Imunidade Inata , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nigericina/farmacologia , Material Particulado/farmacologia , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
3.
Cereb Cortex ; 31(5): 2402-2415, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33341872

RESUMO

Oxytocin (OXT) is a nonapeptide that serves as a neuromodulator in the brain and a hormone participating in parturition and lactation in the periphery. The subiculum is the major output region of the hippocampus and an integral component in the networks that process sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whilst the subiculum expresses the highest OXT-binding sites and is the first brain region to be activated by peripheral application of OXT, the precise actions of OXT in the subiculum have not been determined. Our results demonstrate that application of the selective OXT receptor (OXTR) agonist, [Thr4,Gly7]-oxytocin (TGOT), excited subicular neurons via activation of TRPV1 channels, and depression of K+ channels. The OXTR-mediated excitation of subicular neurons required the functions of phospholipase Cß, protein kinase C, and degradation of phosphatidylinositol 4,5-bisphosphate (PIP2). OXTR-elicited excitation of subicular neurons enhanced long-term potentiation via activation of TRPV1 channels. Our results provide a cellular and molecular mechanism to explain the physiological functions of OXT in the brain.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de Ocitocina/metabolismo , Canais de Cátion TRPV/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Sinalização do Cálcio , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Ocitocina/análogos & derivados , Ocitocina/farmacologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta/efeitos dos fármacos , Fosfolipase C beta/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos , Receptores de Ocitocina/agonistas , Transdução de Sinais , Canais de Cátion TRPV/efeitos dos fármacos
4.
Am J Physiol Cell Physiol ; 320(4): C520-C546, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33326312

RESUMO

Several potassium channels (KCs) have been described throughout the gastrointestinal tract. Notwithstanding, their contribution to both physiologic and pathophysiologic conditions, as inflammatory bowel disease (IBD), remains underexplored. Therefore, we aim to systematically review, for the first time, the evidence on the characteristics and modulation of KCs in intestinal epithelial cells (IECs). PubMed, Scopus, and Web of Science were searched to identify studies focusing on KCs and their modulation in IECs. The included studies were assessed using a reporting inclusiveness checklist. From the 745 identified records, 73 met the inclusion criteria; their reporting inclusiveness was moderate-high. Some studies described the physiological role of KCs, while others explored their importance in pathological settings. Globally, in IBD animal models, apical KCa1.1 channels, responsible for luminal secretion, were upregulated. In human colonocytes, basolateral KCa3.1 channels were downregulated. The pharmacological inhibition of K2P and Kv influenced intestinal barrier function, promoting inflammation. Evidence suggests a strong association between KCs expression and secretory mechanisms in human and animal IECs. Further research is warranted to explore the usefulness of KC pharmacological modulation as a therapeutic target.


Assuntos
Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Potenciais da Membrana , Canais de Potássio/metabolismo , Transdução de Sinais
5.
Physiol Rev ; 94(2): 609-53, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24692356

RESUMO

About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.


Assuntos
Arritmias Cardíacas/metabolismo , Sistema de Condução Cardíaco/metabolismo , Miocárdio/metabolismo , Canais de Potássio/metabolismo , Potássio/metabolismo , Potenciais de Ação , Animais , Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/fisiopatologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Período Refratário Eletrofisiológico
6.
Am J Physiol Heart Circ Physiol ; 320(6): H2201-H2210, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33891515

RESUMO

Our previous study indicated that intravenously administered ivabradine (IVA) augmented the dynamic heart rate (HR) response to moderate-intensity vagal nerve stimulation (VNS). Considering an accentuated antagonism, the results were somewhat paradoxical; i.e., the accentuated antagonism indicates that an activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels via the accumulation of intracellular cyclic adenosine monophosphate (cAMP) augments the HR response to VNS, whereas the inhibition of HCN channels by IVA also augmented the HR response to VNS. To remove the possible influence from the accentuated antagonism, we examined the effects of IVA on the dynamic vagal control of HR under ß-blockade. In anesthetized rats (n = 7), the right vagal nerve was stimulated for 10 min according to binary white noise signals between 0 and 10 Hz (V0-10), between 0 and 20 Hz (V0-20), and between 0 and 40 Hz (V0-40). The transfer function from VNS to HR was estimated. Under ß-blockade (propranolol, 2 mg/kg iv), IVA (2 mg/kg iv) did not augment the asymptotic low-frequency gain but increased the asymptotic high-frequency gain in V0-10 (0.53 ± 0.10 vs. 1.74 ± 0.40 beats/min/Hz, P < 0.01) and V0-20 (0.79 ± 0.14 vs. 2.06 ± 0.47 beats/min/Hz, P < 0.001). These changes, which were observed under a minimal influence from sympathetic background tone, may reflect an increased contribution of the acetylcholine-sensitive potassium channel (IK,ACh) pathway after IVA, because the HR control via the IK,ACh pathway is faster and acts in the frequency range higher than the cAMP-mediated pathway.NEW & NOTEWORTHY Since ivabradine (IVA) inhibits hyperpolarization-activated cyclic nucleotide-gated channels, interactions among the sympathetic effect, vagal effect, and IVA can occur in the control of heart rate (HR). To remove the sympathetic effect, we estimated the transfer function from vagal nerve stimulation to HR under ß-blockade in anesthetized rats. IVA augmented the high-frequency dynamic gain during low- and moderate-intensity vagal nerve stimulation. Untethering the hyperpolarizing effect of acetylcholine-sensitive potassium channels after IVA may be a possible underlying mechanism.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Fármacos Cardiovasculares/farmacologia , Estimulação Elétrica , Frequência Cardíaca/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Ivabradina/farmacologia , Nervo Vago/fisiologia , Animais , Pressão Arterial/efeitos dos fármacos , Pressão Arterial/fisiologia , AMP Cíclico/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Frequência Cardíaca/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Masculino , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Propranolol/farmacologia , Ratos
7.
Int J Mol Sci ; 22(23)2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34884427

RESUMO

Little is known about the effect of lead on the activity of the vacuolar K+ channels. Here, the patch-clamp technique was used to compare the impact of lead (PbCl2) on the slow-activating (SV) and fast-activating (FV) vacuolar channels. It was revealed that, under symmetrical 100-mM K+, the macroscopic currents of the SV channels exhibited a typical slow activation and a strong outward rectification of the steady-state currents, while the macroscopic currents of the FV channels displayed instantaneous currents, which, at the positive potentials, were about three-fold greater compared to the one at the negative potentials. When PbCl2 was added to the bath solution at a final concentration of 100 µM, it decreased the macroscopic outward currents of both channels but did not change the inward currents. The single-channel recordings demonstrated that cytosolic lead causes this macroscopic effect by a decrease of the single-channel conductance and decreases the channel open probability. We propose that cytosolic lead reduces the current flowing through the SV and FV channels, which causes a decrease of the K+ fluxes from the cytosol to the vacuole. This finding may, at least in part, explain the mechanism by which cytosolic Pb2+ reduces the growth of plant cells.


Assuntos
Beta vulgaris/crescimento & desenvolvimento , Chumbo/farmacologia , Canais de Potássio/metabolismo , Vacúolos/metabolismo , Beta vulgaris/efeitos dos fármacos , Beta vulgaris/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Técnicas de Patch-Clamp , Proteínas de Plantas/efeitos dos fármacos , Proteínas de Plantas/metabolismo , Canais de Potássio/efeitos dos fármacos , Vacúolos/efeitos dos fármacos
8.
J Cell Physiol ; 235(6): 5223-5228, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31774182

RESUMO

This work aims to investigate the renal effect of hydrogen sulfide (H2 S), in the experimentally induced diabetic nephropathy, besides the role of activation of АТP-sensitive potassium (KАTP ) channel in that effect. Thirty-two adult male albino rats randomly divided into four groups: Control, streptozotocin-induced diabetic (diabetic nephropathy [DN]), DN+NaHS (the H2 S inducer), and DN+NaHS+Glibenclamide (a selective KАTP channel blocker) groups. Results showed that kidney functions in the diabetic group improved by NaHS proved by the significant decrease in the measured renal injury markers when compared with the diabetic group with an obvious role of inflammation and oxidative stress. However, the improved kidney functions produced by NaHS was reduced by the combination with Glibenclamide. Glibenclamide combination led also to a significant increase in renal total antioxidant capacity, in addition to a significant decrease in renal total nitric oxide (NO) level. Аccordingly, the results from the present work revealed that the renoprotective effects of H2 S in the case of DN through its effects on renal tissue antioxidants and NO can be partially dependent on activation of KАTP channels, while its effect on renal tissue proinflammatory cytokines is independent of it.


Assuntos
Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/genética , Estresse Oxidativo/efeitos dos fármacos , Canais de Potássio/genética , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/patologia , Glibureto/farmacologia , Humanos , Sulfeto de Hidrogênio/farmacologia , Rim/metabolismo , Rim/patologia , Masculino , Óxido Nítrico , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Ratos , Sulfetos/farmacologia
9.
Am J Physiol Heart Circ Physiol ; 319(6): H1347-H1357, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33035439

RESUMO

Reactive oxygen species (ROS) have been shown to prolong cardiac action potential duration resulting in afterdepolarizations, the cellular basis of triggered arrhythmias. As previously shown, protein kinase A type I (PKA I) is readily activated by oxidation of its regulatory subunits. However, the relevance of this mechanism of activation for cardiac pathophysiology is still elusive. In this study, we investigated the effects of oxidation-activated PKA I on cardiac electrophysiology. Ventricular cardiomyocytes were isolated from redox-dead PKA-RI Cys17Ser knock-in (KI) and wild-type (WT) mice and exposed to H2O2 (200 µmol/L) or vehicle (Veh) solution. In WT myocytes, exposure to H2O2 significantly increased oxidation of the regulatory subunit I (RI) and thus its dimerization (threefold increase in PKA RI dimer). Whole cell current clamp and voltage clamp were used to measure cardiac action potentials (APs), transient outward potassium current (Ito) and inward rectifying potassium current (IK1), respectively. In WT myocytes, H2O2 exposure significantly prolonged AP duration due to significantly decreased Ito and IK1 resulting in frequent early afterdepolarizations (EADs). Preincubation with the PKA-specific inhibitor Rp-8-Br-cAMPS (10 µmol/L) completely abolished the H2O2-dependent decrease in Ito and IK1 in WT myocytes. Intriguingly, H2O2 exposure did not prolong AP duration, nor did it decrease Ito, and only slightly enhanced EAD frequency in KI myocytes. Treatment of WT and KI cardiomyocytes with the late INa inhibitor TTX (1 µmol/L) completely abolished EAD formation. Our results suggest that redox-activated PKA may be important for H2O2-dependent arrhythmias and could be important for the development of specific antiarrhythmic drugs.NEW & NOTEWORTHY Oxidation-activated PKA type I inhibits transient outward potassium current (Ito) and inward rectifying potassium current (IK1) and contributes to ROS-induced APD prolongation as well as generation of early afterdepolarizations in murine ventricular cardiomyocytes.


Assuntos
Potenciais de Ação , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Frequência Cardíaca , Miócitos Cardíacos/enzimologia , Canais de Potássio/metabolismo , Potássio/metabolismo , Animais , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Ativação Enzimática , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Oxirredução , Estresse Oxidativo , Canais de Potássio/efeitos dos fármacos , Multimerização Proteica , Fatores de Tempo
10.
Microvasc Res ; 131: 104030, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32531353

RESUMO

Previous studies indicate that sex-related differences exist in the regulation of cutaneous vasodilation, however, the mechanisms remain unresolved. We assessed if sex-differences in young adults exist for cholinergic, nicotinic, and ß-adrenergic cutaneous vasodilation with a focus on nitric oxide synthase (NOS), cyclooxygenase (COX), and K+ channel mechanisms. In twelve young men and thirteen young women, four intradermal forearm skin sites were perfused with the following: 1) lactated Ringer's solution (control), 2) 10 mM Nω-nitro-l-arginine, a non-selective NOS inhibitor, 3) 10 mM ketorolac, a non-selective COX inhibitor, or 4) 50 mM BaCl2, a nonspecific K+ channel blocker. At all four sites, cutaneous vasodilation was induced by 1) 10 mM nicotine, a nicotinic receptor agonist, 2) 100 µM isoproterenol, a nonselective ß-adrenergic receptor agonist, and 3) 2 mM and 2000 mM acetylcholine, an acetylcholine receptor agonist. Nicotine and isoproterenol were administered for 3 min, whereas each acetylcholine dose was administered for 25 min. Regardless of treatment site, cutaneous vasodilation in response to nicotine and a high dose of acetylcholine (2000 mM) were lower in women than men. By contrast, isoproterenol induced cutaneous vasodilation was greater in women vs. men. Irrespective of sex, NOS inhibition or K+ channel blockade attenuated isoproterenol-mediated cutaneous vasodilation, whereas K+ channel blockade decreased nicotine-induced cutaneous vasodilation. Taken together, our findings indicate that while the mechanisms underlying cutaneous vasodilation are comparable between young men and women, sex-related differences in the magnitude of cutaneous vasodilation do exist and this response differs as a function of the receptor agonist.


Assuntos
Vasos Sanguíneos/enzimologia , Óxido Nítrico Sintase/metabolismo , Canais de Potássio/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Colinérgicos/metabolismo , Pele/irrigação sanguínea , Vasodilatação , Agonistas Adrenérgicos beta/farmacologia , Adulto , Vasos Sanguíneos/efeitos dos fármacos , Agonistas Colinérgicos/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Antebraço , Humanos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Receptores Nicotínicos/metabolismo , Fatores Sexuais , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Adulto Jovem
11.
Behav Pharmacol ; 31(2&3): 283-292, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32040017

RESUMO

Morphine sensitization is associated with increased locomotion and stereotypies in rats. This persistent condition has been proposed as a model of manic-like symptoms. Modifications in reward threshold are considered a central feature of mania and have been related to changes in mesocorticolimbic dopaminergic transmission. Thus, to further characterize this model, we investigated reward responses in morphine-sensitized male rats and the mechanisms underlying the behavioral phenotype. In particular, we examined the possible involvement of hyperpolarization-activated cyclic nucleotide-gated channels as they play a critical role in regulating the excitability of dopaminergic neurons. Rats were trained to self-administer sucrose to study whether morphine sensitization affected motivated behavior. Next, the dopaminergic response to sucrose was examined in the nucleus accumbens shell by in vivo microdialysis. To investigate the possible mechanisms underlying the increased dopaminergic transmission in morphine-sensitized rats, HCN2 channel expression levels in mesocorticolimbic regions were analyzed by immunoblotting. Sensitized rats showed an enhanced motivation to work for sucrose that was accompanied by an increased dopaminergic response to sucrose consumption in the nucleus accumbens shell. Moreover, HCN2 expression levels were increased in the ventral tegmental area, suggesting that their increased expression may underpin the enhanced motivation for sucrose reward and nucleus accumbens shell dopaminergic response in sensitized rats. The modified behavioral and dopaminergic reward response observed in sensitized rats supports the suggestion that the condition of morphine sensitization can be regarded as a model of manic symptoms.


Assuntos
Transtorno Bipolar/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais de Potássio/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Expressão Gênica/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Masculino , Morfina/farmacologia , Núcleo Accumbens/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Ratos , Ratos Sprague-Dawley , Recompensa , Transcriptoma/genética , Área Tegmentar Ventral/fisiologia
12.
Europace ; 22(9): 1409-1418, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32676673

RESUMO

AIMS: Non-vitamin K antagonist oral anticoagulants (NOACs) are widely used in the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation (AF). The efficacy of NOACs has been attributed in part to pleiotropic effects that are mediated through effects on thrombin, factor Xa, and their respective receptors. Direct pharmacological effects of NOACs and cardiac ion channels have not been addressed to date. We hypothesized that the favourable clinical outcome of NOAC use may be associated with previously unrecognized effects on atrial repolarizing potassium channels. METHODS AND RESULTS: This study was designed to elucidate acute pharmacological effects of NOACs on cloned ion channels Kv11.1, Kv1.5, Kv4.3, Kir2.1, Kir2.2, and K2P2.1 contributing to IKr, IKur, Ito, IK1, and IK2P K+ currents. Human genes, KCNH2, KCNA5, KCND3, KCNJ2, KCNJ12, and KCNK2, were heterologously expressed in Xenopus laevis oocytes, and currents were recorded using voltage-clamp electrophysiology. Apixaban, dabigatran, edoxaban, and rivaroxaban applied at 1 µM did not significantly affect peak current amplitudes of Kv11.1, Kv1.5, Kv4.3, Kir2.1, Kir2.2, or K2P2.1 K+ channels. Furthermore, biophysical characterization did not reveal significant effects of NOACs on current-voltage relationships of study channels. CONCLUSION: Apixaban, dabigatran, edoxaban, and rivaroxaban did not exhibit direct functional interactions with human atrial K+ channels underlying IKr, IKur, Ito, IK1, and IK2P currents that could account for beneficial clinical outcome associated with the drugs. Indirect or chronic effects and potential underlying signalling mechanisms remain to be investigated.


Assuntos
Anticoagulantes/farmacologia , Fibrilação Atrial , Canais de Potássio/efeitos dos fármacos , Acidente Vascular Cerebral , Administração Oral , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/tratamento farmacológico , Dabigatrana/farmacologia , Humanos , Piridonas/farmacologia , Rivaroxabana/farmacologia , Acidente Vascular Cerebral/prevenção & controle
13.
Med Sci Monit ; 26: e924215, 2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32470968

RESUMO

BACKGROUND Dysfunction of small conductance calcium activated potassium (SK) channels plays a vital role in atrial arrhythmogenesis. Amiodarone and dronedarone are the most effective class III antiarrhythmic drugs. It is unclear whether the antiarrhythmic effect of amiodarone and dronedarone is related to SK channel inhibition. MATERIAL AND METHODS Tissue samples were obtained from the right atria of 46 patients with normal sinus rhythm and 39 patients with chronic atrial fibrillation. Isolated atrial myocytes were obtained by enzymatic dissociation. KCNN2 (SK2) channels were transiently expressed in human embryonic kidney (HEK)-293 cells. SK currents were recorded using whole-cell conventional patch clamp techniques. RESULTS Amiodarone and dronedarone showed a concentration-dependent inhibitory effect on SK currents (IKAS) in atrial myocytes from normal sinus rhythm patients and chronic atrial fibrillation patients. The suppressed efficacy of dronedarone and amiodarone on IKAS was greater in atrial myocytes from chronic atrial fibrillation patients than that from normal sinus rhythm patients. Furthermore, in patients with chronic atrial fibrillation, the IC50 value was 2.42 µM with dronedarone and 8.03 µM with amiodarone. In HEK-293 cells with transiently transfected SK2 channels, both dronedarone and amiodarone had a dose-dependent inhibitory effect on IKAS. The IC50 value was 1.7 µM with dronedarone and 7.2 µM with amiodarone in cells from patients with chronic atrial fibrillation. Compared to amiodarone, dronedarone is more efficacy to inhibit IKAS and could be a potential intervention for patients with chronic atrial fibrillation. CONCLUSIONS Dronedarone provides a great degree of IKAS inhibition in atrial myocytes from chronic atrial fibrillation than amiodarone. IKAS might be a potential target of amiodarone and dronedarone for the management of chronic atrial fibrillation.


Assuntos
Fibrilação Atrial/tratamento farmacológico , Dronedarona/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Adulto , Amiodarona/metabolismo , Amiodarona/farmacologia , Antiarrítmicos/uso terapêutico , Fibrilação Atrial/metabolismo , Cálcio/metabolismo , Dronedarona/metabolismo , Feminino , Células HEK293 , Átrios do Coração/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo
14.
Mar Drugs ; 18(8)2020 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-32748868

RESUMO

Several experimental studies have recently demonstrated that temporary autonomic block using botulinum toxin (BoNT/A1) might be a novel option for the treatment of atrial fibrillation. However, the assessment of antiarrhythmic properties of BoNT has so far been limited, relying exclusively on vagal stimulation and rapid atrial pacing models. The present study examined the antiarrhythmic effect of specially formulated BoNT/A1-chitosan nanoparticles (BTN) in calcium chloride-, barium chloride- and electrically induced arrhythmia rat models. BTN enhanced the effect of BoNT/A1. Subepicardial injection of BTN resulted in a significant antiarrhythmic effect in investigated rat models. BTN formulation antagonizes arrhythmia induced by the activation of Ca, K and Na channels.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/prevenção & controle , Toxinas Botulínicas Tipo A/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Nanopartículas , Potenciais de Ação/efeitos dos fármacos , Animais , Antiarrítmicos/química , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Toxinas Botulínicas Tipo A/química , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Quitosana/química , Modelos Animais de Doenças , Composição de Medicamentos , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Masculino , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos Wistar , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
15.
Int J Mol Sci ; 21(5)2020 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-32121388

RESUMO

Lapatinib (LAP) and sorafenib (SOR) are multitargeted tyrosine kinase inhibitors (TKIs) with antineoplastic properties. In clinical observations, LAP and SOR may contribute to QTc prolongation, but the detailed mechanism for this has been largely unexplored. In this study, we investigated whether LAP and SOR affect the activities of membrane ion channels. Using a small animal model and primary cardiomyocytes, we studied the impact of LAP and SOR on Na+ and K+ currents. We found that LAP-induced QTc prolongation in mice was reversed by isoproterenol. LAP or SOR suppressed the amplitude of the slowly activating delayed-rectifier K+ current (IK(S)) in H9c2 cells in a time- and concentration-dependent fashion. The LAP-mediated inhibition of IK(S) was reversed by adding isoproterenol or meclofenamic acid, but not by adding diazoxide. The steady-state activation curve of IK(S) during exposure to LAP or SOR was shifted toward a less negative potential, with no change in the gating charge required to activate the current. LAP shortened the recovery from IK(S) deactivation. As rapid repetitive stimuli, the IK(S) amplitude decreased; however; the LAP-induced inhibition of IK(S) remained effective. LAP or SOR alone also suppressed inwardly rectifying K+ and voltage-gated Na+ current in neonatal rat ventricular myocytes. The inhibition of ionic currents during exposure to TKIs could be an important mechanism underlying changes in QTc intervals.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Animais , Humanos , Canais Iônicos/antagonistas & inibidores , Transporte de Íons/efeitos dos fármacos , Isoproterenol/farmacologia , Lapatinib/efeitos adversos , Camundongos , Miócitos Cardíacos/metabolismo , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Potássio/química , Potássio/metabolismo , Inibidores de Proteínas Quinases/efeitos adversos , Ratos , Sódio/química , Sódio/metabolismo , Sorafenibe/efeitos adversos
16.
Int J Mol Sci ; 21(1)2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-31948124

RESUMO

Pterostilbene (PTER), a natural dimethylated analog of resveratrol, has been demonstrated to produce anti-neoplastic or neuroprotective actions. However, how and whether this compound can entail any perturbations on ionic currents in electrically excitable cells remains unknown. In whole-cell current recordings, addition of PTER decreased the amplitude of macroscopic Ih during long-lasting hyperpolarization in GH3 cells in a concentration-dependent manner, with an effective IC50 value of 0.84 µM. Its presence also shifted the activation curve of Ih along the voltage axis to a more hyperpolarized potential, by 11 mV. PTER at a concentration greater than 10 µM could also suppress l-type Ca2+ and transient outward K+ currents in GH3 cells. With the addition of PTER, IK(Ca) amplitude was increased, with an EC50 value of 2.23 µM. This increase in IK(Ca) amplitude was attenuated by further addition of verruculogen, but not by tolbutamide or TRAM-39. Neither atropine nor nicotine, in the continued presence of PTER, modified the PTER-stimulated IK(Ca). PTER (10 µM) slightly suppressed the amplitude of l-type Ca2+ current and transient outward K+ current. The presence of PTER (3 µM) was also effective at increasing the open-state probability of large-conductance Ca2+-activated K+ (BKCa) channels identified in hippocampal mHippoE-14 neurons; however, its inability to alter single-channel conductance was detected. Our study highlights evidence to show that PTER has the propensity to perturb ionic currents (e.g., Ih and IK(Ca)), thereby influencing the functional activities of neurons, and neuroendocrine or endocrine cells.


Assuntos
Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiologia , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Potenciais da Membrana/fisiologia , Camundongos , Neurônios/química , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Ratos
17.
Molecules ; 25(11)2020 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-32527034

RESUMO

Carveol is a monoterpene present in the structure of many plant products. It has a variety of biological activities: antioxidant, anticancer and vasorelaxation. However, studies investigating the effect of monoterpenoids on human vessels have not yet been described. Thus, the present study aimed to characterize the effect of (-)-carveol on human umbilical arteries (HUAs). HUA ring preparations were isolated and subjected to isometric tension recordings of umbilical artery smooth muscle contractions. (-)-Carveol exhibited a significant vasorelaxant effect on KCl and 5-HT-induced contractions, obtaining EC50 values of 344.25 ± 8.4 and 175.82 ± 4.05 µM, respectively. The participation of calcium channels in the relaxation produced by (-)-carveol was analyzed using vessels pre-incubated with (-)-carveol (2000 µM) in a calcium-free medium, where the induction of contractions was abolished. The vasorelaxant effect of (-)-carveol on HUAs was reduced by tetraethylammonium (TEA), which increased the (-)-carveol EC50 to 484.87 ± 6.55 µM. The present study revealed that (-)-carveol possesses a vasorelaxant activity in HUAs, which was dependent on the opening of calcium and potassium channels. These results pave the way for further studies involving the use of monoterpenoids for the vasodilatation of HUAs. These molecules have the potential to treat diseases such as pre-eclampsia, which is characterized by resistance in umbilical arteries.


Assuntos
Canais de Cálcio/fisiologia , Monoterpenos Cicloexânicos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Canais de Potássio/fisiologia , Artérias Umbilicais/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Canais de Cálcio/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Humanos , Canais de Potássio/efeitos dos fármacos , Artérias Umbilicais/metabolismo
18.
J Neurosci ; 38(19): 4505-4520, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29650696

RESUMO

Neuropeptide Y (NPY) expression is tightly linked with the development of stress resilience in rodents and humans. Local NPY injections targeting the basolateral amygdala (BLA) produce long-term behavioral stress resilience in male rats via an unknown mechanism. Previously, we showed that activation of NPY Y1 receptors hyperpolarizes BLA principal neurons (PNs) through inhibition of the hyperpolarization-activated, depolarizing H-current, Ih The present studies tested whether NPY treatment induces stress resilience by modulating Ih NPY (10 pmol) was delivered daily for 5 d bilaterally into the BLA to induce resilience; thereafter, the electrophysiological properties of PNs and the expression of Ih in the BLA were characterized. As reported previously, increases in social interaction (SI) times persisted weeks after completion of NPY administration. In vitro intracellular recordings showed that repeated intra-BLA NPY injections resulted in hyperpolarization of BLA PNs at 2 weeks (2W) and 4 weeks (4W) after NPY treatment. At 2W, spontaneous IPSC frequencies were increased, whereas at 4W, resting Ih was markedly reduced and accompanied by decreased levels of HCN1 mRNA and protein expression in BLA. Knock-down of HCN1 channels in the BLA with targeted delivery of lentivirus containing HCN1-shRNA increased SI beginning 2W after injection and induced stress resilience. NPY treatment induced sequential, complementary changes in the inputs to BLA PNs and their postsynaptic properties that reduce excitability, a mechanism that contributes to less anxious behavior. Furthermore, HCN1 knock-down mimicked the increases in SI and stress resilience observed with NPY, indicating the importance of Ih in stress-related behavior.SIGNIFICANCE STATEMENT Resilience improves mental health outcomes in response to adverse situations. Neuropeptide Y (NPY) is associated with decreased stress responses and the expression of resilience in rodents and humans. Single or repeated injections of NPY into the basolateral amygdala (BLA) buffer negative behavioral effects of stress and induce resilience in rats, respectively. Here, we demonstrate that repeated administration of NPY into the BLA unfolds several cellular mechanisms that decrease the activity of pyramidal output neurons. One key mechanism is a reduction in levels of the excitatory ion channel HCN1. Moreover, shRNA knock-down of HCN1 expression in BLA recapitulates some of the actions of NPY and causes potent resilience to stress, indicating that this channel may be a possible target for therapy.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Canais de Potássio/efeitos dos fármacos , Resiliência Psicológica/efeitos dos fármacos , Tonsila do Cerebelo/citologia , Animais , Ansiedade/genética , Ansiedade/psicologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Relações Interpessoais , Masculino , Microinjeções , Neuropeptídeo Y/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
19.
J Cell Physiol ; 234(10): 18086-18097, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30887515

RESUMO

S17 is a clonogenic bone marrow stromal (BMS) cell line derived from mouse that has been extensively used to assess both human and murine hematopoiesis support capacity. However, very little is known about the expression of potassium ion channels and their function in cell survival and migration in these cells. Thus, the present study was designed to characterize potassium ion channels using electrophysiological and molecular biological approaches in S17 BMS cells. The whole-cell configuration of the patch clamp technique has been applied to identify potassium ion currents and reverse transcription polymerase chain reaction (RT-PCR) used to determine their molecular identities. Based on gating kinetics and pharmacological modulation of the macroscopic currents we found the presence of four functional potassium ion channels in S17 BMS cells. These include a current rapidly activated and inactivated, tetraethylammonium-sensitive, (IKV ) in most (50%) cells; a fast activated and rapidly inactivating A-type K + current (IK A -like); a delayed rectifier K + current (IK DR ) and an inward rectifier potassium current (IK IR ), found in, respectively 4.5%, 26% and 24% of these cells. RT-PCR confirmed the presence of mRNA transcripts for the alpha subunit of the corresponding functional ion channels. Additionally, functional assays were performed to investigate the importance of potassium currents in cell survival and migration. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analyses revealed a reduction in cell viability, while wound healing assays revealed reduced migration potential in cells incubated with different potassium channel blockers. In conclusion, our data suggested that potassium currents might play a role in the maintenance of overall S17 cell ionic homeostasis directly affecting cell survival and migration.


Assuntos
Movimento Celular , Células-Tronco Mesenquimais/metabolismo , Canais de Potássio/metabolismo , Potássio/metabolismo , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular , Ativação do Canal Iônico , Cinética , Potenciais da Membrana , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Transdução de Sinais
20.
Pflugers Arch ; 471(7): 1007-1023, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31093757

RESUMO

Ion-transport properties of the epithelium of the cecum, the biggest fermental chamber in non-ruminant species, are largely unknown. Recently, in Ussing chamber experiments, segmental differences in basal short-circuit current (Isc) in rat corpus ceci were observed. The oral segment usually exhibited a much lower or even negative basal Isc in comparison with the aboral segment. The aim of the present study was the closer characterization of these differences. Basal Isc was inhibited by bumetanide and tetrodotoxin in both segments, whereas indomethacin reduced basal Isc only in the aboral corpus. Amiloride did not inhibit basal Isc suggesting that spontaneous anion secretion (but not electrogenic Na+ absorption via ENaC) contributes to the baseline current. In both segments, mucosally applied K+ channel blockers increased Isc indicating a spontaneous K+ secretion. Basolateral depolarization was used to characterize the ion conductances in the apical membrane. When a Cl- gradient was applied, apical Cl- conductance stimulated by carbachol and by forskolin was revealed. When the Cl- gradient was omitted and instead a K+ gradient was used to drive currents across apical K+ channels, a Ba2+-sensititve K+ conductance was observed in both segments, and carbachol stimulated this conductance leading to a negative Isc. Conversely, forskolin induced a positive Isc under these conditions which was dependent on the presence of mucosal Na+ consistent with electrogenic Na+ absorption. This current was reduced by amiloride and several blockers of members of the TRP channel superfamily. These results indicate that similar transport mechanisms are involved in electrogenic ion transport across cecal oral and aboral segments, but with a higher spontaneous prostaglandin production in the aboral segment responsible for higher basal transport rates of both anions and cations.


Assuntos
Ceco/metabolismo , Transporte de Íons/fisiologia , Íons/metabolismo , Amilorida/farmacologia , Animais , Bumetanida/farmacologia , Ceco/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cloretos/metabolismo , Colforsina/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Transporte de Íons/efeitos dos fármacos , Masculino , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Ratos Wistar , Sódio/metabolismo , Tetrodotoxina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA