Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
1.
Glia ; 68(10): 2119-2135, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32220118

RESUMO

Spinal microglia change their phenotype and proliferate after nerve injury, contributing to neuropathic pain. For the first time, we have characterized the electrophysiological properties of microglia and the potential role of microglial potassium channels in the spared nerve injury (SNI) model of neuropathic pain. We observed a strong increase of inward currents restricted at 2 days after injury associated with hyperpolarization of the resting membrane potential (RMP) in microglial cells compared to later time-points and naive animals. We identified pharmacologically and genetically the current as being mediated by Kir2.1 ion channels whose expression at the cell membrane is increased 2 days after SNI. The inhibition of Kir2.1 with ML133 and siRNA reversed the RMP hyperpolarization and strongly reduced the currents of microglial cells 2 days after SNI. These electrophysiological changes occurred coincidentally to the peak of microglial proliferation following nerve injury. In vitro, ML133 drastically reduced the proliferation of BV2 microglial cell line after both 2 and 4 days in culture. In vivo, the intrathecal injection of ML133 significantly attenuated the proliferation of microglia and neuropathic pain behaviors after nerve injury. In summary, our data implicate Kir2.1-mediated microglial proliferation as an important therapeutic target in neuropathic pain.


Assuntos
Proliferação de Células/fisiologia , Microglia/metabolismo , Neuralgia/metabolismo , Bloqueadores dos Canais de Potássio/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Medula Espinal/metabolismo , Animais , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Imidazóis/administração & dosagem , Injeções Espinhais , Masculino , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Neuralgia/prevenção & controle , Fenantrolinas/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos
2.
Biochem Biophys Res Commun ; 532(2): 265-270, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32863001

RESUMO

Methotrexate (MTX) has been widely used for the treatment of many types of autoimmune diseases, such as rheumatoid arthritis, psoriasis and dermatomyositis. However, its pharmacological mechanism is still unclear completely. In this study, we found that MTX is a potent and selective inhibitor of the Kv1.3 channel, a class of potassium channels highly associated with autoimmune diseases. Electrophysiological experiments showed that MTX inhibited human Kv1.3 channel with an IC50 of 41.5 ± 24.9 nM, and 1 µM MTX inhibited 32.6 ± 1.3% and 25.6 ± 2.2% of human Kv1.1 and Kv1.2 channel currents, respectively. These data implied the unique selectivity of MTX towards the Kv1.3 channel. Excitingly, using channel activation and chimeric experiments, we found that MTX bound to the outer pore region of Kv1.3 channel. Mutagenesis experiments in the Kv.3 channel extracellular pore region further showed that the Dsp371, Thr373 and His399 residues of outer pore region of Kv1.3 channel played important roles in MTX inhibiting activities. In conclusion, MTX inhibited Kv1.3 channel by targeting extracellular pore region, which is different form all the report small molecules, such as PAP-1 and 4-AP, but similar with many natural animal toxin peptides, such as ChTX, ShK and BmKTX. To the best of our knowledge, MTX is the first small molecular scaffold targeting the Kv1.3 channel extracellular pore region, suggesting its potential applications for designing novel Kv1.3 lead drugs and treating Kv1.3 channel-associated autoimmune diseases.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Metotrexato/química , Metotrexato/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Sítios de Ligação , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Canal de Potássio Kv1.3/genética , Metotrexato/administração & dosagem , Mutagênese , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/química
3.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R390-R398, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913684

RESUMO

Local skin heating to 42°C causes cutaneous thermal hyperemia largely via nitric oxide (NO) synthase (NOS)-related mechanisms. We assessed the hypothesis that ATP-sensitive K+ (KATP) channels interact with NOS to mediate cutaneous thermal hyperemia. In 13 young adults (6 women, 7 men), cutaneous vascular conductance (CVC) was measured at four intradermal microdialysis sites that were continuously perfused with 1) lactated Ringer solution (control), 2) 5 mM glibenclamide (KATP channel blocker), 3) 20 mM NG-nitro-l-arginine methyl ester (NOS inhibitor), or 4) a combination of KATP channel blocker and NOS inhibitor. Local skin heating to 42°C was administered at all four treatment sites to elicit cutaneous thermal hyperemia. Thirty minutes after the local heating, 1.25 mM pinacidil (KATP channel opener) and subsequently 25 mM sodium nitroprusside (NO donor) were administered to three of the four sites (each 25-30 min). The local heating-induced prolonged elevation in CVC was attenuated by glibenclamide (19%), but the transient initial peak was not. However, glibenclamide had no effect on the prolonged elevation in CVC in the presence of NOS inhibition. Pinacidil caused an elevation in CVC, but this response was abolished at the glibenclamide-treated skin site, demonstrating its effectiveness as a KATP channel blocker. The pinacidil-induced increase in CVC was unaffected by NOS inhibition, whereas the increase in CVC elicited by sodium nitroprusside was partly (15%) inhibited by glibenclamide. In summary, we showed an interactive effect of KATP channels and NOS for the plateau of cutaneous thermal hyperemia. This interplay may reflect a vascular smooth muscle cell KATP channel activation by NO.


Assuntos
Hiperemia/enzimologia , Canais KATP/metabolismo , Microcirculação , Microvasos/enzimologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Pele/irrigação sanguínea , Vasodilatação , Adulto , Velocidade do Fluxo Sanguíneo , Inibidores Enzimáticos/administração & dosagem , Feminino , Humanos , Hiperemia/etiologia , Hiperemia/fisiopatologia , Hipotermia Induzida , Ativação do Canal Iônico , Canais KATP/antagonistas & inibidores , Masculino , Microcirculação/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Microvasos/fisiopatologia , Doadores de Óxido Nítrico/administração & dosagem , Óxido Nítrico Sintase/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/administração & dosagem , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem , Adulto Jovem
4.
Can J Physiol Pharmacol ; 98(11): 753-762, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33095677

RESUMO

The aim of this study was to examine if the peripheral antinociceptive effects of the opioid agonist/antagonist nalbuphine and buprenorphine involve the sequential participation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) synthesis followed by K+ channel opening in the formalin test. Wistar rats (180-220 g) were injected in the dorsal surface of the right hind paw with formalin (1%). Rats received a subcutaneous (s.c.) injection into the dorsal surface of the paw of vehicles or increasing doses of nalbuphine (50-200 µg/paw) or buprenorphine (1-5 µg/paw) 20 min before formalin injection into the paw. Nalbuphine antinociception was reversed by the s.c. injection into the paw of the inhibitor of NO synthesis (NG-nitro-l-arginine methyl ester (L-NAME)), by the inhibitor of guanylyl cyclase (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ)), by the Kir6.1-2, ATP-sensitive K+ channel inhibitors (glibenclamide and glipizide), by the KCa2.1-3, small conductance Ca2+-activated K+ channel blocker (apamin), by the KCa1.1, large conductance Ca2+-activated K+ channel blocker (charybdotoxin), and by the KV, voltage-dependent K+ channel inhibitors (4-aminopyridine (4-AP) and tetraethylammonium chloride (TEA)). The antinociceptive effect produced by buprenorphine was blocked by the s.c. injection of 4-AP and TEA but not by L-NAME, ODQ, glibenclamide, glipizide, apamin, or charybdotoxin. The present results provide evidence for differences in peripheral mechanisms of action between these opioid drugs.


Assuntos
Analgésicos Opioides/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Buprenorfina/farmacologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Glibureto/administração & dosagem , Humanos , Injeções Subcutâneas , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , NG-Nitroarginina Metil Éster/administração & dosagem , Nalbufina/farmacologia , Óxido Nítrico/metabolismo , Nociceptividade/fisiologia , Dor/induzido quimicamente , Dor/diagnóstico , Medição da Dor , Bloqueadores dos Canais de Potássio/administração & dosagem , Ratos , Receptores Opioides/metabolismo , Transdução de Sinais/fisiologia
5.
Curr Pain Headache Rep ; 24(12): 77, 2020 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-33270149

RESUMO

PURPOSE OF REVIEW: Migraine remains a challenging condition to treat, thus highlighting the need for a better understanding of its molecular mechanisms. This review intends to unravel a new emerging target in migraine pathophysiology, the adenosine 5'-triphosphate-sensitive K+ (KATP) channel. RECENT FINDINGS: KATP channel is a common denominator in the cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) mediated intracellular cascades, both of which are involved in migraine. Intravenous infusion of KATP channel opener, levcromakalim, provoked migraine attack associated with dilation of extracerebral arteries in all persons with migraine. Preclinical and clinical studies implicate KATP channels in migraine initiation. KATP channel is a novel therapeutic target for the acute and preventive treatment of migraine. Future studies are warranted to provide a better understanding of the role of KATP channel subgroups in migraine.


Assuntos
Canais KATP/agonistas , Canais KATP/antagonistas & inibidores , Transtornos de Enxaqueca/tratamento farmacológico , Transtornos de Enxaqueca/fisiopatologia , Bloqueadores dos Canais de Potássio/administração & dosagem , Animais , Broncodilatadores/efeitos adversos , Cromakalim/efeitos adversos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Canais KATP/metabolismo , Transtornos de Enxaqueca/induzido quimicamente , Transtornos de Enxaqueca/metabolismo
6.
J Biol Chem ; 293(18): 7040-7057, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29545312

RESUMO

Cardiac potassium channels encoded by human ether-à-go-go-related gene (hERG) are major targets for structurally diverse drugs associated with acquired long QT syndrome. This study characterized hERG channel inhibition by a minimally structured high-affinity hERG inhibitor, Cavalli-2, composed of three phenyl groups linked by polymethylene spacers around a central amino group, chosen to probe the spatial arrangement of side chain groups in the high-affinity drug-binding site of the hERG pore. hERG current (IhERG) recorded at physiological temperature from HEK293 cells was inhibited with an IC50 of 35.6 nm with time and voltage dependence characteristic of blockade contingent upon channel gating. Potency of Cavalli-2 action was markedly reduced for attenuated inactivation mutants located near (S620T; 54-fold) and remote from (N588K; 15-fold) the channel pore. The S6 Y652A and F656A mutations decreased inhibitory potency 17- and 75-fold, respectively, whereas T623A and S624A at the base of the selectivity filter also decreased potency (16- and 7-fold, respectively). The S5 helix F557L mutation decreased potency 10-fold, and both F557L and Y652A mutations eliminated voltage dependence of inhibition. Computational docking using the recent cryo-EM structure of an open channel hERG construct could only partially recapitulate experimental data, and the high dependence of Cavalli-2 block on Phe-656 is not readily explainable in that structure. A small clockwise rotation of the inner (S6) helix of the hERG pore from its configuration in the cryo-EM structure may be required to optimize Phe-656 side chain orientations compatible with high-affinity block.


Assuntos
Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/química , Bloqueadores dos Canais de Potássio/farmacologia , Alanina/química , Simulação por Computador , Microscopia Crioeletrônica , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Ativação do Canal Iônico/efeitos dos fármacos , Simulação de Acoplamento Molecular , Mutação , Técnicas de Patch-Clamp , Fenilalanina/química , Bloqueadores dos Canais de Potássio/administração & dosagem , Ligação Proteica , Conformação Proteica
7.
Ann Noninvasive Electrocardiol ; 24(6): e12679, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31347753

RESUMO

BACKGROUND: Human ether-à-go-go-related gene (hERG) potassium-channel block represents a harmful side effect of drug therapy that may cause torsade de pointes (TdP). Analysis of ventricular repolarization through electrocardiographic T-wave features represents a noninvasive way to accurately evaluate the TdP risk in drug-safety studies. This study proposes an artificial neural network (ANN) for noninvasive electrocardiography-based classification of the hERG potassium-channel block. METHODS: The data were taken from the "ECG Effects of Ranolazine, Dofetilide, Verapamil, and Quinidine in Healthy Subjects" Physionet database; they consisted of median vector magnitude (VM) beats of 22 healthy subjects receiving a single 500 µg dose of dofetilide. Fourteen VM beats were considered for each subject, relative to time-points ranging from 0.5 hr before to 14.0 hr after dofetilide administration. For each VM, changes in two indexes accounting for the early and the late phases of repolarization, ΔERD30% and ΔTS/A , respectively, were computed as difference between values at each postdose time-point and the predose time-point. Thus, the dataset contained 286 ΔERD30% -ΔTS/A pairs, partitioned into training, validation, and test sets (114, 29, and 143 pairs, respectively) and used as inputs of a two-layer feedforward ANN with two target classes: high block (HB) and low block (LB). Optimal ANN (OANN) was identified using the training and validation sets and tested on the test set. RESULTS: Test set area under the receiver operating characteristic was 0.91; sensitivity, specificity, accuracy, and precision were 0.93, 0.83, 0.92, and 0.96, respectively. CONCLUSION: OANN represents a reliable tool for noninvasive assessment of the hERG potassium-channel block.


Assuntos
Eletrocardiografia/métodos , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Redes Neurais de Computação , Fenetilaminas/administração & dosagem , Bloqueadores dos Canais de Potássio/administração & dosagem , Sulfonamidas/administração & dosagem , Humanos
8.
J Neurosci ; 37(23): 5648-5658, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28483976

RESUMO

The voltage-gated K+ channel Kv2.1 has been intimately linked with neuronal apoptosis. After ischemic, oxidative, or inflammatory insults, Kv2.1 mediates a pronounced, delayed enhancement of K+ efflux, generating an optimal intracellular environment for caspase and nuclease activity, key components of programmed cell death. This apoptosis-enabling mechanism is initiated via Zn2+-dependent dual phosphorylation of Kv2.1, increasing the interaction between the channel's intracellular C-terminus domain and the SNARE (soluble N-ethylmaleimide-sensitive factor activating protein receptor) protein syntaxin 1A. Subsequently, an upregulation of de novo channel insertion into the plasma membrane leads to the critical enhancement of K+ efflux in damaged neurons. Here, we investigated whether a strategy designed to interfere with the cell death-facilitating properties of Kv2.1, specifically its interaction with syntaxin 1A, could lead to neuroprotection following ischemic injury in vivo The minimal syntaxin 1A-binding sequence of Kv2.1 C terminus (C1aB) was first identified via a far-Western peptide screen and used to create a protherapeutic product by conjugating C1aB to a cell-penetrating domain. The resulting peptide (TAT-C1aB) suppressed enhanced whole-cell K+ currents produced by a mutated form of Kv2.1 mimicking apoptosis in a mammalian expression system, and protected cortical neurons from slow excitotoxic injury in vitro, without influencing NMDA-induced intracellular calcium responses. Importantly, intraperitoneal administration of TAT-C1aB in mice following transient middle cerebral artery occlusion significantly reduced ischemic stroke damage and improved neurological outcome. These results provide strong evidence that targeting the proapoptotic function of Kv2.1 is an effective and highly promising neuroprotective strategy.SIGNIFICANCE STATEMENT Kv2.1 is a critical regulator of apoptosis in central neurons. It has not been determined, however, whether the cell death-enabling function of this K+ channel can be selectively targeted to improve neuronal survival following injury in vivo The experiments presented here demonstrate that the cell death-specific role of Kv2.1 can be uniquely modulated to provide neuroprotection in an animal model of acute ischemic stroke. We thus reveal a novel therapeutic strategy for neurological disorders that are accompanied by Kv2.1-facilitated forms of cell death.


Assuntos
Apoptose/efeitos dos fármacos , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia , Animais , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Feminino , Masculino , Bloqueadores dos Canais de Potássio/administração & dosagem , Proteínas Qa-SNARE/antagonistas & inibidores , Proteínas Qa-SNARE/metabolismo , Ratos , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
9.
Neurobiol Dis ; 109(Pt A): 137-147, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29045814

RESUMO

A balance between excitation and inhibition is necessary to maintain stable brain network dynamics. Traditionally, seizure activity is believed to arise from the breakdown of this delicate balance in favor of excitation with loss of inhibition. Surprisingly, recent experimental evidence suggests that this conventional view may be limited, and that inhibition plays a prominent role in the development of epileptiform synchronization. Here, we explored the role of the KCC2 co-transporter in the onset of inhibitory network-induced seizures. Our experiments in acute mouse brain slices, of either sex, revealed that optogenetic stimulation of either parvalbumin- or somatostatin-expressing interneurons induced ictal discharges in rodent entorhinal cortex during 4-aminopyridine application. These data point to a proconvulsive role of GABAA receptor signaling that is independent of the inhibitory input location (i.e., dendritic vs. somatic). We developed a biophysically realistic network model implementing dynamics of ion concentrations to explore the mechanisms leading to inhibitory network-induced seizures. In agreement with experimental results, we found that stimulation of the inhibitory interneurons induced seizure-like activity in a network with reduced potassium A-current. Our model predicts that interneuron stimulation triggered an increase of interneuron firing, which was accompanied by an increase in the intracellular chloride concentration and a subsequent KCC2-dependent gradual accumulation of the extracellular potassium promoting epileptiform ictal activity. When the KCC2 activity was reduced, stimulation of the interneurons was no longer able to induce ictal events. Overall, our study provides evidence for a proconvulsive role of GABAA receptor signaling that depends on the involvement of the KCC2 co-transporter.


Assuntos
Sincronização Cortical , Epilepsia/fisiopatologia , Interneurônios/fisiologia , Potássio/metabolismo , Convulsões/fisiopatologia , Simportadores/fisiologia , 4-Aminopiridina/administração & dosagem , Animais , Córtex Entorrinal/metabolismo , Córtex Entorrinal/fisiopatologia , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Feminino , Interneurônios/metabolismo , Masculino , Camundongos , Redes Neurais de Computação , Parvalbuminas/metabolismo , Bloqueadores dos Canais de Potássio/administração & dosagem , Receptores de GABA-A/fisiologia , Convulsões/induzido quimicamente , Convulsões/metabolismo , Somatostatina/metabolismo , Simportadores/metabolismo , Cotransportadores de K e Cl-
10.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R693-R699, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29341826

RESUMO

µ-Opioid G protein-coupled receptors (MOR) interact with ion channels to decrease neuronal excitability. In humans, intrathecal administration of the MOR agonist fentanyl inhibits the exercise pressor reflex, an effect that can be attributed to either the opening of inward rectifying potassium channels (GIRK) or the closing of N-type calcium channels. The purpose of this study was to determine if the highly selective MOR agonist [d-Ala2, N-MePhe4,Gly-ol]-enkephalin (DAMGO) attenuates the exercise pressor reflex and which of these two channels are responsible for this effect. In decerebrate rats, we determined the effect of intrathecal injection of either tertiapin-LQ, which blocks the GIRK channel or ω-conotoxin-GVIA, which blocks the N-type calcium channel on the exercise pressor reflex, which was evoked by contracting the triceps surae muscles. Initially, we established that intrathecal injection of DAMGO inhibited the exercise pressor reflex relative to no intrathecal injection or intrathecal saline injection ( P < 0.001, n = 5). We then found that intrathecal injection of two doses of tertiapin-LQ (1 and 10 µg) had no effect on the exercise pressor reflex ( n = 6 and n = 7, respectively; P > 0.05). Importantly, neither dose of tertiapin-LQ prevented the DAMGO-induced inhibition of the exercise pressor reflex. Last, we found that intrathecal injection of ω-conotoxin-GVIA markedly attenuated the exercise pressor reflex ( P < 0.001, n = 7). The cardioaccelerator response to contraction did not appear to be effected in any of the experiments. We conclude that N-type voltage-gated calcium channel inhibition appears to be the mechanism by which MOR activation inhibits the exercise pressor reflex in decerebrate rats.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ativação do Canal Iônico , Músculo Esquelético/inervação , Inibição Neural , Esforço Físico , Receptores Opioides mu/metabolismo , Reflexo , Medula Espinal/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Bloqueadores dos Canais de Cálcio/administração & dosagem , Sinalização do Cálcio/efeitos dos fármacos , Estado de Descerebração , Ala(2)-MePhe(4)-Gly(5)-Encefalina/administração & dosagem , Injeções Espinhais , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Contração Muscular , Inibição Neural/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Bloqueadores dos Canais de Potássio/administração & dosagem , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos , Reflexo/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos
11.
Microvasc Res ; 117: 22-27, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29247720

RESUMO

Local skin heating to 42°C causes rapid increases in cutaneous perfusion (initial peak), followed by a brief nadir and subsequent sustained elevation (plateau). Several studies have demonstrated that nitric oxide synthase (NOS) largely contributes to the plateau response during local heating. In this study, we tested the hypothesis that voltage-gated potassium (Kv) channels contribute to the plateau of the cutaneous vasodilation during local heating through NOS-dependent mechanisms. Eleven young males (25±4years) participated in this study wherein cutaneous vascular conductance (CVC) was measured at four intradermal microdialysis sites that were continuously perfused with either 1) lactated Ringer (Control), 2) 10mM 4-aminopyridine (Kv channel blocker), 3) 10mM Nω-Nitro-L-arginine (NOS inhibitor), or 4) a combination of 4-aminopyridine and Nω-Nitro-L-arginine. In comparison to the Control site, the inhibition of Kv channels alone attenuated the increase in CVC observed at the initial peak, nadir, and plateau phases measured during local heating; in contrast, the inhibition of NOS alone attenuated the increase in CVC at the nadir and plateau phases only (e.g., plateau response: Control site: 59±5%max, Kv channel blockade site: 49±8%max, NOS inhibition site: 35±11%max, combined inhibition site: 40±12%max). Further, no effect of Kv channel blockade on CVC was measured at any phase of the local heating response when the modulating influence of NOS was simultaneously removed. We show that Kv channels and NOS contribute to the local heating mediated sustained increase (i.e., plateau) in cutaneous vasodilation in an interactive manner. (243/250 words).


Assuntos
Artérias/enzimologia , Calefação , Hipertermia Induzida , Óxido Nítrico Sintase/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Pele/irrigação sanguínea , Vasodilatação , Administração Cutânea , Adulto , Artérias/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Humanos , Masculino , Microdiálise , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/administração & dosagem , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Adulto Jovem
12.
Can J Physiol Pharmacol ; 96(1): 18-25, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28892643

RESUMO

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and a major cause of morbidity and mortality. Traditional antiarrhythmic agents used for restoration of sinus rhythm have limited efficacy in long-term AF and they may possess ventricular proarrhythmic adverse effects, especially in patients with structural heart disease. The acetylcholine receptor-activated potassium channel (IK,ACh) represents an atrial selective target for future AF management. We investigated the effects of the IK,ACh blocker tertiapin-Q (TQ), a derivative of the honeybee toxin tertiapin, on chronic atrial tachypacing-induced AF in conscious dogs, without the influence of anesthetics that modulate a number of cardiac ion channels. Action potentials (APs) were recorded from right atrial trabeculae isolated from dogs with AF. TQ significantly and dose-dependently reduced AF incidence and AF episode duration, prolonged atrial effective refractory period, and prolonged AP duration. The reference drugs propafenone and dofetilide, both used in the clinical management of AF, exerted similar effects against AF in vivo. Dofetilide prolonged atrial AP duration, whereas propafenone increased atrial conduction time. TQ and propafenone did not affect the QT interval, whereas dofetilide prolonged the QT interval. Our results show that inhibition of IK,ACh may represent a novel, atrial-specific target for the management of AF in chronic AF.


Assuntos
Potenciais de Ação , Fibrilação Atrial/tratamento farmacológico , Remodelamento Atrial , Estado de Consciência , Átrios do Coração/fisiopatologia , Bloqueadores dos Canais de Potássio/uso terapêutico , Canais de Potássio/metabolismo , Receptores Colinérgicos/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Fibrilação Atrial/fisiopatologia , Remodelamento Atrial/efeitos dos fármacos , Venenos de Abelha/administração & dosagem , Venenos de Abelha/farmacologia , Venenos de Abelha/uso terapêutico , Estado de Consciência/efeitos dos fármacos , Cães , Eletrocardiografia , Átrios do Coração/efeitos dos fármacos , Masculino , Fenetilaminas/administração & dosagem , Fenetilaminas/farmacologia , Fenetilaminas/uso terapêutico , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/farmacologia , Propafenona/administração & dosagem , Propafenona/farmacologia , Propafenona/uso terapêutico , Período Refratário Eletrofisiológico/efeitos dos fármacos , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico
13.
J Neuroinflammation ; 14(1): 203, 2017 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-29037241

RESUMO

BACKGROUND: Reactive astrogliosis is one of the significantly pathological features in ischemic stroke accompanied with changes in gene expression, morphology, and proliferation. KCa3.1 was involved in TGF-ß-induced astrogliosis in vitro and also contributed to astrogliosis-mediated neuroinflammation in neurodegeneration disease. METHODS: Wild type mice and KCa3.1-/- mice were subjected to permanent middle cerebral artery occlusion (pMCAO) to evaluate the infarct areas by 2,3,5-triphenyltetrazolium hydrochloride staining and neurological deficit. KCa3.1 channels expression and cell localization in the brain of pMCAO mice model were measured by immunoblotting and immunostaining. Glia activation and neuron loss was measured by immunostaining. DiBAC4 (3) and Fluo-4AM were used to measure membrane potential and cytosolic Ca2+ level in oxygen-glucose deprivation induced reactive astrocytes in vitro. RESULTS: Immunohistochemistry on pMCAO mice infarcts showed strong upregulation of KCa3.1 immunoreactivity in reactive astrogliosis. KCa3.1-/- mice exhibited significantly smaller infarct areas on pMCAO and improved neurological deficit. Both activated gliosis and neuronal loss were attenuated in KCa3.1-/- pMCAO mice. In the primary cultured astrocytes, the expressions of KCa3.1 and TRPV4 were increased associated with upregulation of astrogliosis marker GFAP induced by oxygen-glucose deprivation. The activation of KCa3.1 hyperpolarized membrane potential and, by promoting the driving force for calcium, induced calcium entry through TRPV4, a cation channel of the transient receptor potential family. Double-labeled staining showed that KCa3.1 and TRPV4 channels co-localized in astrocytes. Blockade of KCa3.1 or TRPV4 inhibited the phenotype switch of reactive astrogliosis. CONCLUSIONS: Our data suggested that KCa3.1 inhibition might represent a promising therapeutic strategy for ischemia stroke.


Assuntos
Isquemia Encefálica/metabolismo , Sistemas de Liberação de Medicamentos , Gliose/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/deficiência , Bloqueadores dos Canais de Potássio/administração & dosagem , Acidente Vascular Cerebral/metabolismo , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Gliose/tratamento farmacológico , Gliose/patologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia
14.
Toxicol Appl Pharmacol ; 322: 89-96, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28284858

RESUMO

Excessive consumption of alcohol is a well-established risk factor of atrial fibrillation (AF). However, the effects of moderate alcohol drinking remain to be elucidated. This study was designed to determine the effects of moderate ethanol ingestion on atrial fibrillation and the electrophysiological mechanisms. In acetylcholine-induced canine and mouse AF models, the moderate ethanol prevented the generation and persistence of AF through prolonging the latent period of AF and shortening the duration of AF. The action potential duration (APD) was remarkably prolonged under the concentration range of 12.5-50.0mM ethanol in guinea pig atrial myocytes. Ultra-rapid delayed rectified potassium currents (IKv1.5) were markedly inhibited by 12.5-50.0mM ethanol in a concentration-dependent manner. Ethanol with 50.0mM could inhibit rapid delayed rectifier potassium currents (IhERG). Ethanol under 6.25-50.0mM did not affect on inward rectifier potassium currents (IKir2.1). Collectively, the present study provided an evidence that moderate ethanol intake can prolong the APD of atrial myocytes by inhibition of IKv1.5 and IhERG, which contributed to preventing the development and duration of AF.


Assuntos
Antiarrítmicos/administração & dosagem , Fibrilação Atrial/tratamento farmacológico , Etanol/administração & dosagem , Canal de Potássio Kv1.5/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/administração & dosagem , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Fibrilação Atrial/fisiopatologia , Cães , Relação Dose-Resposta a Droga , Cobaias , Células HEK293 , Humanos , Canal de Potássio Kv1.5/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Miócitos Cardíacos/fisiologia
15.
Immunity ; 29(4): 602-14, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18835197

RESUMO

Effector memory T (Tem) cells are essential mediators of autoimmune disease and delayed-type hypersensitivity (DTH), a convenient model for two-photon imaging of Tem cell participation in an inflammatory response. Shortly (3 hr) after entry into antigen-primed ear tissue, Tem cells stably attached to antigen-bearing antigen-presenting cells (APCs). After 24 hr, enlarged Tem cells were highly motile along collagen fibers and continued to migrate rapidly for 18 hr. Tem cells rely on voltage-gated Kv1.3 potassium channels to regulate calcium signaling. ShK-186, a specific Kv1.3 blocker, inhibited DTH and suppressed Tem cell enlargement and motility in inflamed tissue but had no effect on homing to or motility in lymph nodes of naive and central memory T (Tcm) cells. ShK-186 effectively treated disease in a rat model of multiple sclerosis. These results demonstrate a requirement for Kv1.3 channels in Tem cells during an inflammatory immune response in peripheral tissues. Targeting Kv1.3 allows for effector memory responses to be suppressed while central memory responses remain intact.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Hipersensibilidade Tardia/imunologia , Memória Imunológica , Canal de Potássio Kv1.3/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Movimento Celular/efeitos dos fármacos , Infecções por Chlamydia/tratamento farmacológico , Infecções por Chlamydia/imunologia , Chlamydia trachomatis/imunologia , Colágeno , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Feminino , Hipersensibilidade Tardia/metabolismo , Memória Imunológica/efeitos dos fármacos , Canal de Potássio Kv1.3/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Ativação Linfocitária/efeitos dos fármacos , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/imunologia , Ovalbumina/imunologia , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/uso terapêutico , Proteínas/farmacologia , Ratos , Ratos Endogâmicos Lew , Receptores CCR7/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
16.
J Electrocardiol ; 50(3): 277-281, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28238302

RESUMO

BACKGROUND: Patients with repetitive ventricular tachyarrhythmias - so-called electrical storm - frequently require antiarrhythmic drugs. Amiodarone is widely used for the treatment of electrical storm but is ineffective in some patients. Therefore, we investigated the efficacy of stepwise administration of nifekalant, a pure potassium channel blocker, and mexiletine for electrical storm. METHODS: This study included 44 patients with repetitive ventricular tachyarrhythmias who received stepwise therapy with nifekalant and mexiletine for electrical storm. Nifekalant was initially administered, and mexiletine was subsequently added if nifekalant failed to control ventricular tachyarrhythmias. RESULTS: Nifekalant completely suppressed recurrences of ventricular arrhythmias in 28 patients (64%), including 6 patients in whom oral amiodarone failed to control arrhythmias. In 9 of 16 patients in whom nifekalant was partially effective but failed to suppress ventricular arrhythmias, mexiletine was added. The addition of mexiletine prevented recurrences of ventricular tachyarrhythmias in 5 of these 9 patients (56%). There was no death associated with electrical storm. In total, the stepwise treatment with nifekalant and mexiletine was effective in preventing ventricular tachyarrhythmias in 33 of 44 patients (75%). There was no difference in cycle length of the ventricular tachycardia, QRS interval, QT interval, or left ventricular ejection fraction between patients who responded to antiarrhythmic drugs and those who did not. During follow-up, 8 patients had repetitive ventricular tachyarrhythmia recurrences, and the stepwise treatment was effective in 6 of these 8 patients (75%). CONCLUSIONS: The stepwise treatment with nifekalant and mexiletine was highly effective in the suppression of electrical storm.


Assuntos
Eletroencefalografia/efeitos dos fármacos , Mexiletina/administração & dosagem , Bloqueadores dos Canais de Potássio/administração & dosagem , Pirimidinonas/administração & dosagem , Bloqueadores dos Canais de Sódio/administração & dosagem , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/mortalidade , Administração Oral , Humanos , Japão/epidemiologia , Prevalência , Recidiva , Estudos Retrospectivos , Fatores de Risco , Taxa de Sobrevida , Taquicardia Ventricular/diagnóstico , Resultado do Tratamento
17.
J Neurosci ; 35(5): 2101-11, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653366

RESUMO

Cerebral ischemic stroke is a worldwide cause of mortality/morbidity and thus an important focus of research to decrease the severity of brain injury. Therapeutic options for acute stroke are still limited. In neurons throughout the brain, "M-type" K(+) currents, underlain by KCNQ subunits 2-5, play dominant roles in control over excitability, and are thus implicated in myriad neurological and psychiatric disorders. Although KCNQ channel openers, such as retigabine, have emerged as anti-epilepsy drugs, their effects on ischemic injury remain unknown. Here, we investigated the protective effects of M-channel openers on stroke-induced brain injury in mouse photothrombotic and middle cerebral artery occlusion (MCAo) models. Both photothrombosis and MCAo led to rapid, predictable, and consistently sized necrotic brain lesions, inflammatory responses, and behavioral deficits. Administration of three distinct M-channel openers at 0-6 h after ischemic injury significantly decreased brain infarct size and inflammation, and prevented neurological dysfunction, although they were more effective when administered 0-3 h poststroke. Thus, we show beneficial effects against stroke-induced brain injury and neuronal death through pharmacological regulation of ion channels that control neuronal excitability.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Canais de Potássio KCNQ/antagonistas & inibidores , Animais , Antracenos/administração & dosagem , Antracenos/farmacologia , Antracenos/uso terapêutico , Carbamatos/administração & dosagem , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Quimioterapia Combinada , Canais de Potássio KCNQ/agonistas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenilenodiaminas/administração & dosagem , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/uso terapêutico
18.
J Peripher Nerv Syst ; 21(2): 85-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26968589

RESUMO

Fampridine-PR is a voltage-gated potassium channel inhibitor potentially improving nerve conduction in demyelinated axons. Based on its established clinical efficacy in patients with demyelination in the central nervous system, we assessed if fampridine-PR is also effective in patients with inflammatory demyelination of the peripheral nerve. In this small open-label study, 10 patients with chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) were treated with fampridine-PR 10 mg BID for 28 days and assessed clinically as well as by nerve conduction studies. In this study, Fampridine-PR failed to improve CIDP based on clinical measures and nerve conduction studies. Our findings suggest that Fampridine-PR appears to be ineffective in demyelinating polyneuropathies. These observations may indicate a more complex mode of action beyond improving action potential conduction in demyelinated axons.


Assuntos
4-Aminopiridina/administração & dosagem , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/tratamento farmacológico , Bloqueadores dos Canais de Potássio/administração & dosagem , Resultado do Tratamento , Avaliação da Deficiência , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença
19.
Anesthesiology ; 122(3): 571-84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25321870

RESUMO

BACKGROUND: Propofol is widely used clinically for the induction and maintenance of anesthesia. Clinical case reports have shown that propofol has an antiatrial tachycardia/fibrillation effect; however, the related ionic mechanisms are not fully understood. The current study investigates the effects of propofol on human cardiac potassium channels. METHODS: The whole cell patch voltage clamp technique was used to record transient outward potassium current (Ito) and ultrarapidly activating delayed rectifier potassium current (IKur) in human atrial myocytes and hKv1.5, human ether-à-go-go-related gene (hERG), and hKCNQ1/hKCNE1 channels stably expressed in HEK 293 cells. Current clamp mode was used to record action potentials in human atrial myocytes. RESULTS: In human atrial myocytes, propofol inhibited Ito in a concentration-dependent manner (IC50 = 33.5 ± 2.0 µM for peak current, n = 6) by blocking open channels without affecting the voltage-dependent kinetics or the recovery time constant; propofol decreased IKur (IC50 = 35.3 ± 1.9 µM, n = 6) in human atrial myocytes and inhibited hKv1.5 current expressed in HEK 293 cells by preferentially binding to the open channels. Action potential duration at 90% repolarization was slightly prolonged by 30 µM propofol in human atrial myocytes. In addition, propofol also suppressed hERG and hKCNQ1/hKCNE1 channels expressed in HEK 293 cells. CONCLUSION: Propofol inhibits multiple human cardiac potassium channels, including human atrial Ito and IKur, as well as hKv1.5, hERG, and hKCNQ1/hKCNE1 channels stably expressed in HEK 293 cells, and slightly prolongs human atrial action potential duration, which may contribute to the antiatrial tachycardia/fibrillation effects observed in patients who receive propofol.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/administração & dosagem , Canais de Potássio/fisiologia , Propofol/administração & dosagem , Idoso , Anestésicos Intravenosos , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade
20.
Circ Res ; 113(8): 1023-32, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23940309

RESUMO

RATIONALE: Reactive hyperemia (RH) in the forearm circulation is an important marker of cardiovascular health, yet the underlying vasodilator signaling pathways are controversial and thus remain unclear. OBJECTIVE: We hypothesized that RH occurs via activation of inwardly rectifying potassium (KIR) channels and Na(+)/K(+)-ATPase and is largely independent of the combined production of the endothelial autocoids nitric oxide (NO) and prostaglandins in young healthy humans. METHODS AND RESULTS: In 24 (23±1 years) subjects, we performed RH trials by measuring forearm blood flow (FBF; venous occlusion plethysmography) after 5 minutes of arterial occlusion. In protocol 1, we studied 2 groups of 8 subjects and assessed RH in the following conditions. For group 1, we studied control (saline), KIR channel inhibition (BaCl2), combined inhibition of KIR channels and Na(+)/K(+)-ATPase (BaCl2 and ouabain, respectively), and combined inhibition of KIR channels, Na(+)/K(+)-ATPase, NO, and prostaglandins (BaCl2, ouabain, L-NMMA [N(G)-monomethyl-L-arginine] and ketorolac, respectively). Group 2 received ouabain rather than BaCl2 in the second trial. In protocol 2 (n=8), the following 3 RH trials were performed: control; L-NMMA plus ketorolac; and L-NMMA plus ketorolac plus BaCl2 plus ouabain. All infusions were intra-arterial (brachial). Compared with control, BaCl2 significantly reduced peak FBF (-50±6%; P<0.05), whereas ouabain and L-NMMA plus ketorolac did not. Total FBF (area under the curve) was attenuated by BaCl2 (-61±3%) and ouabain (-44±12%) alone, and this effect was enhanced when combined (-87±4%), nearly abolishing RH. L-NMMA plus ketorolac did not impact total RH FBF before or after administration of BaCl2 plus ouabain. CONCLUSIONS: Activation of KIR channels is the primary determinant of peak RH, whereas activation of both KIR channels and Na(+)/K(+)-ATPase explains nearly all of the total (AUC) RH in humans.


Assuntos
Artéria Braquial/enzimologia , Antebraço/irrigação sanguínea , Hemodinâmica , Hiperemia/enzimologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Adolescente , Adulto , Análise de Variância , Velocidade do Fluxo Sanguíneo , Artéria Braquial/efeitos dos fármacos , Artéria Braquial/fisiopatologia , Estudos de Casos e Controles , Inibidores de Ciclo-Oxigenase/administração & dosagem , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Hiperemia/fisiopatologia , Infusões Intra-Arteriais , Masculino , Microcirculação , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Pletismografia , Bloqueadores dos Canais de Potássio/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Prostaglandinas/metabolismo , Fluxo Sanguíneo Regional , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Fatores de Tempo , Vasodilatação , Vasodilatadores/administração & dosagem , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa