Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Life Sci Alliance ; 7(10)2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39089879

RESUMEN

The large-conductance calcium-activated potassium (BKCa) channel, which is crucial for urinary bladder smooth muscle relaxation, is a potential target for overactive bladder treatment. Our prior work unveiled CTIBD as a promising BKCa channel activator, altering V 1/2 and G max This study investigates CTIBD's activation mechanism, revealing its independence from the Ca2+ and membrane voltage sensing of the BKCa channel. Cryo-electron microscopy disclosed that two CTIBD molecules bind to hydrophobic regions on the extracellular side of the lipid bilayer. Key residues (W22, W203, and F266) are important for CTIBD binding, and their replacement with alanine reduces CTIBD-mediated channel activation. The triple-mutant (W22A/W203A/F266A) channel showed the smallest V 1/2 shift with a minimal impact on activation and deactivation kinetics by CTIBD. At the single-channel level, CTIBD treatment was much less effective at increasing P o in the triple mutant, mainly because of a drastically increased dissociation rate compared with the WT. These findings highlight CTIBD's mechanism, offering crucial insights for developing small-molecule treatments for BKCa-related pathophysiological conditions.


Asunto(s)
Agonistas de los Canales de Cloruro , Microscopía por Crioelectrón , Canales de Potasio de Gran Conductancia Activados por el Calcio , Animales , Humanos , Sitios de Unión , Calcio/metabolismo , Células HEK293 , Activación del Canal Iónico , Cinética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/agonistas , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Membrana Dobles de Lípidos/metabolismo , Mutación , Unión Proteica , Agonistas de los Canales de Cloruro/química , Agonistas de los Canales de Cloruro/farmacología
2.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36835507

RESUMEN

Elevated TNF-α levels in serum and broncho-alveolar lavage fluid of acute lung injury patients correlate with mortality rates. We hypothesized that pharmacological plasma membrane potential (Em) hyperpolarization protects against TNF-α-induced CCL-2 and IL-6 secretion from human pulmonary endothelial cells through inhibition of inflammatory Ca2+-dependent MAPK pathways. Since the role of Ca2+ influx in TNF-α-mediated inflammation remains poorly understood, we explored the role of L-type voltage-gated Ca2+ (CaV) channels in TNF-α-induced CCL-2 and IL-6 secretion from human pulmonary endothelial cells. The CaV channel blocker, Nifedipine, decreased both CCL-2 and IL-6 secretion, suggesting that a fraction of CaV channels is open at the significantly depolarized resting Em of human microvascular pulmonary endothelial cells (-6 ± 1.9 mV), as shown by whole-cell patch-clamp measurements. To further explore the role of CaV channels in cytokine secretion, we demonstrated that the beneficial effects of Nifedipine could also be achieved by Em hyperpolarization via the pharmacological activation of large conductance K+ (BK) channels with NS1619, which elicited a similar decrease in CCL-2 but not IL-6 secretion. Using functional gene enrichment analysis tools, we predicted and validated that known Ca2+-dependent kinases, JNK-1/2 and p38, are the most likely pathways to mediate the decrease in CCL-2 secretion.


Asunto(s)
Células Epiteliales Alveolares , Quimiocina CCL2 , Canales de Potasio de Gran Conductancia Activados por el Calcio , Neumonía , Factor de Necrosis Tumoral alfa , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Nifedipino/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Neumonía/metabolismo , Neumonía/prevención & control , Quimiocina CCL2/metabolismo
3.
Neurol India ; 70(4): 1601-1609, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36076665

RESUMEN

Background: Neuroprotection in traumatic brain injury (TBI) is an unmet medical need. Objective: We evaluated two agents, aglepristone (progesterone receptor antagonist) and N-salicyloyltryptamine (STP) (activator of Maxi-K channel in GH3 cells), for neuroprotection in Feeney's weight drop model of TBI. Material and Methods: Forty-eight male Wistar rats were divided into six groups (n = 8 per group). A battery of six neurobehavioral tests was evaluated at the end of the first week (EO1W), second week (EO2W), and third week (EO3W). In addition, histopathological and immunohistochemistry (BAX, Bcl-2, and M30 Cytodeath) tests were performed at EO3W. Results: Aglepristone at 10 mg/kg showed significant neuroprotection compared to control as assessed by Rota-rod test at EO1W, VEFP right paw and 28-point neurobehavioral test at EO2W, MWM test at EO3W, and positive histopathological and IHC findings. Aglepristone at 20 mg/kg showed negative results as assessed by BAX expression, downregulation of Bcl-2, and positive M30 Cytodeath, thereby suggesting toxicity at higher doses. STP 100 mg/kg showed modest neuroprotective activity but failed to show a dose-response relationship at a dose of 50 mg/kg. Conclusion: The study shows that progesterone receptor antagonists have neuroprotection at lower doses and toxicity at higher doses.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Canales de Potasio de Gran Conductancia Activados por el Calcio , Fármacos Neuroprotectores , Receptores de Progesterona , Animales , Lesiones Encefálicas/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Proteínas Portadoras , Modelos Animales de Enfermedad , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Masculino , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Progesterona/antagonistas & inhibidores , Proteína X Asociada a bcl-2/metabolismo
4.
Cardiovasc Drugs Ther ; 35(4): 719-732, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33245463

RESUMEN

PURPOSE: In the present study, the therapeutic efficacy of a selective BKCa channel opener (compound X) in the treatment of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) was investigated. METHODS: PAH was induced in male Wistar rats by a single injection of MCT. After two weeks, the MCT-treated group was divided into two groups that were either treated with compound X or vehicle. Compound X was administered daily at 28 mg/kg. Electrocardiographic, echocardiographic, and haemodynamic analyses were performed; ex vivo evaluations of pulmonary artery reactivity, right ventricle (RV) and lung histology as well as expression levels of α and ß myosin heavy chain, brain natriuretic peptide, and cytokines (TNFα and IL10) in heart tissue were performed. RESULTS: Pulmonary artery rings of the PAH group showed a lower vasodilatation response to acetylcholine, suggesting endothelial dysfunction. Compound X promoted strong vasodilation in pulmonary artery rings of both control and MCT-induced PAH rats. The untreated hypertensive rats presented remodelling of pulmonary arterioles associated with increased resistance to pulmonary flow; increased systolic pressure, hypertrophy and fibrosis of the RV; prolongation of the QT and Tpeak-Tend intervals (evaluated during electrocardiogram); increased lung and liver weights; and autonomic imbalance with predominance of sympathetic activity. On the other hand, treatment with compound X reduced pulmonary vascular remodelling, pulmonary flow resistance and RV hypertrophy and afterload. CONCLUSION: The use of a selective and potent opener to activate the BKCa channels promoted improvement of haemodynamic parameters and consequent prevention of RV maladaptive remodelling in rats with MCT-induced PAH.


Asunto(s)
Agonistas de los Canales de Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio , Hipertensión Arterial Pulmonar , Quinolinas/farmacología , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Agonistas de los Canales de Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacocinética , Modelos Animales de Enfermedad , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/fisiopatología , Ratas , Ratas Wistar , Resultado del Tratamiento , Remodelación Vascular/efectos de los fármacos , Función Ventricular Derecha/efectos de los fármacos
5.
Pflugers Arch ; 472(10): 1481-1494, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32748018

RESUMEN

In myotonia, reduced Cl- conductance of the mutated ClC-1 channels causes hindered muscle relaxation after forceful voluntary contraction due to muscle membrane hyperexcitability. Repetitive contraction temporarily decreases myotonia, a phenomena called "warm up." The underlying mechanism for the reduction of hyperexcitability in warm-up is currently unknown. Since potassium displacement is known to reduce excitability in, for example, muscle fatigue, we characterized the role of potassium in native myotonia congenita (MC) muscle. Muscle specimens of ADR mice (an animal model for low gCl- conductance myotonia) were exposed to increasing K+ concentrations. To characterize functional effects of potassium ion current, the muscle of ADR mice was exposed to agonists and antagonists of the big conductance Ca2+-activated K+ channel (BK) and the voltage-gated Kv7 channel. Effects were monitored by functional force and membrane potential measurements. By increasing [K+]0 to 5 mM, the warm-up phenomena started earlier and at [K+]0 7 mM only weak myotonia was detected. The increase of [K+]0 caused a sustained membrane depolarization accompanied with a reduction of myotonic bursts in ADR mice. Retigabine, a Kv7.2-Kv7.5 activator, dose-dependently reduced relaxation deficit of ADR myotonic muscle contraction and promoted the warm-up phenomena. In vitro results of this study suggest that increasing potassium conductivity via activation of voltage-gated potassium channels enhanced the warm-up phenomena, thereby offering a potential therapeutic treatment option for myotonia congenita.


Asunto(s)
Canales de Cloruro/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Miotonía Congénita/metabolismo , Potasio/metabolismo , Animales , Cloruros/metabolismo , Canales de Potasio KCNQ/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Potenciales de la Membrana , Ratones , Contracción Muscular , Mutación , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología
6.
ACS Chem Biol ; 15(8): 2098-2106, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32667185

RESUMEN

Heme catabolism by heme oxygenase (HO) with a decrease in intracellular heme concentration and a concomitant local release of CO and Fe2+ has the potential to regulate BKCa channels. Here, we show that the iron-based photolabile CO-releasing molecule CORM-S1 [dicarbonyl-bis(cysteamine)iron(II)] coreleases CO and Fe2+, making it a suitable light-triggered source of these downstream products of HO activity. To investigate the impact of CO, iron, and cysteamine on BKCa channel activation, human Slo1 (hSlo1) was expressed in HEK293T cells and studied with electrophysiological methods. Whereas hSlo1 channels are activated by CO and even more strongly by Fe2+, Fe3+ and cysteamine possess only marginal activating potency. Investigation of hSlo1 mutants revealed that Fe2+ modulates the channels mainly through the Mg2+-dependent activation mechanism. Flash photolysis of CORM-S1 suits for rapid and precise delivery of Fe2+ and CO in biological settings.


Asunto(s)
Monóxido de Carbono/metabolismo , Compuestos Ferrosos/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Fotólisis , Sitios de Unión , Calcio/metabolismo , Compuestos Ferrosos/metabolismo , Células HEK293 , Hemo/metabolismo , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Magnesio/metabolismo , Técnicas de Placa-Clamp
7.
Bioorg Med Chem ; 28(16): 115609, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32690264

RESUMEN

As a member of transient receptor potential family, the transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium-permeable cation channel, which belongs to non-voltage gated Ca2+ channel. Large-conductance Ca2+-activated K+ channel (BKCa) represents a unique superfamily of Ca2+-activated K+ channel (KCa) that is both voltage and intracellular Ca2+ dependent. Not surprisingly, aberrant function of either TRPV4 or BKCa in neurons has been associated with brain disorders, such as Alzheimer's disease, cerebral ischemia, brain tumor, epilepsy, as well as headache. In these diseases, vascular dysfunction is a common characteristic. Notably, endothelial and smooth muscle TRPV4 can mediate BKCa to regulate cerebral blood flow and pressure. Therefore, in this review, we not only discuss the diverse functions of TRPV4 and BKCa in neurons to integrate relative signaling pathways in the context of cerebral physiological and pathological situations respectively, but also reveal the relationship between TRPV4 and BKCa in regulation of cerebral vascular tone as an etiologic factor. Based on these analyses, this review demonstrates the effective mechanisms of compounds targeting these two channels, which may be potential therapeutic strategies for diseases in the brain.


Asunto(s)
Encefalopatías/tratamiento farmacológico , Descubrimiento de Drogas , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Encefalopatías/metabolismo , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Terapia Molecular Dirigida , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores
8.
Arch Biochem Biophys ; 688: 108410, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32446891

RESUMEN

Kidneys from deceased donors used for transplantation are placed in cold storage (CS) solution during the search for a matched recipient. However, CS induces mitochondrial and cellular injury, which exacerbates renal graft dysfunction, highlighting the need for therapeutic interventions. Using an in vitro model of renal CS, we recently reported that pharmacological activation of the mitochondrial BK channel (mitoBK) during CS protected against CS-induced mitochondrial injury and cell death. Here, we used an in vivo syngeneic rat model of renal CS (18 h) followed by transplantation (24 h reperfusion) (CS + Tx) to similarly evaluate whether addition of a mitoBK activator to the CS solution can alleviate CS + Tx-induced renal injury. Western blots detected the pore-forming α subunit of the BK channel in mitochondrial fractions from rat kidneys, and mitoBK protein level was reduced after CS + Tx compared to sham surgery. The addition of the BK activator NS11021 (3 µM) to the CS solution partially protected against CS + Tx-induced mitochondrial respiratory dysfunction, oxidative protein nitration, and cell death, but not acute renal dysfunction (SCr and BUN). In summary, the current preclinical study shows that pharmacologically targeting mitoBK channels during CS may be a promising therapeutic intervention to prevent CS + Tx-induced mitochondrial and renal injury.


Asunto(s)
Trasplante de Riñón/efectos adversos , Riñón/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Mitocondrias/efectos de los fármacos , Tetrazoles/farmacología , Tiourea/análogos & derivados , Animales , Muerte Celular/efectos de los fármacos , Criopreservación , Riñón/metabolismo , Riñón/patología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Mitocondrias/metabolismo , Ratas , Tiourea/farmacología
9.
Cardiovasc Drugs Ther ; 33(5): 581-588, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31705225

RESUMEN

PURPOSE: Small and big conductance Ca2+-sensitive potassium (KCa) channels are involved in cardioprotective measures aiming at reducing myocardial reperfusion injury. For levosimendan, infarct size-reducing effects were shown. Whether activation of these channels is involved in levosimendan-induced postconditioning is unknown. We hypothesized that levosimendan exerts a concentration-dependent cardioprotective effect and that both types of Ca2+-sensitive potassium channels are involved. METHODS: In a prospective blinded experimental laboratory investigation, hearts of male Wistar rats were randomized and placed on a Langendorff system, perfused with Krebs-Henseleit buffer at a constant pressure of 80 mmHg. All hearts were subjected to 33 min of global ischemia and 60 min of reperfusion. At the onset of reperfusion, hearts were perfused with various concentrations of levosimendan (0.03-1 µM) in order to determine a concentration-response relationship. To elucidate the involvement of KCa-channels for the observed cardioprotection, in the second set of experiments, 0.3 µM levosimendan was administered in combination with the subtype-specific KCa-channel inhibitors paxilline (1 µM, big KCa-channel) and NS8593 (0.1 µM, small KCa-channel) respectively. Infarct size was determined by tetrazolium chloride (TTC) staining. RESULTS: Infarct size in controls was 60 ± 7% and 59 ± 6% respectively. Levosimendan at a concentration of 0.3 µM reduced infarct size to 30 ± 5% (P < 0.0001 vs. control). Higher concentrations of levosimendan did not induce a stronger effect. Paxilline but not NS8593 completely abolished levosimendan-induced cardioprotection while both substances alone had no effect on infarct size. CONCLUSIONS: Cardioprotection by levosimendan-induced postconditioning shows a binary phenomenon, either ineffective or with maximal effect. The cardioprotective effect requires activation of big but not small KCa channels.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Precondicionamiento Isquémico Miocárdico , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Simendán/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Preparación de Corazón Aislado , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Wistar , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
10.
Cells ; 8(10)2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31575085

RESUMEN

The arachidonic acid metabolism through 5-lipoxygenase (5-LO) pathways is involved in modulating both tumorigenesis and angiogenesis. Although anti-carcinogenic activities of certain 5-LO inhibitors have been reported, the role of zileuton, a well known 5-LO inhibitor, on the endothelial cell proliferation and angiogenesis has not been fully elucidated. Here, we report that zileuton has an anti-angiogenic effect, and the underlying mechanisms involved activation of the large-conductance Ca2+-activated K+ (BK) channel. Our results show that zileuton significantly prevented vascular endothelial growth factor (VEGF)-induced proliferation of human umbilical vein endothelial cells (HUVECs) in vitro, as well as in vivo. However, such anti-angiogenic effect of zileuton was abolished by iberiotoxin (IBTX), a BK channel blocker, suggesting zileuton-induced activation of BK channel was critical for the observed anti-angiogenic effect of zileuton. Furthermore, the anti-angiogenic effect of zileuton was, at least, due to the activation of pro-apoptotic signaling cascades which was also abolished by IBTX. Additionally, zileuton suppressed the expression of VCAM-1, ICAM-1, ETS related gene (Erg) and the production of nitric oxide (NO). Taken together, our results show that zileuton prevents angiogenesis by activating the BK channel dependent-apoptotic pathway, thus highlighting its therapeutic capacity in angiogenesis-related diseases, such as cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Hidroxiurea/análogos & derivados , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Inhibidores de la Lipooxigenasa/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Hidroxiurea/farmacología , Ratones , Ratones Endogámicos C57BL
11.
J Comput Neurosci ; 46(3): 233-256, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31025235

RESUMEN

The large conductance voltage and calcium activated potassium (BK) channels play a crucial role in regulating the excitability of detrusor smooth muscle, which lines the wall of the urinary bladder. These channels have been widely characterized in terms of their molecular structure, pharmacology and electrophysiology. They control the repolarising and hyperpolarising phases of the action potential, thereby regulating the firing frequency and contraction profiles of the smooth muscle. Several groups have reported varied profiles of BK currents and I-V curves under similar experimental conditions. However, no single computational model has been able to reconcile these apparent discrepancies. In view of the channels' physiological importance, it is imperative to understand their mechanistic underpinnings so that a realistic model can be created. This paper presents a computational model of the BK channel, based on the Hodgkin-Huxley formalism, constructed by utilising three activation processes - membrane potential, calcium inflow from voltage-gated calcium channels on the membrane and calcium released from the ryanodine receptors present on the sarcoplasmic reticulum. In our model, we attribute the discrepant profiles to the underlying cytosolic calcium received by the channel during its activation. The model enables us to make heuristic predictions regarding the nature of the sub-membrane calcium dynamics underlying the BK channel's activation. We have employed the model to reproduce various physiological characteristics of the channel and found the simulated responses to be in accordance with the experimental findings. Additionally, we have used the model to investigate the role of this channel in electrophysiological signals, such as the action potential and spontaneous transient hyperpolarisations. Furthermore, the clinical effects of BK channel openers, mallotoxin and NS19504, were simulated for the detrusor smooth muscle cells. Our findings support the proposed application of these drugs for amelioration of the condition of overactive bladder. We thus propose a physiologically realistic BK channel model which can be integrated with other biophysical mechanisms such as ion channels, pumps and exchangers to further elucidate its micro-domain interaction with the intracellular calcium environment.


Asunto(s)
Señalización del Calcio/fisiología , Simulación por Computador , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Miocitos del Músculo Liso/fisiología , Vejiga Urinaria/fisiología , Acetofenonas/farmacología , Benzopiranos/farmacología , Calcio/fisiología , Citosol/metabolismo , Fenómenos Electrofisiológicos , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Potenciales de la Membrana/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/metabolismo , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Vejiga Urinaria Hiperactiva/fisiopatología
12.
Science ; 363(6429): 875-880, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30792303

RESUMEN

Potassium (K+) channels have been evolutionarily tuned for activation by diverse biological stimuli, and pharmacological activation is thought to target these specific gating mechanisms. Here we report a class of negatively charged activators (NCAs) that bypass the specific mechanisms but act as master keys to open K+ channels gated at their selectivity filter (SF), including many two-pore domain K+ (K2P) channels, voltage-gated hERG (human ether-à-go-go-related gene) channels and calcium (Ca2+)-activated big-conductance potassium (BK)-type channels. Functional analysis, x-ray crystallography, and molecular dynamics simulations revealed that the NCAs bind to similar sites below the SF, increase pore and SF K+ occupancy, and open the filter gate. These results uncover an unrecognized polypharmacology among K+ channel activators and highlight a filter gating machinery that is conserved across different families of K+ channels with implications for rational drug design.


Asunto(s)
Clorobencenos/farmacología , Canal de Potasio ERG1/agonistas , Canal de Potasio ERG1/química , Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Tetrahidronaftalenos/farmacología , Tetrazoles/farmacología , Tiourea/análogos & derivados , ortoaminobenzoatos/farmacología , Animales , Células CHO , Clorobencenos/química , Cricetulus , Cristalografía por Rayos X , Diseño de Fármacos , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Dominios Proteicos , Tetrahidronaftalenos/química , Tetrazoles/química , Tiourea/química , Tiourea/farmacología , Xenopus , ortoaminobenzoatos/química
13.
Cardiovasc Drugs Ther ; 32(5): 427-434, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30120617

RESUMEN

PURPOSE: Activation of mitochondrial large-conductance Ca2+-sensitive potassium (mBKCa)-channels is a crucial step for cardioprotection by preconditioning. Whether activation of these channels is involved in levosimendan-induced preconditioning is unknown. We investigated if cardioprotection by levosimendan requires activation of mBKCa-channels in the rat heart in vitro. METHODS: In a prospective blinded experimental laboratory investigation, hearts of male Wistar rats were randomized and placed on a Langendorff system, perfused with Krebs-Henseleit buffer at a constant pressure of 80 mmHg. All hearts were subjected to 33 min of global ischemia and 60 min of reperfusion. Before ischemia, hearts were perfused with different concentrations of levosimendan (0.03-1 µM) for determination of a dose-effect curve. In a second set of experiments, 0.3 µM levosimendan was administered in combination with the mBKCa-channel inhibitor paxilline (1 µM). Infarct size was determined by TTC staining. RESULTS: In control, animal's infarct size was 58 ± 7%. Levosimendan at a concentration of 0.3 µM reduced infarct size to 30 ± 7% (P < 0.05 vs. control). Higher concentrations with 1 µM levosimendan did not confer stronger protection. Paxilline completely blocked levosimendan-induced cardioprotection while paxilline alone had no effect on infarct size. CONCLUSIONS: This study shows that activation of mBKCa-channels plays a pivotal role in levosimendan-induced preconditioning.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Mitocondrias Cardíacas/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Simendán/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Indoles/farmacología , Preparación de Corazón Aislado , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Bloqueadores de los Canales de Potasio/farmacología , Ratas Wistar , Función Ventricular Izquierda/efectos de los fármacos
14.
Neurogastroenterol Motil ; 30(7): e13312, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29488290

RESUMEN

BACKGROUND: Muscarinic acetylcholine receptor (mAChR) activation is an important factor to enhance the motility of gastrointestinal (GI) smooth muscle. Large conductance Ca2+ -activated potassium (BK) channels are widely expressed in GI smooth muscle. Roles of BK in carbachol (a mAChR agonist) induced enhancement of GI motility and the molecular mechanisms remains unknown and were investigated in this study. METHODS: Colonic smooth muscle (CSM) strip was perfused to record motility in vitro. The patch-clamp technique was used to record BK currents. RT-PCR was used to detect the expression of BK channels in rat CSM tissues. Two different types BK channels were constructed in HEK293 cells to investigate the regulation mechanism. Paired t tests were set with a P < .05 regarded as significant. KEY RESULTS: Carbachol enhanced CSM contraction through M3 receptor (M3 R) were attenuated by IbTX, an inhibitor of BK. Carbachol inhibited BK currents in CSM cells and Go6983, an inhibitor of protein kinase C (PKC), reversed the effect. PKC activator, phorbol 12-myristate 13-acetate (PMA), inhibited BK currents. Two types of BK channels (ZERO-BK and STREX-BK) were detected in CSM. ZERO- but not STREX-BK channels expressed in HEK293 cells were inhibited by PMA. CONCLUSION: Our results provide strong evidence that inhibition of ZERO-BK but not STREX-BK channels via PKC pathway is involved in the enhancement of CSM motility by mAChR activation. Besides the activation of BK by an increase in intracellular calcium, inhibition of BK played an important role in GI motility regulation during mAChR activation.


Asunto(s)
Carbacol/farmacología , Colon/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Músculo Liso/efectos de los fármacos , Proteína Quinasa C/metabolismo , Animales , Bencimidazoles/farmacología , Agonistas Colinérgicos/farmacología , Colon/enzimología , Motilidad Gastrointestinal/fisiología , Células HEK293 , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Masculino , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso/enzimología , Ratas , Ratas Sprague-Dawley
15.
Int J Mol Sci ; 19(2)2018 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-29370072

RESUMEN

Potassium channel openers (KCOs) have been shown to play a role in cytoprotection through the activation of mitochondrial potassium channels. Recently, in several reports, a number of data has been described as off-target actions for KCOs. In the present study, we investigated the effects of BKCa channel openers CGS7181, CGS7184, NS1619, and NS004 in neuronal cells. For the purpose of this research, we used a rat brain, the mouse hippocampal HT22 cells, and the human astrocytoma U-87 MG cell line. We showed that CGS7184 activated the mitochondrial BKCa (mitoBKCa) channel in single-channel recordings performed on astrocytoma mitoplasts. Moreover, when applied to the rat brain homogenate or isolated rat brain mitochondria, CGS7184 increased the oxygen consumption rate, and can thus be considered a potentially cytoprotective agent. However, experiments on intact neuronal HT22 cells revealed that both CGS7181 and CGS7184 induced HT22 cell death in a concentration- and time-dependent manner. By contrast, we did not observe cell death when NS1619 or NS004 was applied. CGS7184 toxicity was not abolished by BKCa channel inhibitors, suggesting that the observed effects were independent of a BKCa-type channel activity. CGS7184 treatment resulted in an increase of cytoplasmic Ca2+ concentration that likely involved efflux from internal calcium stores and the activation of calpains (calcium-dependent proteases). The cytotoxic effect of the channel opener was partially reversed by a calpain inhibitor. Our data show that KCOs under study not only activate mitoBKCa channels from brain tissue, but also induce cell death when used in cellular models.


Asunto(s)
Indoles/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Moduladores del Transporte de Membrana/farmacología , Proteínas Mitocondriales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Calpaína/metabolismo , Línea Celular Tumoral , Humanos , Indoles/toxicidad , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Masculino , Moduladores del Transporte de Membrana/toxicidad , Proteínas Mitocondriales/agonistas , Ratas , Ratas Wistar
16.
Cereb Cortex ; 28(2): 433-446, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27999123

RESUMEN

We studied the effect of Amyloid ß 1-42 oligomers (Abeta42) on Ca2+ dependent excitability profile of hippocampal neurons. Abeta42 is one of the Amyloid beta peptides produced by the proteolytic processing of the amyloid precursor protein and participates in the initiating event triggering the progressive dismantling of synapses and neuronal circuits. Our experiments on cultured hippocampal network reveal that Abeta42 increases intracellular Ca2+ concentration by 46% and inhibits firing discharge by 19%. More precisely, Abeta42 differently regulates ryanodine (RyRs), NMDA receptors (NMDARs), and voltage gated calcium channels (VGCCs) by increasing Ca2+ release through RyRs and inhibiting Ca2+ influx through NMDARs and VGCCs. The overall increased intracellular Ca2+ concentration causes stimulation of K+ current carried by big conductance Ca2+ activated potassium (BK) channels and hippocampal network firing inhibition. We conclude that Abeta42 alters neuronal function by means of at least 4 main targets: RyRs, NMDARs, VGCCs, and BK channels. The development of selective modulators of these channels may in turn be useful for developing effective therapies that could enhance the quality of life of AD patients during the early onset of the pathology.


Asunto(s)
Potenciales de Acción/fisiología , Péptidos beta-Amiloides/farmacología , Hipocampo/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Factores de Tiempo
17.
Am J Physiol Heart Circ Physiol ; 313(5): H988-H999, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28822969

RESUMEN

Activation of large-conductance Ca2+-activated K+ (BKCa) channels evokes cell survival programs that mitigate intestinal ischemia and reperfusion (I/R) inflammation and injury 24 h later. The goal of the present study was to determine the roles of reactive oxygen species (ROS) and heme oxygenase (HO)-1 in delayed acquisition of tolerance to I/R induced by pretreatment with the BKCa channel opener NS-1619. Superior mesentery arteries were occluded for 45 min followed by reperfusion for 70 min in wild-type (WT) or HO-1-null (HO-1-/-) mice that were pretreated with NS-1619 or saline vehicle 24 h earlier. Intravital microscopy was used to quantify the numbers of rolling and adherent leukocytes. Mucosal permeability, tumor necrosis factor-α (TNF-α) levels, and HO-1 activity and expression in jejunum were also determined. I/R induced leukocyte rolling and adhesion, increased intestinal TNF-α levels, and enhanced mucosal permeability in WT mice, effects that were largely abolished by pretreatment with NS-1619. The anti-inflammatory and mucosal permeability-sparing effects of NS-1619 were prevented by coincident treatment with the HO-1 inhibitor tin protoporphyrin-IX or a cell-permeant SOD mimetic, Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), in WT mice. NS-1619 also increased jejunal HO-1 activity in WT animals, an effect that was attenuated by treatment with the BKCa channel antagonist paxilline or MnTBAP. I/R also increased postischemic leukocyte rolling and adhesion and intestinal TNF-α levels in HO-1-/- mice to levels comparable to those noted in WT animals. However, NS-1619 was ineffective in preventing these effects in HO-1-deficient mice. In summary, our data indicate that NS-1619 induces the development of an anti-inflammatory phenotype and mitigates postischemic mucosal barrier disruption in the small intestine by a mechanism that may involve ROS-dependent HO-1 activity.NEW & NOTEWORTHY Antecedent treatment with the large-conductance Ca2+-activated K+ channel opener NS-1619 24 h before ischemia-reperfusion limits postischemic tissue injury by an oxidant-dependent mechanism. The present study shows that NS-1619-induced oxidant production prevents ischemia-reperfusion-induced inflammation and mucosal barrier disruption in the small intestine by provoking increases in heme oxygenase-1 activity.


Asunto(s)
Bencimidazoles/farmacología , Hemo-Oxigenasa 1/efectos de los fármacos , Inflamación/prevención & control , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Proteínas de la Membrana/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/prevención & control , Animales , Hemo-Oxigenasa 1/genética , Inflamación/etiología , Precondicionamiento Isquémico , Leucocitos/efectos de los fármacos , Leucocitos/enzimología , Leucocitos/metabolismo , Activación de Macrófagos , Masculino , Proteínas de la Membrana/genética , Arteria Mesentérica Superior/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/patología , Daño por Reperfusión/complicaciones , Daño por Reperfusión/fisiopatología , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Int Rev Neurobiol ; 128: 439-75, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27238271

RESUMEN

High conductance, calcium-activated potassium (BK) channels (KCa1.1) are important in regulating physiologic responses in many types of tissues and, as such, present opportunities for development of new therapeutic agents. Both channel agonists and inhibitors could have therapeutic utility, depending on medical application under consideration. However, characterization of molecular pharmacology of BK channels is incomplete and has been difficult to accomplish because of paucity of chemical leads that are acceptable templates for Medicinal Chemistry investigation. Only through continued prosecution of new high-throughput screening campaigns can this situation be rectified. Examples are presented of BK channel agonist and inhibitor discovery paradigms which will be useful for progressing BK channel future drug discovery strategies.


Asunto(s)
Descubrimiento de Drogas , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Moduladores del Transporte de Membrana/uso terapéutico , Calcio/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Potasio/metabolismo
19.
Neurochem Res ; 41(8): 1982-92, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27097551

RESUMEN

Seizure activity is linked to astrocyte activation as well as dysfunctional cortical neuron excitability produced from changes in calcium-activated potassium (KCa) channel function. Ciliary neurotrophic factor-treated astrocyte conditioned medium (CNTF-ACM) can be used to investigate the peripheral effects of activated astrocytes upon cortical neurons. However, CNTF-ACM's effect upon KCa channel activity in cultured cortical neurons has not yet been investigated. Whole-cell patch clamp recordings were performed in rat cortical neurons to evaluate CNTF-ACM's effects upon charybdotoxin-sensitive large-conductance KCa (BK) channel currents and apamin-sensitive small-conductance KCa (SK) channel current. Biotinylation and RT-PCR were applied to assess CNTF-ACM's effects upon the protein and mRNA expression, respectively, of the SK channel subunits SK2 and SK3 and the BK channel subunits BKα1 and BKß3. An anti-fibroblast growth factor-2 (FGF-2) monoclonal neutralizing antibody was used to assess the effects of the FGF-2 component of CNTF-ACM. CNTF-ACM significantly increased KCa channel current density, which was predominantly attributable to gains in BK channel activity (p < 0.05). CNTF-ACM produced a significant increase in BKα1 and BKß3 expression (p < 0.05) but had no significant effect upon SK2 or SK3 expression (p > 0.05). Blocking FGF-2 produced significant reductions in KCa channel current density (p > 0.05) as well as BKα1 and BKß3 expression in CNTF-ACM-treated neurons (p > 0.05). CNTF-ACM significantly enhances BK channel activity in rat cortical neurons and that FGF-2 is partially responsible for these effects. CNTF-induced astrocyte activation results in secretion of neuroactive factors which may affect neuronal excitability and resultant seizure activity in mammalian cortical neurons.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Factor Neurotrófico Ciliar/farmacología , Medios de Cultivo Condicionados/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
20.
Am J Physiol Cell Physiol ; 310(4): C284-92, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26659726

RESUMEN

Large-conductance Ca(2+)-activated K(+) (BKCa) channels are thought to play a key role in the regulation of corpus cavernosum smooth muscle (CCSM) excitability. Few BKCa channel openers have been accepted for clinical development. The effect of the novel BKCa channel opener GoSlo-SR5-130 on electrical activity in isolated rabbit CCSM cells and mechanical activity in strips of rabbit CCSM was examined. Single-channel currents were observed in inside-out patches. These channels were sensitive to Ca(2+), blocked by penitrem A, and had a conductance of 291 ± 20 pS (n = 7). In the presence of GoSlo-SR5-130, the number of open BKCa channels increased. Using voltage-ramp protocols, GoSlo-SR5-130 caused currents to activate at more negative potentials in a concentration-dependent manner, shifting the half-maximal activation voltage potential to the left on the voltage axis. Therefore, BKCa channels were open within the physiological range of membrane potentials in the presence of GoSlo-SR5-130. GoSlo-SR5-130 also resulted in an increase in the activity of spontaneous transient outward currents in myocytes isolated from CCSM, and this effect was reversed by iberiotoxin. In current-clamp mode, GoSlo-SR5-130 hyperpolarized the cell membrane. Isometric tension recording of strips of rabbit corpus cavernosum showed that GoSlo-SR5-130 inhibited spontaneous contractions in a concentration-dependent manner. This effect was reversed in the presence of iberiotoxin, suggesting that GoSlo-SR5-130 exerts its effect through BKCa channels. These findings suggest that GoSlo-SR5-130 is an effective tool for the study of BKCa channels and that these channels can modulate CCSM activity and are possible targets for the treatment of erectile dysfunction.


Asunto(s)
Antraquinonas/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Erección Peniana/efectos de los fármacos , Pene/irrigación sanguínea , Potasio/metabolismo , Ácidos Sulfónicos/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Animales , Relación Dosis-Respuesta a Droga , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Conejos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...