Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Physiol Biochem ; 55(S3): 46-64, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33667331

RESUMEN

BACKGROUND/AIMS: Tea, produced from the evergreen Camellia sinensis, has reported therapeutic properties against multiple pathologies, including hypertension. Although some studies validate the health benefits of tea, few have investigated the molecular mechanisms of action. The KCNQ5 voltage-gated potassium channel contributes to vascular smooth muscle tone and neuronal M-current regulation. METHODS: We applied electrophysiology, myography, mass spectrometry and in silico docking to determine effects and their underlying molecular mechanisms of tea and its components on KCNQ channels and arterial tone. RESULTS: A 1% green tea extract (GTE) hyperpolarized cells by augmenting KCNQ5 activity >20-fold at resting potential; similar effects of black tea were inhibited by milk. In contrast, GTE had lesser effects on KCNQ2/Q3 and inhibited KCNQ1/E1. Tea polyphenols epicatechin gallate (ECG) and epigallocatechin-3-gallate (EGCG), but not epicatechin or epigallocatechin, isoform-selectively hyperpolarized KCNQ5 activation voltage dependence. In silico docking and mutagenesis revealed that activation by ECG requires KCNQ5-R212, at the voltage sensor foot. Strikingly, ECG and EGCG but not epicatechin KCNQ-dependently relaxed rat mesenteric arteries. CONCLUSION: KCNQ5 activation contributes to vasodilation by tea; ECG and EGCG are candidates for future anti-hypertensive drug development.


Asunto(s)
Catequina/análogos & derivados , Canales de Potasio KCNQ/química , Canal de Potasio KCNQ1/química , Arterias Mesentéricas/efectos de los fármacos , Extractos Vegetales/farmacología , Té/química , Animales , Sitios de Unión , Catequina/química , Catequina/farmacología , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Arterias Mesentéricas/fisiología , Leche/química , Simulación del Acoplamiento Molecular , Miografía , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Extractos Vegetales/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Técnicas de Cultivo de Tejidos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Xenopus laevis
2.
Mol Cell ; 81(1): 25-37.e4, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33238160

RESUMEN

Among the five KCNQ channels, also known as the Kv7 voltage-gated potassium (Kv) channels, KCNQ2-KCNQ5 control neuronal excitability. Dysfunctions of KCNQ2-KCNQ5 are associated with neurological disorders such as epilepsy, deafness, and neuropathic pain. Here, we report the cryoelectron microscopy (cryo-EM) structures of human KCNQ4 and its complexes with the opener retigabine or the blocker linopirdine at overall resolutions of 2.5, 3.1, and 3.3 Å, respectively. In all structures, a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule inserts its head group into a cavity within each voltage-sensing domain (VSD), revealing an unobserved binding mode for PIP2. Retigabine nestles in each fenestration, inducing local shifts. Instead of staying within the central pore, linopirdine resides in a cytosolic cavity underneath the inner gate. Electrophysiological analyses of various mutants corroborated the structural observations. Our studies reveal the molecular basis for the modulatory mechanism of neuronal KCNQ channels and provide a framework for structure-facilitated drug discovery targeting these important channels.


Asunto(s)
Carbamatos/farmacología , Indoles/farmacología , Canales de Potasio KCNQ , Fenilendiaminas/farmacología , Piridinas/farmacología , Animales , Microscopía por Crioelectrón , Humanos , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Mutación , Fosfatidilinositol 4,5-Difosfato/metabolismo , Dominios Proteicos , Células Sf9 , Spodoptera
3.
Commun Biol ; 3(1): 356, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641720

RESUMEN

Many commonly consumed plants are used as folk medicines, often with unclear molecular mechanisms. Recent studies uncovered the ubiquitous and influential KCNQ family of voltage-gated potassium (Kv) channels as a therapeutic target for several medicinal plant compounds. Capers - immature flower buds of Capparis spinosa - have been consumed for food and medicinal purposes for millennia. Here, we show that caper extract hyperpolarizes cells expressing KCNQ1 or KCNQ2/3 Kv channels. Capers are the richest known natural source of quercetin, the most consumed dietary flavonoid. Quercetin potentiated KCNQ1/KCNE1, KCNQ2/3 and KCNQ4 currents but, unusually, not KCNQ5. Strikingly, quercetin augmented both activation and inactivation of KCNQ1, via a unique KCNQ activation mechanism involving sites atop the voltage sensor and in the pore. The findings uncover a novel potential molecular basis for therapeutic effects of quercetin-rich foods and a new chemical space for atypical modes of KCNQ channel modulation.


Asunto(s)
Canales de Potasio KCNQ/agonistas , Quercetina/farmacología , Animales , Sitios de Unión , Capparis/química , Canales de Potasio KCNQ/química , Canal de Potasio KCNQ2/agonistas , Canal de Potasio KCNQ3/agonistas , Oocitos , Técnicas de Placa-Clamp , Extractos Vegetales/farmacología , Estructura Terciaria de Proteína , Rutina/farmacología , Xenopus laevis
4.
Pharmacol Res ; 159: 105039, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32565313

RESUMEN

The gasotransmitter hydrogen sulfide (H2S) is involved in the regulation of the vascular tone and an impairment of its endogenous production may play a role in hypertension. Thus, the administration of exogenous H2S may be a possible novel and effective strategy to control blood pressure. Some natural and synthetic sulfur compounds are suitable H2S-donors, exhibiting long-lasting H2S release; however, novel H2S-releasing agents are needed to improve the pharmacological armamentarium for the treatment of cardiovascular diseases. For this purpose, N-phenylthiourea (PTU) and N,N'-diphenylthiourea (DPTU) compounds have been investigated as potential H2S-donors. The thioureas showed long-lasting H2S donation in cell free environment and in human aortic smooth muscle cells (HASMCs). In HASMCs, DPTU caused membrane hyperpolarization, mediated by activation of KATP and Kv7 potassium channels. The thiourea derivatives promoted vasodilation in rat aortic rings, which was abolished by KATP and Kv7 blockers. The vasorelaxing effects were also observed in angiotensin II-constricted coronary vessels. In conclusion, thiourea represents an original H2S-donor functional group, which releases H2S with slow and long lasting kinetic, and promotes typical H2S-mediated vascular effects. Such a moiety will be extremely useful for developing original cardiovascular drugs and new chemical tools for investigating the pharmacological roles of H2S.


Asunto(s)
Sulfuro de Hidrógeno/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Feniltiourea/farmacología , Tiourea/análogos & derivados , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Humanos , Preparación de Corazón Aislado , Canales KATP/agonistas , Canales KATP/metabolismo , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/metabolismo , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratas Wistar , Tiourea/farmacología
5.
Arterioscler Thromb Vasc Biol ; 40(5): 1207-1219, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32188278

RESUMEN

OBJECTIVE: Intravenous acetaminophen/paracetamol (APAP) is well documented to cause hypotension. Since the patients receiving intravenous APAP are usually critically ill, any severe hemodynamic changes, as with those associated with APAP, can be life-threatening. The mechanism underlying this dangerous iatrogenic effect of APAP was unknown. Approach and Results: Here, we show that intravenous APAP caused transient hypotension in rats, which was attenuated by the Kv7 channel blocker, linopirdine. APAP metabolite N-acetyl-p-benzoquinone imine caused vasodilatation of rat mesenteric arteries ex vivo. This vasodilatation was sensitive to linopirdine and also the calcitonin gene-related peptide antagonist, BIBN 4096. Further investigation revealed N-acetyl-p-benzoquinone imine stimulates calcitonin gene-related peptide release from perivascular nerves, causing a cAMP-dependent activation of Kv7 channels. We also show that N-acetyl-p-benzoquinone imine enhances Kv7.4 and Kv7.5 channels overexpressed in oocytes, suggesting that it can activate Kv7.4 and Kv7.5 channels directly, to elicit vasodilatation. CONCLUSIONS: Direct and indirect activation of Kv7 channels by the APAP metabolite N-acetyl-p-benzoquinone imine decreases arterial tone, which can lead to a drop in blood pressure. Our findings provide a molecular mechanism and potential preventive intervention for the clinical phenomenon of intravenous APAP-dependent transient hypotension.


Asunto(s)
Acetaminofén/toxicidad , Presión Sanguínea/efectos de los fármacos , Hipotensión/inducido químicamente , Canales de Potasio KCNQ/agonistas , Arterias Mesentéricas/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Acetaminofén/metabolismo , Animales , Benzoquinonas , Hipotensión/metabolismo , Hipotensión/fisiopatología , Iminas , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Masculino , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratas Wistar , Transducción de Señal , Xenopus laevis
6.
Pharmacology ; 105(7-8): 471-476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32062659

RESUMEN

INTRODUCTION: Gout arthritis is an inflammatory disease characterized by severe acute pain. The goal of pharmacological gout arthritis treatments is to reduce pain, and thereby increase the patient's quality of life. The Kv7/M channel activators retigabine and flupirtine show analgesic efficacy in animal models of osteoarthritic pain. We hypothesized that these drugs may also alleviate gout arthritis pain. OBJECTIVE: To determine the effects of retigabine and flupirtine on pain behavior associated with monosodium urate (MSU)-induced gout arthritis. METHODS: The gout arthritis model was established with an intra-articular injection of MSU into the right ankle joint, animals were treated with retigabine or flupirtine, and pain-related behaviors were assessed. RESULTS: Retigabine and flupirtine significantly increased the mechanical threshold and prolonged the paw withdrawal latency in a rat model of gout arthritis pain in a dose-dependent manner. The antinociceptive effects of retigabine and flupirtine were fully antagonized by the Kv7/M channel blocker XE991. CONCLUSION: Retigabine and flupirtine showed antinociceptive effects for MSU-induced gout pain at different times during pain development.


Asunto(s)
Aminopiridinas/farmacología , Analgésicos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Gotosa/tratamiento farmacológico , Carbamatos/farmacología , Dolor/tratamiento farmacológico , Fenilendiaminas/farmacología , Aminopiridinas/uso terapéutico , Analgésicos/uso terapéutico , Animales , Artritis Experimental/inducido químicamente , Artritis Gotosa/inducido químicamente , Conducta Animal/efectos de los fármacos , Carbamatos/uso terapéutico , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/efectos de los fármacos , Masculino , Dolor/inducido químicamente , Fenilendiaminas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Ácido Úrico/toxicidad
7.
J Pharmacol Exp Ther ; 373(1): 72-80, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31969383

RESUMEN

Spinal cord injury (SCI) usually leads to acute neuronal death and delayed secondary degeneration, resulting in sensory dysfunction, paralysis, and chronic pain. Excessive excitation is one of the critical factors leading to secondary neural damage initiated by various insults. KCNQ/Kv7 channels are highly expressed in spinal neurons and axons and play an important role in controlling their excitability. Enhancing KCNQ channel activity by using its specific opener retigabine could thus be a plausible treatment strategy to reduce the pathology after SCI. We produced contusive SCI at T10 in adult male rats, which then received 10 consecutive days' treatment with retigabine or vehicle starting 3 hours or 3 days after contusion. Two different concentrations and two different delivery methods were applied. Delivery of retigabine via Alzet osmotic pumps, but not intraperitoneal injections 3 hours after contusion, promoted recovery of locomotor function. Remarkably, retigabine delivery in both methods significantly attenuated the development of mechanical stimuli-induced hyperreflexia and spontaneous pain; however, no significant difference in the thermal threshold was observed. Although retigabine delivered 3 days after contusion significantly attenuated the development of mechanical hypersensitivity and spontaneous pain, the locomotor function is not improved by the delayed treatments. Finally, we found that early application of retigabine attenuates the inflammatory activity in the spinal cord and increases the survival of white matter after SCI. Our results suggest that decreasing neuronal excitability by targeting KCNQ/Kv7 channels at acute stage aids the recovery of locomotor function and attenuates the development of neuropathic pain after SCI. SIGNIFICANCE STATEMENT: Several pharmacological interventions have been proposed for spinal cord injury (SCI) treatment, but none have been shown to be both effective and safe in clinical trials. Necrotic neuronal death and chronic pain are often the cost of pathological neural excitation after SCI. We show that early, brief application of retigabine could aid locomotor and sensory neurobehavioral recovery after SCI, supporting the use of this drug in the clinic to promote motor and sensory function in patients with SCI.


Asunto(s)
Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/metabolismo , Locomoción/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Carbamatos/farmacología , Carbamatos/uso terapéutico , Locomoción/efectos de los fármacos , Masculino , Moduladores del Transporte de Membrana/farmacología , Moduladores del Transporte de Membrana/uso terapéutico , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Vértebras Torácicas/lesiones
8.
Mol Psychiatry ; 25(6): 1323-1333, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-30385872

RESUMEN

Major depressive disorder (MDD) is a leading cause of disability worldwide, yet current treatment strategies remain limited in their mechanistic diversity. Recent evidence has highlighted a promising novel pharmaceutical target-the KCNQ-type potassium channel-for the treatment of depressive disorders, which may exert a therapeutic effect via functional changes within the brain reward system, including the ventral striatum. The current study assessed the effects of the KCNQ channel opener ezogabine (also known as retigabine) on reward circuitry and clinical symptoms in patients with MDD. Eighteen medication-free individuals with MDD currently in a major depressive episode were enrolled in an open-label study and received ezogabine up to 900 mg/day orally over the course of 10 weeks. Resting-state functional magnetic resonance imaging data were collected at baseline and posttreatment to examine brain reward circuitry. Reward learning was measured using a computerized probabilistic reward task. After treatment with ezogabine, subjects exhibited a significant reduction of depressive symptoms (Montgomery-Asberg Depression Rating Scale score change: -13.7 ± 9.7, p < 0.001, d = 2.08) and anhedonic symptoms (Snaith-Hamilton Pleasure Scale score change: -6.1 ± 5.3, p < 0.001, d = 1.00), which remained significant even after controlling for overall depression severity. Improvement in depression was associated with decreased functional connectivity between the ventral caudate and clusters within the mid-cingulate cortex and posterior cingulate cortex (n = 14, voxel-wise p < 0.005). In addition, a subgroup of patients tested with a probabilistic reward task (n = 9) showed increased reward learning following treatment. These findings highlight the KCNQ-type potassium channel as a promising target for future drug discovery efforts in mood disorders.


Asunto(s)
Carbamatos/farmacología , Carbamatos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Activación del Canal Iónico/efectos de los fármacos , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Estriado Ventral/efectos de los fármacos , Trastorno Depresivo Mayor/metabolismo , Femenino , Humanos , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/metabolismo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recompensa , Estriado Ventral/metabolismo
9.
FASEB J ; 33(8): 9154-9166, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31063701

RESUMEN

Voltage-gated Kv7/KCNQ/M potassium channels play an essential role in the control of membrane potential and neuronal excitability. Activation of the neuronal Kv7/KCNQ/M-current represents an attractive therapeutic strategy for treatment of hyperexcitability-related neuropsychiatric disorders such as epilepsy, pain, and depression, which is an unmet medical need. In this study, we synthesized and characterized a novel compound, N-(4-(2-bromo-6,7-dihydrothieno[3,2-c]pyridin-5(4H)-yl)-2,6-dimethylphenyl)-3,3-dimethylbutanamide (SCR2682) 2,6-dimethyl-4-(piperidin-yl) phenyl)-amide derivative, that exhibits selective and potent activation of neuronal Kv7/KCNQ/M-channels. Whole-cell patch-clamp recordings of human embryonic kidney 293 cells expressing Kv7.2/Kv7.3 channels show that SCR2682 selectively activates the channel current in a dose-dependent manner with an EC50 of 9.8 ± 0.4 nM, which is ∼100-fold more potent than a U.S. Food and Drug Administration-approved antiepileptic drug (retigabine) for treatment of partial epilepsy. SCR2682 shifts voltage-dependent activation of the Kv7.2/7.3 current toward more negative membrane potential, to about -37 mV (V1/2). SCR2682 also activates the native M-current in rat hippocampal or cortical neurons, causing marked hyperpolarization and potent inhibition of neuronal firings. Mechanistically, mutating the tryptophan residue 236 located at the fifth transmembrane segment of Kv7.2 abolishes the chemical activation of the channel by SCR2682. Furthermore, intraperitoneal or intragastric administration of SCR2682 results in a dose-dependent inhibition of seizures by maximal electroshock. Taken together, our findings demonstrate that a novel small molecule, SCR2682, selectively and potently activates neuronal Kv7 channels and reverses epileptic seizures in rodents. Thus, SCR2682 may warrant further evaluation for clinical development of antiepileptic therapy.-Zhang, F., Liu, Y., Tang, F., Liang, B., Chen, H., Zhang, H., Wang, K. Electrophysiological and pharmacological characterization of a novel and potent neuronal Kv7 channel opener SCR2682 for antiepilepsy.


Asunto(s)
Anticonvulsivantes/farmacología , Canal de Potasio KCNQ2/agonistas , Canal de Potasio KCNQ3/agonistas , Moduladores del Transporte de Membrana/farmacología , Piridinas/farmacología , Sustitución de Aminoácidos , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/química , Células Cultivadas , Canal de Potasio ERG1/antagonistas & inhibidores , Epilepsia/tratamiento farmacológico , Células HEK293 , Humanos , Canales de Potasio KCNQ/agonistas , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Moduladores del Transporte de Membrana/síntesis química , Moduladores del Transporte de Membrana/química , Ratones , Mutagénesis Sitio-Dirigida , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Piridinas/síntesis química , Piridinas/química , Ratas , Convulsiones/tratamiento farmacológico
10.
J Pain ; 20(5): 528-539, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30471428

RESUMEN

Paclitaxel-induced peripheral neuropathy (PIPN) and associated neuropathic pain are the most common and serious adverse effects experienced by cancer patients receiving paclitaxel treatment. These effects adversely impact daily activities and consequently the quality of life, sometimes forcing the suspension of treatment and negatively influencing survival. Patients are usually at high risk of developing PIPN if paclitaxel induces acute pain, which strongly suggests that an acute increase in the excitability of nociceptors underlies the chronic alterations of PIPN. KCNQ/Kv7 channels are widely expressed in the primary sensory neurons to modulate their excitability. In the present study, we show that targeting KCNQ/Kv7 channels at an early stage is an effective strategy to attenuate the development of PIPN. We found that paclitaxel did not decrease the expression level of KCNQ/Kv7 channels in the primary sensory neurons as detected by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting. However, retigabine, which is a specific KCNQ/Kv7 channel opener, attenuated significantly the development of PIPN, as shown by both morphologic and behavioral evidence. We also observed that retigabine had no obvious effect on the chemosensitivity of breast cancer cells to paclitaxel. Although retigabine has been approved by the FDA as an anticonvulsant, our study suggests that this drug can be repurposed to attenuate the development of PIPN. PERSPECTIVE: Paclitaxel-induced peripheral neuropathy and associated neuropathic pain are severe and resistant to intervention. The results of our study demonstrated that retigabine (a clinically available medicine) can be used to attenuate the development of paclitaxel-induced peripheral neuropathy.


Asunto(s)
Carbamatos/farmacología , Canales de Potasio KCNQ/agonistas , Neuralgia/prevención & control , Fármacos Neuroprotectores/farmacología , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/prevención & control , Fenilendiaminas/farmacología , Animales , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Reposicionamiento de Medicamentos , Humanos , Canales de Potasio KCNQ/metabolismo , Masculino , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Paclitaxel/farmacología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley
11.
Epilepsia ; 59(2): 358-368, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29265344

RESUMEN

OBJECTIVES: Antiseizure drugs are the leading therapeutic choice for treatment of epilepsy, but their efficacy is limited by pharmacoresistance and the occurrence of unwanted side effects. Here, we examined the therapeutic efficacy of KCNQ channel activation by retigabine in preventing seizures and neurocardiac dysfunction in 2 potassium channelopathy mouse models of epilepsy with differing severity that have been associated with increased risk of sudden unexpected death in epilepsy (SUDEP): the Kcna1-/- model of severe epilepsy and the Kcnq1A340E/A340E model of mild epilepsy. METHODS: A combination of behavioral, seizure threshold, electrophysiologic, and gene expression analyses was used to determine the effects of KCNQ activation in mice. RESULTS: Behaviorally, Kcna1-/- mice exhibited unexpected hyperexcitability instead of the expected sedative-like response. In flurothyl-induced seizure tests, KCNQ activation decreased seizure latency by ≥50% in Kcnq1 strain mice but had no effect in the Kcna1 strain, suggesting the influence of genetic background. However, in simultaneous electroencephalography and electrocardiography recordings, KCNQ activation significantly reduced spontaneous seizure frequency in Kcna1-/- mice by ~60%. In Kcnq1A340E/A340E mice, KCNQ activation produced adverse cardiac effects including profound bradycardia and abnormal increases in heart rate variability and atrioventricular conduction blocks. Analyses of Kcnq2 and Kcnq3 mRNA levels revealed significantly elevated Kcnq2 expression in Kcna1-/- brains, suggesting that drug target alterations may contribute to the altered drug responses. SIGNIFICANCE: This study shows that treatment strategies in channelopathy may have unexpected outcomes and that effective rebalancing of channel defects requires improved understanding of channel interactions at the circuit and tissue levels. The efficacy of KCNQ channel activation and manifestation of adverse effects were greatly affected by genetic background, potentially limiting KCNQ modulation as a way to prevent neurocardiac dysfunction in epilepsy and thereby SUDEP risk. Our data also uncover a potential role for KCNQ2-5 channels in autonomic control of chronotropy.


Asunto(s)
Anticonvulsivantes/farmacología , Carbamatos/farmacología , Epilepsia/tratamiento farmacológico , Frecuencia Cardíaca/efectos de los fármacos , Canales de Potasio KCNQ/agonistas , Canal de Potasio KCNQ1/genética , Canal de Potasio Kv.1.1/genética , Fenilendiaminas/farmacología , Animales , Bloqueo Atrioventricular , Conducta Animal , Bradicardia , Canalopatías , Muerte Súbita , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Epilepsia Refractaria/tratamiento farmacológico , Epilepsia Refractaria/genética , Electroencefalografía , Epilepsia/genética , Perfilación de la Expresión Génica , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/genética , Ratones , Proteínas del Tejido Nervioso/genética , Farmacogenética , Pruebas de Farmacogenómica , ARN Mensajero/metabolismo , Transcriptoma
12.
J Neurophysiol ; 118(6): 2991-3006, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28855291

RESUMEN

The precise role and mechanisms underlying efferent modulation of peripheral vestibular afferent function are not well understood in mammals. Clarifying the details of efferent action may lead to new strategies for clinical management of debilitating disturbances in vestibular and balance function. Recent evidence in turtle indicates that efferent modulation of M-currents is likely one mechanism for modifying afferent discharge. M-currents depend in part on KCNQ potassium conductances (Kv7), which can be adjusted through efferent activation of M1, M3, and/or M5 muscarinic acetylcholine receptors (mAChRs). How KCNQ channels and altered M-currents affect vestibular afferent function in vivo is unclear, and whether such a mechanism operates in mammals is unknown. In this study we used the KCNQ antagonist XE991 and the KCNQ activator retigabine in anesthetized mice to evaluate the effects of M-current modulation on peripheral vestibular responses to transient head motion. At low doses of XE991, responses were modestly enhanced, becoming larger in amplitude and shorter in latency. Higher doses of XE991 produced transient response enhancement, followed by steady-state suppression where latencies and thresholds increased and amplitudes decreased. Retigabine produced opposite effects. Auditory function was also impacted, based on results of companion auditory brain stem response testing. We propose that closure of KCNQ channels transforms vestibular afferent behavior by suppressing responses to transient high-frequency stimuli while simultaneously enhancing responses to sustained low-frequency stimulation. Our results clearly demonstrate that KCNQ channels are critical for normal mammalian vestibular function and suggest that efferent action may utilize these mechanisms to modulate the dynamic characteristics and gain of vestibular afferent responses.NEW & NOTEWORTHY The role of calyceal KCNQ channels and associated M-current in normal mammalian vestibular function is unknown. Our results show that calyceal KCNQ channels are critical for normal vestibular function in the intact mammal. The findings provide evidence that efferent modulation of M-currents may act normally to differentially adjust the sensitivity of vestibular neurons to transient and tonic stimulation and that such mechanisms may be targeted to achieve effective clinical management of vestibular disorders.


Asunto(s)
Movimientos de la Cabeza , Neuronas Motoras/fisiología , Vestíbulo del Laberinto/fisiología , Animales , Antracenos/farmacología , Carbamatos/farmacología , Potenciales Evocados , Femenino , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/metabolismo , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/metabolismo , Fenilendiaminas/farmacología
13.
Basic Clin Pharmacol Toxicol ; 120(1): 46-51, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27377794

RESUMEN

Central Kv7 (KCNQ) channels are voltage-dependent potassium channels composed of different combinations of four Kv7 subunits, being differently expressed in the brain. Notably, striatal dopaminergic neurotransmission is strongly suppressed by systemic administration of the pan-Kv7 channel opener retigabine. The effect of retigabine likely involves the inhibition of the activity in mesencephalic dopaminergic neurons projecting to the striatum, but whether Kv7 channels expressed in the striatum may also play a role is not resolved. We therefore assessed the effect of intrastriatal retigabine administration on striatal neuronal excitability in the rat determined by c-Fos immunoreactivity, a marker of neuronal activation. When retigabine was applied locally in the striatum, this resulted in a marked reduction in the number of c-Fos-positive neurons after a strong excitatory striatal stimulus induced by acute systemic haloperidol administration in the rat. The relative mRNA levels of Kv7 subunits in the rat striatum were found to be Kv7.2 = Kv7.3 = Kv7.5 > >Kv7.4. These data suggest that intrastriatal Kv7 channels play a direct role in regulating striatal excitability in vivo.


Asunto(s)
Carbamatos/farmacología , Cuerpo Estriado/efectos de los fármacos , Canales de Potasio KCNQ/agonistas , Moduladores del Transporte de Membrana/farmacología , Neuronas Aferentes/efectos de los fármacos , Neuronas Eferentes/efectos de los fármacos , Fenilendiaminas/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/farmacología , Biomarcadores/metabolismo , Carbamatos/administración & dosificación , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Excitabilidad Cortical/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/farmacología , Inyecciones Intraventriculares , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Masculino , Moduladores del Transporte de Membrana/administración & dosificación , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Neuronas Eferentes/citología , Neuronas Eferentes/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Fenilendiaminas/administración & dosificación , Subunidades de Proteína/agonistas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Wistar
14.
Arterioscler Thromb Vasc Biol ; 36(12): 2404-2411, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27789473

RESUMEN

OBJECTIVE: To establish the role of Kv7 channels in EPAC (exchange protein directly activated by cAMP)-dependent relaxations of the rat vasculature and to investigate whether this contributes to ß-adrenoceptor-mediated vasorelaxations. APPROACH AND RESULTS: Isolated rat renal and mesenteric arteries (RA and MA, respectively) were used for isometric tension recording to study the relaxant effects of a specific EPAC activator and the ß-adrenoceptor agonist isoproterenol in the presence of potassium channel inhibitors and cell signaling modulators. Isolated myocytes were used in proximity ligation assay studies to detect localization of signaling intermediaries with Kv7.4 before and after cell stimulation. Our studies showed that the EPAC activator (8-pCPT-2Me-cAMP-AM) produced relaxations and enhanced currents of MA and RA that were sensitive to linopirdine (Kv7 inhibitor). Linopirdine also inhibited isoproterenol-mediated relaxations in both RA and MA. In the MA, isoproterenol relaxations were sensitive to EPAC inhibition, but not protein kinase A inhibition. In contrast, isoproterenol relaxations in RA were attenuated by protein kinase A but not by EPAC inhibition. Proximity ligation assay showed a localization of Kv7.4 with A-kinase anchoring protein in both vessels in the basal state, which increased only in the RA with isoproterenol stimulation. In the MA, but not the RA, a localization of Kv7.4 with both Rap1a and Rap2 (downstream of EPAC) increased with isoproterenol stimulation. CONCLUSIONS: EPAC-dependent vasorelaxations occur in part via activation of Kv7 channels. This contributes to the isoproterenol-mediated relaxation in mesenteric, but not renal, arteries.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Canales de Potasio KCNQ/metabolismo , Arterias Mesentéricas/metabolismo , Arteria Renal/metabolismo , Vasodilatación , Proteínas de Anclaje a la Quinasa A/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Factores de Intercambio de Guanina Nucleótido/agonistas , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Células HEK293 , Humanos , Técnicas In Vitro , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/genética , Masculino , Potenciales de la Membrana , Arterias Mesentéricas/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Wistar , Arteria Renal/efectos de los fármacos , Transducción de Señal , Transfección , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Proteínas de Unión al GTP rap1/metabolismo
15.
J Neurophysiol ; 116(5): 2114-2124, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27512022

RESUMEN

KCNQ/Kv7 channels form a slow noninactivating K+ current, also known as the M current. They activate in the subthreshold range of membrane potentials and regulate different aspects of excitability in neurons of the central nervous system. In spinal motoneurons (MNs), KCNQ/Kv7 channels have been identified in the somata, axonal initial segment, and nodes of Ranvier, where they generate a slow, noninactivating, K+ current sensitive to both muscarinic receptor-mediated inhibition and KCNQ/Kv7 channel blockers. In this study, we thoroughly reevaluated the function of up- and downregulation of KCNQ/Kv7 channels in mouse immature spinal MNs. Using electrophysiological techniques together with specific pharmacological modulators of the activity of KCNQ/Kv7 channels, we show that enhancement of the activity of these channels decreases the excitability of spinal MNs in mouse neonates. This action on MNs results from a combination of hyperpolarization of the resting membrane potential, a decrease in the input resistance, and depolarization of the voltage threshold. On the other hand, the effect of inhibition of KCNQ/Kv7 channels suggested that these channels play a limited role in regulating basal excitability. Computer simulations confirmed that pharmacological enhancement of KCNQ/Kv7 channel activity decreases excitability and also suggested that the effects of inhibition of KCNQ/Kv7 channels on the excitability of spinal MNs do not depend on a direct effect in these neurons but likely on spinal cord synaptic partners. These results indicate that KCNQ/Kv7 channels have a fundamental role in the modulation of the excitability of spinal MNs acting both in these neurons and in their local presynaptic partners.


Asunto(s)
Regulación hacia Abajo/fisiología , Canales de Potasio KCNQ/fisiología , Neuronas Motoras/fisiología , Médula Espinal/fisiología , Regulación hacia Arriba/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Regulación hacia Abajo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Ratones , Neuronas Motoras/efectos de los fármacos , Técnicas de Cultivo de Órganos , Médula Espinal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
16.
Neuropharmacology ; 109: 131-138, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27263036

RESUMEN

The activation of Kv7 channels and the resulting M-current is a powerful mechanism to control neuronal excitability with profound effects in pain pathways. Despite the lack of specific data on the expression and role of these channels in nociceptive processing, much attention has been paid at exploring their potential value as targets for analgesia. Here we have characterized the spinal actions of two novel subunit selective Kv7 activators, ICA-069673 and ML213, and compared their effects to those of retigabine that acts with similar affinity on all neuronal Kv7 channels. Spinal reflexes were recorded in a mouse spinal cord in vitro preparation to allow the testing of the compounds on native spinal pathways at known concentrations. As retigabine, novel compounds depressed spinal segmental transmission with particularly strong effects on wind up, showing an adequate pro-analgesic profile. ML213 presented the highest potency. In contrast to retigabine, the effects of ICA-069673 and ML213 were blocked by XE-991 even at the highest concentrations used, suggesting specific effect on Kv7 channels. In addition, the effects of ICA-069673 on repetitive stimulation are consistent with a mode of action involving state or activity dependent interaction with the channels. Compared to retigabine, novel Kv7 openers maintain strong depressant effects on spinal nociceptive transmission showing an improved specificity on Kv7 channels. The differential effects obtained with these Kv7 openers may indicate the existence of several Kv7 conformations in spinal circuits.


Asunto(s)
Anticonvulsivantes/farmacología , Carbamatos/farmacología , Canales de Potasio KCNQ/fisiología , Fenilendiaminas/farmacología , Reflejo/fisiología , Médula Espinal/fisiología , Anilidas/farmacología , Animales , Compuestos Bicíclicos con Puentes/farmacología , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Masculino , Ratones , Reflejo/efectos de los fármacos , Médula Espinal/efectos de los fármacos
17.
Acta Pharmacol Sin ; 37(8): 1054-62, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27264315

RESUMEN

AIM: The aim of this study was to examine the activation of neuronal Kv7/KCNQ channels by a novel modified Kv7 opener QO58-lysine and to test the anti-nociceptive effects of QO58-lysine on inflammatory pain in rodent models. METHODS: Assays including whole-cell patch clamp recordings, HPLC, and in vivo pain behavioral evaluations were employed. RESULTS: QO58-lysine caused instant activation of Kv7.2/7.3 currents, and increasing the dose of QO58-lysine resulted in a dose-dependent activation of Kv7.2/Kv7.3 currents with an EC50 of 1.2±0.2 µmol/L. QO58-lysine caused a leftward shift of the voltage-dependent activation of Kv7.2/Kv7.3 to a hyperpolarized potential at V1/2=-54.4±2.5 mV from V1/2=-26.0±0.6 mV. The half-life in plasma (t1/2) was derived as 2.9, 2.7, and 3.0 h for doses of 12.5, 25, and 50 mg/kg, respectively. The absolute bioavailabilities for the three doses (12.5, 25, and 50 mg/kg) of QO58-lysine (po) were determined as 13.7%, 24.3%, and 39.3%, respectively. QO58-lysine caused a concentration-dependent reduction in the licking times during phase II pain induced by the injection of formalin into the mouse hindpaw. In the Complete Freund's adjuvant (CFA)-induced inflammatory pain model in rats, oral or intraperitoneal administration of QO58-lysine resulted in a dose-dependent increase in the paw withdrawal threshold, and the anti-nociceptive effect on mechanical allodynia could be reversed by the channel-specific blocker XE991 (3 mg/kg). CONCLUSION: Taken together, our findings show that a modified QO58 compound (QO58-lysine) can specifically activate Kv7.2/7.3/M-channels. Oral or intraperitoneal administration of QO58-lysine, which has improved bioavailability and a half-life of approximately 3 h in plasma, can reverse inflammatory pain in rodent animal models.


Asunto(s)
Canales de Potasio KCNQ/agonistas , Lisina/farmacología , Dimensión del Dolor/efectos de los fármacos , Animales , Antracenos/farmacología , Disponibilidad Biológica , Carbamatos/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lisina/antagonistas & inhibidores , Lisina/farmacocinética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Fenilendiaminas/farmacología , Ratas
18.
Pharmacol Ther ; 165: 14-25, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27179745

RESUMEN

Smooth muscle cells provide crucial contractile functions in visceral, vascular, and lung tissues. The contractile state of smooth muscle is largely determined by their electrical excitability, which is in turn influenced by the activity of potassium channels. The activity of potassium channels sustains smooth muscle cell membrane hyperpolarization, reducing cellular excitability and thereby promoting smooth muscle relaxation. Research over the past decade has indicated an important role for Kv7 (KCNQ) voltage-gated potassium channels in the regulation of the excitability of smooth muscle cells. Expression of multiple Kv7 channel subtypes has been demonstrated in smooth muscle cells from viscera (gastrointestinal, bladder, myometrial), from the systemic and pulmonary vasculature, and from the airways of the lung, from multiple species, including humans. A number of clinically used drugs, some of which were developed to target Kv7 channels in other tissues, have been found to exert robust effects on smooth muscle Kv7 channels. Functional studies have indicated that Kv7 channel activators and inhibitors have the ability to relax and contact smooth muscle preparations, respectively, suggesting a wide range of novel applications for the pharmacological tool set. This review summarizes recent findings regarding the physiological functions of Kv7 channels in smooth muscle, and highlights potential therapeutic applications based on pharmacological targeting of smooth muscle Kv7 channels throughout the body.


Asunto(s)
Diseño de Fármacos , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Bloqueadores de los Canales de Potasio/uso terapéutico , Sistema Respiratorio/efectos de los fármacos , Vísceras/efectos de los fármacos , Animales , Broncoconstricción/efectos de los fármacos , Broncoconstrictores/uso terapéutico , Broncodilatadores/uso terapéutico , Humanos , Canales de Potasio KCNQ/metabolismo , Terapia Molecular Dirigida , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Bloqueadores de los Canales de Potasio/efectos adversos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/fisiopatología , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Vísceras/metabolismo , Vísceras/fisiopatología
19.
J Pharm Pharmacol ; 68(4): 494-502, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26969140

RESUMEN

OBJECTIVES: This study investigated the effect of tannic acid (TA), a plant-derived hydrolyzable polyphenol, on Kv7.4 and Kv7.5 K(+) channels and rat mesenteric artery. METHODS: Whole-cell patch clamp experiments were used to record the Kv7.4 and Kv7.3/7.5 K(+) currents expressed in HEK293 cells; and the tension changes of mesenteric arteries isolated from rats were recorded using small vessel myography apparatus. KEY FINDINGS: Tannic acid increases the Kv7.4 and Kv7.3/7.5 K(+) currents in a concentration-dependent manner (median effective concentration (EC50 ) = 27.3 ± 3.6 µm and EC50 = 23.1 ± 3.9 µm, respectively). In addition, 30 µm TA shifts the G-V curve of Kv7.4 and Kv7.3/7.5 K(+) currents to the left by 14.18 and 25.24 mV, respectively, and prolongs the deactivation time constants by 184.44 and 154.77 ms, respectively. Moreover, TA relaxes the vascular tension of rat mesenteric arteries in a concentration-dependent manner (half inhibitory concentration (IC50 ) = 148.7 ± 13.4 µm). CONCLUSION: These results confirms the vasodilatory effects of TA on rat mesenteric artery and the activating effects on the Kv7.4 and Kv7.3/7.5 K(+) channels, which may be a mechanism to explain the vasodilatory effect and this mechanism can be used in the research of antihypertension.


Asunto(s)
Antihipertensivos/farmacología , Canales de Potasio KCNQ/agonistas , Canal de Potasio KCNQ3/agonistas , Arterias Mesentéricas/efectos de los fármacos , Taninos/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Técnicas In Vitro , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Masculino , Potenciales de la Membrana , Miografía , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Ratas Sprague-Dawley , Factores de Tiempo , Transfección
20.
J Pharmacol Exp Ther ; 357(1): 94-102, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26869667

RESUMEN

The voltage-gated KV7 (KCNQ) potassium channels are activated by ischemia and involved in hypoxic vasodilatation. We investigated the effect of KV7 channel modulation on cardiac ischemia and reperfusion injury and its interaction with cardioprotection by ischemic preconditioning (IPC). Reverse-transcription polymerase chain reaction revealed expression of KV7.1, KV7.4, and KV7.5 in the left anterior descending rat coronary artery and all KV7 subtypes (KV7.1-KV7.5) in the left and right ventricles of the heart. Isolated hearts were subjected to no-flow global ischemia and reperfusion with and without IPC. Infarct size was quantified by 2,3,5-triphenyltetrazolium chloride staining. Two blockers of KV7 channels, XE991 [10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone] (10 µM) and linopirdine (10 µM), reduced infarct size and exerted additive infarct reduction to IPC. An opener of KV7 channels, flupirtine (10 µM) abolished infarct size reduction by IPC. Hemodynamics were measured using a catheter inserted in the left ventricle and postischemic left ventricular recovery improved in accordance with reduction of infarct size and deteriorated with increased infarct size. XE991 (10 µM) reduced coronary flow in the reperfusion phase and inhibited vasodilatation in isolated small branches of the left anterior descending coronary artery during both simulated ischemia and reoxygenation. KV7 channels are expressed in rat coronary arteries and myocardium. Inhibition of KV7 channels exerts cardioprotection and opening of KV7 channels abrogates cardioprotection by IPC. Although safety issues should be further addressed, our findings suggest a potential role for KV7 blockers in the treatment of ischemia-reperfusion injury.


Asunto(s)
Canales de Potasio KCNQ/antagonistas & inhibidores , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Bloqueadores de los Canales de Potasio/farmacología , Aminopiridinas/uso terapéutico , Animales , Antracenos/uso terapéutico , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Indoles/uso terapéutico , Precondicionamiento Isquémico Miocárdico , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/genética , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Miocardio/metabolismo , Piridinas/uso terapéutico , Ratas , Ratas Wistar , Vasodilatación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA