Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36835507

RESUMO

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.


Assuntos
Células Epiteliais Alveolares , Quimiocina CCL2 , Canais de Potássio Ativados por Cálcio de Condutância Alta , Pneumonia , Fator de Necrose Tumoral alfa , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Nifedipino/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Pneumonia/metabolismo , Pneumonia/prevenção & controle , Quimiocina CCL2/metabolismo
2.
Neurol India ; 70(4): 1601-1609, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36076665

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Canais de Potássio Ativados por Cálcio de Condutância Alta , Fármacos Neuroprotetores , Receptores de Progesterona , Animais , Lesões Encefálicas/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Proteínas de Transporte , Modelos Animais de Doenças , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Masculino , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Progesterona/antagonistas & inibidores , Proteína X Associada a bcl-2/metabolismo
3.
Bioorg Med Chem ; 28(16): 115609, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32690264

RESUMO

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.


Assuntos
Encefalopatias/tratamento farmacológico , Descoberta de Drogas , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Encefalopatias/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Terapia de Alvo Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores
4.
ACS Chem Biol ; 15(8): 2098-2106, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32667185

RESUMO

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.


Assuntos
Monóxido de Carbono/metabolismo , Compostos Ferrosos/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Fotólise , Sítios de Ligação , Cálcio/metabolismo , Compostos Ferrosos/metabolismo , Células HEK293 , Heme/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Magnésio/metabolismo , Técnicas de Patch-Clamp
5.
Arch Biochem Biophys ; 688: 108410, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32446891

RESUMO

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.


Assuntos
Transplante de Rim/efeitos adversos , Rim/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Mitocôndrias/efeitos dos fármacos , Tetrazóis/farmacologia , Tioureia/análogos & derivados , Animais , Morte Celular/efeitos dos fármacos , Criopreservação , Rim/metabolismo , Rim/patologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Masculino , Mitocôndrias/metabolismo , Ratos , Tioureia/farmacologia
6.
Cells ; 8(10)2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31575085

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Hidroxiureia/análogos & derivados , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Inibidores de Lipoxigenase/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Hidroxiureia/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
7.
J Comput Neurosci ; 46(3): 233-256, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31025235

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , Simulação por Computador , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Miócitos de Músculo Liso/fisiologia , Bexiga Urinária/fisiologia , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Cálcio/fisiologia , Citosol/metabolismo , Fenômenos Eletrofisiológicos , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Potenciais da Membrana/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático/metabolismo , Bexiga Urinária Hiperativa/tratamento farmacológico , Bexiga Urinária Hiperativa/fisiopatologia
8.
Science ; 363(6429): 875-880, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30792303

RESUMO

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.


Assuntos
Clorobenzenos/farmacologia , Canal de Potássio ERG1/agonistas , Canal de Potássio ERG1/química , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Tetra-Hidronaftalenos/farmacologia , Tetrazóis/farmacologia , Tioureia/análogos & derivados , ortoaminobenzoatos/farmacologia , Animais , Células CHO , Clorobenzenos/química , Cricetulus , Cristalografia por Raios X , Desenho de Fármacos , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Domínios Proteicos , Tetra-Hidronaftalenos/química , Tetrazóis/química , Tioureia/química , Tioureia/farmacologia , Xenopus , ortoaminobenzoatos/química
9.
Int J Mol Sci ; 19(2)2018 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-29370072

RESUMO

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.


Assuntos
Indóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Moduladores de Transporte de Membrana/farmacologia , Proteínas Mitocondriais/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Calpaína/metabolismo , Linhagem Celular Tumoral , Humanos , Indóis/toxicidade , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Masculino , Moduladores de Transporte de Membrana/toxicidade , Proteínas Mitocondriais/agonistas , Ratos , Ratos Wistar
10.
Am J Physiol Heart Circ Physiol ; 313(5): H988-H999, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28822969

RESUMO

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.


Assuntos
Benzimidazóis/farmacologia , Heme Oxigenase-1/efeitos dos fármacos , Inflamação/prevenção & controle , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Proteínas de Membrana/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Heme Oxigenase-1/genética , Inflamação/etiologia , Precondicionamento Isquêmico , Leucócitos/efeitos dos fármacos , Leucócitos/enzimologia , Leucócitos/metabolismo , Ativação de Macrófagos , Masculino , Proteínas de Membrana/genética , Artéria Mesentérica Superior/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa/patologia , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/fisiopatologia , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Exp Gerontol ; 56: 99-105, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24727217

RESUMO

The beneficial effects of many cardioprotective strategies including ischemic or pharmacological conditioning are reduced in the aged heart. The underlying reason(s) for the age-dependent loss of cardioprotection is unclear. Recently, we demonstrated that protein kinase A (PKA) dependent cardioprotection is lost in the aged heart. However, activation of large-conductance Ca(2+)-sensitive K(+) (BK(Ca)) channels, a putative PKA downstream target, initiated cardioprotection also in the aged heart. Therefore, we aimed to investigate whether 1) BK(Ca) channels are critically involved in PKA activation induced cardioprotection and 2) the age-dependent loss of cardioprotection is caused by differences in PKA regulation. Using an in vivo rat model with regional myocardial ischemia, we treated young (2-4 months) and aged (22-24 months) Wistar rats with PKA activator forskolin, BK(Ca) channel activator NS1619 and/or BK(Ca) channel blocker iberiotoxin. Forskolin induced infarct size reduction was 1) age-dependent and 2) prevented by iberiotoxin. The effect of forskolin on myocardial PKA activity was comparable in young and aged animals. In addition, NS1619 initiated cardioprotection also in the aged heart both when administered before ischemia and during early reperfusion phase. Activation of BK(Ca) channels is critically involved in forskolin induced cardioprotection. The age-dependency of forskolin induced cardioprotection is not caused by age-dependent differences in PKA activation. Pharmacological targeting of BK(Ca) channels before or after myocardial ischemia is a promising therapeutic strategy to protect the aged heart from ischemia and reperfusion injury.


Assuntos
Envelhecimento/metabolismo , Benzimidazóis/farmacologia , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativadores de Enzimas/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Transdução de Sinais/efeitos dos fármacos , Fatores Etários , Envelhecimento/patologia , Animais , Citoproteção , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Masculino , Terapia de Alvo Molecular , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos Wistar
12.
Immunology ; 141(3): 345-52, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24138091

RESUMO

Increasing evidence has demonstrated that Toll-like receptor 4 (TLR4) -mediated systemic inflammatory response syndrome accompanied by multiple organ failure, is one of the most common causes of death in patients with severe acute pancreatitis. Recent reports have revealed that heparan sulphate (HS) proteoglycan, a component of extracellular matrices, potentiates the activation of intracellular pro-inflammatory responses via TLR4, contributing to the aggravation of acute pancreatitis. However, little is known about the participants in the HS/TLR4-mediated inflammatory cascades. Our previous work provided a clue that a membrane potassium channel (MaxiK) is responsible for HS-induced production of inflammatory cytokines. Therefore, in this report we attempted to reveal the roles of MaxiK in the activation of macrophages stimulated by HS. Our results showed that incubation of RAW264.7 cells with HS up-regulated MaxiK and TLR4 expression levels. HS could also activate MaxiK channels to promote the efflux of potassium ions from cells, as measured by the elevated activity of caspase-1, whereas this was significantly abolished by treatment with paxilline, a specific blocker of the MaxiK channel. Moreover, it was found that paxilline substantially inhibited HS-induced activation of several different transcription factors in macrophages, including nuclear factor-κB, p38 and interferon regulatory factor-3, followed by decreased production of tumour necrosis factor-α and interferon-ß. Taken together, our investigation provides evidence that the HS/TLR4-mediated intracellular inflammatory cascade depends on the activation of MaxiK, which may offer an important opportunity for a new approach in therapeutic strategies of severe acute pancreatitis.


Assuntos
Membrana Celular/efeitos dos fármacos , Heparitina Sulfato/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Potássio/metabolismo , Animais , Caspase 1/metabolismo , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Imunidade Inata/efeitos dos fármacos , Fator Regulador 3 de Interferon/metabolismo , Interleucina-1beta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Macrófagos/metabolismo , Potenciais da Membrana , Camundongos , NF-kappa B/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Am J Physiol Cell Physiol ; 305(6): C609-22, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23804200

RESUMO

We used the perforated patch-clamp technique at 37°C to investigate the mechanisms underlying the activation of a transient large-conductance K(+) (tBK) current in rabbit urethral smooth muscle cells. The tBK current required an elevation of intracellular Ca(2+), resulting from ryanodine receptor (RyR) activation via Ca(2+)-induced Ca(2+) release, triggered by Ca(2+) influx through L-type Ca(2+) (CaV) channels. Carbachol inhibited tBK current by reducing Ca(2+) influx and Ca(2+) release and altered the shape of spike complexes recorded under current-clamp conditions. The tBK currents were blocked by iberiotoxin and penitrem A (300 and 100 nM, respectively) and were also inhibited when external Ca(2+) was removed or the CaV channel inhibitors nifedipine (10 µM) and Cd(2+) (100 µM) were applied. The tBK current was inhibited by caffeine (10 mM), ryanodine (30 µM), and tetracaine (100 µM), suggesting that RyR-mediated Ca(2+) release contributed to the activation of the tBK current. When IP3 receptors (IP3Rs) were blocked with 2-aminoethoxydiphenyl borate (2-APB, 100 µM), the amplitude of the tBK current was not reduced. However, when Ca(2+) release via IP3Rs was evoked with phenylephrine (1 µM) or carbachol (1 µM), the tBK current was inhibited. The effect of carbachol was abolished when IP3Rs were blocked with 2-APB or by inhibition of muscarinic receptors with the M3 receptor antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide (1 µM). Under current-clamp conditions, bursts of action potentials could be evoked with depolarizing current injection. Carbachol reduced the number and amplitude of spikes in each burst, and these effects were reduced in the presence of 2-APB. In the presence of ryanodine, the number and amplitude of spikes were also reduced, and carbachol was without further effect. These data suggest that IP3-generating agonists can modulate the electrical activity of rabbit urethral smooth muscle cells and may contribute to the effects of neurotransmitters on urethral tone.


Assuntos
Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Uretra/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Compostos de Boro/farmacologia , Cafeína/farmacologia , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Carbacol/farmacologia , Feminino , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Masculino , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Micotoxinas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Peptídeos/farmacologia , Potássio/metabolismo , Coelhos , Receptor Muscarínico M3/metabolismo , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Tetracaína/farmacologia , Uretra/efeitos dos fármacos
14.
J Neurophysiol ; 109(10): 2528-41, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23446695

RESUMO

Purkinje cells have specialized intrinsic ionic conductances that generate high-frequency action potentials. Disruptions of their Ca or Ca-activated K (KCa) currents correlate with altered firing patterns in vitro and impaired motor behavior in vivo. To examine the properties of somatic KCa currents, we recorded voltage-clamped KCa currents in Purkinje cell bodies isolated from postnatal day 17-21 mouse cerebellum. Currents were evoked by endogenous Ca influx with approximately physiological Ca buffering. Purkinje somata expressed voltage-activated, Cd-sensitive KCa currents with iberiotoxin (IBTX)-sensitive (>100 nS) and IBTX-insensitive (>75 nS) components. IBTX-sensitive currents activated and partially inactivated within milliseconds. Rapid, incomplete macroscopic inactivation was also evident during 50- or 100-Hz trains of 1-ms depolarizations. In contrast, IBTX-insensitive currents activated more slowly and did not inactivate. These currents were insensitive to the small- and intermediate-conductance KCa channel blockers apamin, scyllatoxin, UCL1684, bicuculline methiodide, and TRAM-34, but were largely blocked by 1 mM tetraethylammonium. The underlying channels had single-channel conductances of ∼150 pS, suggesting that the currents are carried by IBTX-resistant (ß4-containing) large-conductance KCa (BK) channels. IBTX-insensitive currents were nevertheless increased by small-conductance KCa channel agonists EBIO, chlorzoxazone, and CyPPA. During trains of brief depolarizations, IBTX-insensitive currents flowed during interstep intervals, and the accumulation of interstep outward current was enhanced by EBIO. In current clamp, EBIO slowed spiking, especially during depolarizing current injections. The two components of BK current in Purkinje somata likely contribute differently to spike repolarization and firing rate. Moreover, augmentation of BK current may partially underlie the action of EBIO and chlorzoxazone to alleviate disrupted Purkinje cell firing associated with genetic ataxias.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células de Purkinje/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Benzimidazóis/farmacologia , Cádmio/farmacologia , Cálcio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Cerebelo/fisiologia , Clorzoxazona/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Células de Purkinje/fisiologia , Pirazóis/farmacologia , Pirimidinas/farmacologia
15.
Microcirculation ; 20(2): 170-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23002811

RESUMO

OBJECTIVE: Test the hypothesis that exercise training increases the contribution of BK(Ca) channels to endothelium-mediated dilation in coronary arterioles from collateral-dependent myocardial regions of chronically occluded pig hearts and may function downstream of H2O2. METHODS: An ameroid constrictor was placed around the proximal left circumflex coronary artery to induce gradual occlusion in Yucatan miniature swine. Eight weeks postoperatively, pigs were randomly assigned to sedentary or exercise training (treadmill; 14 week) regimens. RESULTS: Exercise training significantly enhanced bradykinin-mediated dilation in collateral-dependent arterioles (~125 µm diameter) compared with sedentary pigs. The BK(Ca) -channel blocker, iberiotoxin alone or in combination with the H2O2 scavenger, polyethylene glycol catalase, reversed exercise training-enhanced dilation in collateral-dependent arterioles. Iberiotoxin-sensitive whole-cell K+ currents (i.e., BK(Ca)-channel currents) were not different between smooth muscle cells of nonoccluded and collateral-dependent arterioles of sedentary and exercise trained groups. CONCLUSIONS: These data provide evidence that BK(Ca)-channel activity contributes to exercise training-enhanced endothelium-dependent dilation in collateral-dependent coronary arterioles despite no change in smooth muscle BK(Ca)-channel current. Taken together, our findings suggest that a component of the bradykinin signaling pathway, which stimulates BK(Ca) channels, is enhanced by exercise training in collateral-dependent arterioles and suggest a potential role for H2O2 as the mediator.


Assuntos
Arteríolas/fisiologia , Circulação Coronária/fisiologia , Endotélio Vascular/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Condicionamento Físico Animal/fisiologia , Vasodilatação/fisiologia , Animais , Bradicinina/farmacologia , Bradicinina/fisiologia , Catalase/metabolismo , Catalase/farmacologia , Circulação Colateral/fisiologia , Feminino , Peróxido de Hidrogênio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Músculo Liso Vascular/fisiologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Polietilenoglicóis/farmacologia , Potássio/metabolismo , Distribuição Aleatória , Superóxido Dismutase/metabolismo , Suínos , Porco Miniatura
16.
Sheng Li Xue Bao ; 64(2): 121-8, 2012 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-22513460

RESUMO

The aim of the present study was to study the effect of ß-estradiol (ß-E(2)) on the large-conductance Ca(2+)-activated potassium (BK(Ca)) channel in mesenteric artery smooth muscle cells (SMCs). The mesenteric arteries were obtained from post-menopause female patients with abdominal surgery, and the SMCs were isolated from the arteries using an enzymatic disassociation. According to the sources, the SMCs were divided into non-hypertension (NH) and essential hypertension (EH) groups. Single channel patch clamp technique was used to investigate the effect of ß-E(2) and ICI 182780 (a specific blocker of estrogen receptor) on BK(Ca) in the SMCs. The results showed the opening of BK(Ca) in the SMCs was voltage and calcium dependent, and could be blocked by IbTX. ß-E(2) (100 µmol/L) significantly increased open probability (Po) of BK(Ca) in both NH and EH groups. After ß-E(2) treatment, NH group showed higher Po of BK(Ca) compared with EH group. ICI 182780 could inhibit the activating effect of ß-E(2) on BK(Ca) in no matter NH or EH groups. These results suggest ß-E(2) activates BK(Ca) in mesenteric artery SMCs from post-menopause women via estrogen receptor, but hypertension may decline the activating effect of ß-E(2) on BK(Ca).


Assuntos
Estradiol/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Artérias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Pós-Menopausa/fisiologia , Idoso , Estradiol/análogos & derivados , Feminino , Fulvestranto , Humanos , Hipertensão/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Artérias Mesentéricas/fisiologia , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Técnicas de Patch-Clamp , Receptores de Estrogênio/antagonistas & inibidores
17.
Hear Res ; 285(1-2): 13-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22366501

RESUMO

There are two types of hair cells in the sensory epithelium of vestibular end organ. Type II vestibular hair cell (VHC II) is innervated by the efferent nerve endings, which employ a cholinergic inhibition mediated by SK channels through the activation of α9-containing nAChR. Our previous studies demonstrated that a BK-type cholinergic inhibition was present in guinea pig VHCs II, which may be mediated by an unknown mAChR. In this study, BK channel activities triggered by ACh were studied to determine the mAChR subtype and function. We found the BK channel was insensitive to α9-containing nAChR antagonists and m1, m3, m4 muscarinic antagonists, but potently inhibited by the m2 muscarinic antagonist. Muscarinic agonists could mimic the effect of ACh and be blocked by m2 antagonist. cAMP analog activated the BK current and adenyl cyclase (AC) inhibitor inhibited the ACh response. Inhibitor of Giα subunit failed to affect the BK current, but inhibitor of Giα and Gißγ subunits showed a potent inhibition to these currents. Our findings provide the physiological evidence that mAChRs may locate in guinea pig VHCs II, and m2 mAChRs may play a dominant role in BK-type cholinergic inhibition. The activation of m2 mAChRs may stimulate Gißγ-mediated excitation of AC/cAMP activities and lead to the phosphorylation of Ca(2+) channels, resulting in the influx of Ca(2+) and opening of the BK channel.


Assuntos
Células Ciliadas Vestibulares/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Receptor Muscarínico M2/metabolismo , Acetilcolina/farmacologia , Adenilil Ciclases/metabolismo , Animais , AMP Cíclico/metabolismo , Fenômenos Eletrofisiológicos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Cobaias , Células Ciliadas Vestibulares/classificação , Células Ciliadas Vestibulares/efeitos dos fármacos , Técnicas In Vitro , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Modelos Biológicos , Agonistas Muscarínicos/farmacologia , Antagonistas Muscarínicos/farmacologia , Técnicas de Patch-Clamp
18.
Mol Pharmacol ; 81(4): 567-77, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22241372

RESUMO

High-conductance calcium-activated potassium (Maxi-K) channels are present in smooth muscle where they regulate tone. Activation of Maxi-K channels causes smooth muscle hyperpolarization and shortening of action-potential duration, which would limit calcium entry through voltage-dependent calcium channels leading to relaxation. Although Maxi-K channels appear to indirectly mediate the relaxant effects of a number of agents, activators that bind directly to the channel with appropriate potency and pharmacological properties useful for proof-of-concept studies are not available. Most agents identified to date display significant polypharmacy that severely compromises interpretation of experimental data. In the present study, a high-throughput, functional, cell-based assay for identifying Maxi-K channel agonists was established and used to screen a large sample collection (>1.6 million compounds). On the basis of potency and selectivity, a family of tetrahydroquinolines was further characterized. Medicinal chemistry efforts afforded identification of compound X, from which its two enantiomers, Y and Z, were resolved. In in vitro assays, Z is more potent than Y as a channel activator. The same profile is observed in tissues where the ability of either agent to relax precontracted smooth muscles, via a potassium channel-dependent mechanism, is demonstrated. These data, taken together, suggest that direct activation of Maxi-K channels represents a mechanism to be explored for the potential treatment of a number of diseases associated with smooth muscle hyperexcitability.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Músculo Liso/fisiologia , Animais , Células CHO , Cromatografia Líquida , Cricetinae , Cricetulus , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Relaxamento Muscular
19.
Mol Pharmacol ; 81(1): 21-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21984254

RESUMO

Large conductance, Ca(2+)-activated K channel proteins are involved in a wide range of physiological activities, so there is considerable interest in the pharmacology of large conductance calcium-activated K (BK) channels. One potent activator of BK channels is mallotoxin (MTX), which produces a very large hyperpolarizing shift of the voltage gating of heterologously expressed BK channels and causes a dramatic increase in the activity of BK channels in human smooth muscle cells. However, we found that MTX shifted the steady-state activation of BK channels in native parotid acinar cells by only 6 mV. This was not because the parotid BK isoform (parSlo) is inherently insensitive to MTX as MTX shifted the activation of heterologously expressed parSlo channels by 70 mV. Even though MTX had a minimal effect on steady-state activation of parotid BK channels, it produced an approximate 2-fold speeding of the channel-gating kinetics. The BK channels in parotid acinar cells have a much more hyperpolarized voltage activation range than BK channels in most other cell types. We found that this is probably attributable to an accessory protein, LRRC26, which is expressed in parotid glands: expressed parSlo + LRRC26 channels were resistant to the actions of MTX. Another class of BK activators is the benzimidazalones that includes 1,3-dihydro-1-(2-hydroxy-5-(trifluoromethyl)phenyl)-5-(trifluoromethyl)-2H-benzimidazol-2-one (NS-1619). Although the LRRC26 accessory protein strongly inhibited the ability of MTX to activate BK channels, we found that it had only a small effect on the action of NS-1619 on BK channels. Thus, the LRRC26 BK channel accessory protein selectively alters the pharmacology of BK channels.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas de Neoplasias/fisiologia , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Animais , Benzimidazóis/metabolismo , Benzimidazóis/farmacologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Glândula Parótida/efeitos dos fármacos , Glândula Parótida/metabolismo , Técnicas de Patch-Clamp , Picrotoxina/análogos & derivados , Picrotoxina/toxicidade , Canais de Potássio/agonistas , Canais de Potássio/biossíntese
20.
Cell Calcium ; 51(2): 131-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22204737

RESUMO

The mouse cortical collecting duct (CCD) M-1 cells were grown to confluency on coverslips to assess the interaction between TRPV4 and Ca(2+)-activated K(+) channels. Immunocytochemistry demonstrated strong expression of TRPV4, along with the CCD marker, aquaporin-2, and the Ca(2+)-activated K(+) channels, the small conductance SK3 (K(Ca)2.3) channel and large conductance BKα channel (K(Ca)1.1). TRPV4 overexpression studies demonstrated little physical dependency of the K(+) channels on TRPV4. However, activation of TRPV4 by hypotonic swelling (or GSK1016790A, a selective agonist) or inhibition by the selective antagonist, HC-067047, demonstrated a strong dependency of SK3 and BK-α activation on TRPV4-mediated Ca(2+) influx. Selective inhibition of BK-α channel (Iberiotoxin) or SK3 channel (apamin), thereby depolarizing the cells, further revealed a significant dependency of TRPV4-mediated Ca(2+) influx on activation of both K(+) channels. It is concluded that a synergistic cross-talk exists between the TRPV4 channel and SK3 and BK-α channels to provide a tight functional regulation between the channel groups. This cross-talk may be progressive in nature where the initial TRPV4-mediated Ca(2+) influx would first activate the highly Ca(2+)-sensitive SK3 channel which, in turn, would lead to enhanced Ca(2+) influx and activation of the less Ca(2+)-sensitive BK channel.


Assuntos
Cálcio/metabolismo , Túbulos Renais Coletores/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Apamina/farmacologia , Aquaporina 2/biossíntese , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Túbulos Renais Coletores/citologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Leucina/análogos & derivados , Leucina/farmacologia , Camundongos , Peptídeos/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA