Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Cells ; 12(6)2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36980269

RESUMO

Cantú syndrome (CS) is caused by the gain of function mutations in the ABCC9 and KCNJ8 genes encoding, respectively, for the sulfonylureas receptor type 2 (SUR2) and the inwardly rectifier potassium channel 6.1 (Kir6.1) of the ATP-sensitive potassium (KATP) channels. CS is a multi-organ condition with a cardiovascular phenotype, neuromuscular symptoms, and skeletal malformations. Glibenclamide has been proposed for use in CS, but even in animals, the drug is incompletely effective against severe mutations, including the Kir6.1wt/V65M. Patch-clamp experiments showed that zoledronic acid (ZOL) fully reduced the whole-cell KATP currents in bone calvaria cells from wild type (WT/WT) and heterozygous Kir6.1wt/V65MCS mice, with IC50 for ZOL block < 1 nM in each case. ZOL fully reduced KATP current in excised patches in skeletal muscle fibers in WT/WT and CS mice, with IC50 of 100 nM in each case. Interestingly, KATP currents in the bone of heterozygous SUR2wt/A478V mice were less sensitive to ZOL inhibition, showing an IC50 of ~500 nM and a slope of ~0.3. In homozygous SUR2A478V/A478V cells, ZOL failed to fully inhibit the KATP currents, causing only ~35% inhibition at 100 µM, but was responsive to glibenclamide. ZOL reduced the KATP currents in Kir6.1wt/VMCS mice in both skeletal muscle and bone cells but was not effective in the SUR2[A478V] mice fibers. These data indicate a subunit specificity of ZOL action that is important for appropriate CS therapies.


Assuntos
Músculo Esquelético , Ácido Zoledrônico , Animais , Camundongos , Trifosfato de Adenosina , Modelos Animais de Doenças , Glibureto/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Ácido Zoledrônico/farmacologia , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Receptores de Sulfonilureias/efeitos dos fármacos , Receptores de Sulfonilureias/metabolismo
2.
Invest Ophthalmol Vis Sci ; 63(2): 15, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35129587

RESUMO

Purpose: To evaluate the effect of ATP-sensitive potassium channel openers cromakalim prodrug 1 (CKLP1) and diazoxide on IOP in three independent mouse models of ocular hypertension. Methods: Baseline IOP was measured in TGFß2 overexpression, steroid-induced, and iris dispersion (DBA/2J) ocular hypertension mouse models, followed by once daily eyedrop administration with CKLP1 (5 mM) or diazoxide (5 mM). The IOP was measured in conscious animals with a handheld rebound tonometer. Aqueous humor dynamics were assessed by a constant perfusion method. Effect of treatment on ocular tissues was evaluated by transmission electron microscopy. Results: CKLP1 decreased the IOP by 20% in TGFß2 overexpressing mice (n = 6; P < 0.0001), 24% in steroid-induced ocular hypertensive mice (n = 8; P < 0.0001), and 43% in DBA/2J mice (n = 15; P < 0.0001). Diazoxide decreased the IOP by 32% in mice with steroid-induced ocular hypertension (n = 13; P < 0.0001) and by 41% in DBA/2J mice (n = 4; P = 0.005). An analysis of the aqueous humor dynamics revealed that CKLP1 decreased the episcleral venous pressure by 29% in TGFß2 overexpressing mice (n = 13; P < 0.0001) and by 72% in DBA/2J mice (n = 4 control, 3 treated; P = 0.0002). Diazoxide lowered episcleral venous pressure by 35% in steroid-induced ocular hypertensive mice (n = 3; P = 0.03). Tissue histology and cell morphology appeared normal when compared with controls. Accumulation of extracellular matrix was reduced in CKLP1- and diazoxide-treated eyes in the steroid-induced ocular hypertension model. Conclusions: ATP-sensitive potassium channel openers CKLP1 and diazoxide effectively decreased the IOP in ocular hypertensive animal models by decreasing the episcleral venous pressure, supporting a potential therapeutic application of these agents in ocular hypertension and glaucoma.


Assuntos
Cromakalim/administração & dosagem , Diazóxido/administração & dosagem , Pressão Intraocular/efeitos dos fármacos , Canais KATP/efeitos dos fármacos , Hipertensão Ocular/tratamento farmacológico , Animais , Anti-Hipertensivos/administração & dosagem , Modelos Animais de Doenças , Olho/ultraestrutura , Canais KATP/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Microscopia Eletrônica de Transmissão , Hipertensão Ocular/metabolismo , Hipertensão Ocular/fisiopatologia , Soluções Oftálmicas
3.
Eur J Clin Invest ; 52(2): e13683, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34587304

RESUMO

BACKGROUND: In the present work, we investigated the cardioprotective potential of pyridoxal-5-phosphate (PLP) in old rats as a cofactor of enzymes that synthesize hydrogen sulphide (H2 S). MATERIALS AND METHODS: PLP was administered per os in a dose of 0.7 mg per kg daily for 2 weeks. Rats were divided into three groups (adult, old and old +PLP) of 20 animals. The cardiac mRNA levels of genes encoding H2 S-synthesizing enzymes cystathionine-γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST), uncoupling proteins (UCP3), subunits of ATP-sensitive potassium (KATP ) channels were determined using real-time polymerase chain reaction analysis. We also studied the effect of PLP-administration on the content of H2 S, oxidative stress, the activities of inducible and constitutive NO-synthase (iNOS, cNOS), arginase and nitrate reductase in the heart homogenates as well as cardiac resistance to ischemia-reperfusion in Langendorff-isolated heart model. RESULTS: It was shown that PLP restored mRNA levels of CSE, 3-MST and UCP3 genes, and H2 S content and also significantly increased the expression of SUR2 and Kir6.1 (2.2 and 3.3 times, respectively) in the heart of old rats. PLP significantly reduced the formation of superoxide, malondialdehyde, diene conjugates as well as the activity of iNOS and arginase. PLP significantly increased constitutive synthesis of NO and prevented reperfusion disturbances of the heart function after ischemia. CONCLUSIONS: Thus, PLP-administration in old rats was associated with up-expression of CSE, 3-MST, UCP3 and SUR2 and Kir6.1 subunits of KATP channels, and also increased cNOS activity and reduced oxidative stress and prevented reperfusion dysfunction of the heart in ischemia-reperfusion.


Assuntos
Cardiotônicos/farmacologia , Cistationina gama-Liase/efeitos dos fármacos , Cistationina gama-Liase/fisiologia , Canais KATP/efeitos dos fármacos , Canais KATP/fisiologia , Fosfato de Piridoxal/farmacologia , Sulfurtransferases/efeitos dos fármacos , Sulfurtransferases/fisiologia , Envelhecimento , Animais , Cistationina gama-Liase/genética , Regulação da Expressão Gênica , Coração/efeitos dos fármacos , Canais KATP/genética , Masculino , Ratos , Ratos Wistar , Sulfurtransferases/genética
4.
Pak J Pharm Sci ; 34(5(Supplementary)): 1983-1988, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34836870

RESUMO

Moxifloxacin and gemifloxacin were tested on isolated rabbits' jejunal preparations as little is known about its effects on gastrointestinal tissues. Moxifloxacin and gemifloxacin were tested in concentrations 0.01-10µg/mL for possible effect(s) on isolated rabbits' jejunal preparations. The drugs were applied on spontaneous, on low K+ (20mM)-induced contractions and on high K+ (80mM)-induced contractions. Response was plotted as % of its respective controls. EC50 for Moxifloxacin and Gemifloxacin on spontaneous (without Glibenclamide) contractions are 2.83±0.5µg/mL and 1.11±0.2µg/mL, respectively. Moxifloxacin and Gemifloxacin relaxed the low K+ (20mM) -induced contractions, which were inhibited in presence of Glibenclamide (3µM). Our result indicates that the relaxant activity of Moxifloxacin and Gemifloxacin is mediated possibly through activation of ATP-sensitive potassium channels (KATP). The relaxant effect of Moxifloxacin and Gemifloxacin is predominantly mediated by activation of ATP-Sensitive potassium channels (KATP), which could be cause of one of relaxing mechanisms.


Assuntos
Gemifloxacina/farmacologia , Canais KATP/efeitos dos fármacos , Moxifloxacina/farmacologia , Parassimpatolíticos/farmacologia , Animais , Bioensaio , Feminino , Glibureto/farmacologia , Jejuno/efeitos dos fármacos , Masculino , Contração Muscular/efeitos dos fármacos , Relaxantes Musculares Centrais/farmacologia , Relaxamento Muscular/efeitos dos fármacos , Coelhos
5.
J Ocul Pharmacol Ther ; 37(5): 251-260, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33784195

RESUMO

Purpose: To evaluate pharmacokinetic parameters and ocular hypotensive effects of cromakalim prodrug 1 (CKLP1) in normotensive large animal models. Methods: Optimal CKLP1 concentration was determined by dose response and utilized in short- (5-8 days) and long-term (60 days) evaluation in hound dogs (n = 5) and African Green Monkeys (n = 5). Blood pressure was recorded 3-5 times per week with a tail cuff. Concentrations of CKLP1 and the parent compound levcromakalim were assessed in hound dog plasma and select tissues by LC-MS/MS after bilateral ocular treatment with CKLP1 for 8 days. Pharmacokinetic parameters were calculated from days 1, 4, and 8 data. After necropsy, histology was assessed in 43 tissue samples from each animal. Results: In hound dogs and African Green monkeys, 10 mM CKLP1 (optimal concentration) significantly lowered intraocular pressure (IOP) by 18.9% ± 1.1% and 16.7% ± 6.7%, respectively, compared with control eyes (P < 0.05). During treatment, no significant change in systolic or diastolic blood pressure was observed in either species (P > 0.1). Average values for half-life of CKLP1 was 295.3 ± 140.4 min, Cmax, 10.5 ± 1.6 ng/mL, and area under the concentration vs. time curve (AUClast) 5261.4 ± 918.9 ng·min/mL. For levcromakalim, average values of half-life were 96.2 ± 27 min, Cmax 1.2 ± 0.2 ng/mL, and AUClast 281.2 ± 110.8 ng·min/mL. No significant pathology was identified. Conclusions: CKLP1 lowered IOP in hound dogs and African green monkeys with no effect on systemic blood pressure. Ocular topical treatment of CKLP1 showed excellent tolerability even after extended treatment periods.


Assuntos
Anti-Hipertensivos/farmacocinética , Cromakalim/farmacocinética , Pressão Intraocular/efeitos dos fármacos , Canais KATP/efeitos dos fármacos , Administração Oftálmica , Administração Tópica , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/farmacologia , Área Sob a Curva , Autopsia/métodos , Pressão Sanguínea/efeitos dos fármacos , Chlorocebus aethiops , Cromakalim/administração & dosagem , Cromakalim/farmacologia , Cães , Relação Dose-Resposta a Droga , Feminino , Meia-Vida , Modelos Animais , Primatas , Pró-Fármacos
6.
Cell Rep ; 34(4): 108690, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33503433

RESUMO

Hallmarks of mature ß cells are restricted proliferation and a highly energetic secretory state. Paradoxically, cyclin-dependent kinase 2 (CDK2) is synthesized throughout adulthood, its cytosolic localization raising the likelihood of cell cycle-independent functions. In the absence of any changes in ß cell mass, maturity, or proliferation, genetic deletion of Cdk2 in adult ß cells enhanced insulin secretion from isolated islets and improved glucose tolerance in vivo. At the single ß cell level, CDK2 restricts insulin secretion by increasing KATP conductance, raising the set point for membrane depolarization in response to activation of the phosphoenolpyruvate (PEP) cycle with mitochondrial fuels. In parallel with reduced ß cell recruitment, CDK2 restricts oxidative glucose metabolism while promoting glucose-dependent amplification of insulin secretion. This study provides evidence of essential, non-canonical functions of CDK2 in the secretory pathways of quiescent ß cells.


Assuntos
Linfócitos B/metabolismo , Quinase 2 Dependente de Ciclina/uso terapêutico , Canais KATP/efeitos dos fármacos , Animais , Quinase 2 Dependente de Ciclina/farmacologia , Humanos , Camundongos
7.
Cardiovasc Drugs Ther ; 35(1): 87-101, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33057968

RESUMO

PURPOSE: This study aimed to investigate whether inhibition of glucagon-like peptide-1 (GLP-1) on pressure overload induced cardiac hypertrophy and apoptosis is related to activation of ATP sensitive potassium (KATP) channels. METHODS: Male SD rats were randomly divided into five groups: sham, control (abdominal aortic constriction), GLP-1 analog liraglutide (0.3 mg/kg/twice day), KATP channel blocker glibenclamide (5 mg/kg/day), and liraglutide plus glibenclamide. RESULTS: Relative to the control on week 16, liraglutide upregulated protein and mRNA levels of KATP channel subunits Kir6.2/SUR2 and their expression in the myocardium, vascular smooth muscle, aortic endothelium, and cardiac microvasculature. Consistent with a reduction in aortic wall thickness (61.4 ± 7.6 vs. 75.0 ± 7.6 µm, p < 0.05), liraglutide enhanced maximal aortic endothelium-dependent relaxation in response to acetylcholine (71.9 ± 8.7 vs. 38.6 ± 4.8%, p < 0.05). Along with a reduction in heart to body weight ratio (2.6 ± 0.1 vs. 3.4 ± 0.4, mg/g, p < 0.05) by liraglutide, hypertrophied cardiomyocytes (371.0 ± 34.4 vs. 933.6 ± 156.6 µm2, p < 0.05) and apoptotic cells (17.5 ± 8.2 vs. 44.7 ± 7.9%, p < 0.05) were reduced. Expression of anti-apoptotic protein BCL-2 and contents of myocardial ATP were augmented, and expression of cleaved-caspase 3 and levels of serum Tn-I/-T were reduced. Echocardiography and hemodynamic measurement showed that cardiac systolic function was enhanced as evidenced by increased ejection fraction (88.4 ± 4.8 vs. 73.8 ± 5.1%, p < 0.05) and left ventricular systolic pressure (105.2 ± 10.8 vs. 82.7 ± 7.9 mmHg, p < 0.05), and diastolic function was preserved as shown by a reduction of ventricular end-diastolic pressure (-3.1 ± 2.9 vs. 6.7 ± 2.8 mmHg, p < 0.05). Furthermore, left ventricular internal diameter at end-diastole (5.8 ± 0.5 vs. 7.7 ± 0.6 mm, p < 0.05) and left ventricular internal diameter at end-systole (3.0 ± 0.6 vs. 4.7 ± 0.4 mm, p < 0.05) were improved. Dietary administration of glibenclamide alone did not alter all the parameters measured but significantly blocked liraglutide-exerted cardioprotection. CONCLUSION: Liraglutide ameliorates cardiac hypertrophy and apoptosis, potentially via activating KATP channel-mediated signaling pathway. These data suggest that liraglutide might be considered as an adjuvant therapy to treat patients with heart failure.


Assuntos
Apoptose/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glibureto/farmacologia , Canais KATP/efeitos dos fármacos , Liraglutida/farmacologia , Animais , Cardiomegalia , Quimioterapia Combinada , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
8.
Int J Neurosci ; 131(10): 1004-1011, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32408781

RESUMO

OBJECTIVE: The leukotrienes are inflammatory mediators. In the present study, the analgesic role of local montelukast, a cysteinyl leukotriene receptor antagonist, and the possible involvement of L-arginine/NO/cGMP/KATP channel pathway and PPARγ receptors was assessed in the formalin test in rats. METHODS AND RESULTS: The local administration of montelukast into the hind paw produced dose-related analgesia during both phases of the formalin test. Furthermore, pre-treatment with L-NAME, methylene blue, and glibenclamide prevented montelukast (10 µg/paw)-induced antinociception in both early and late phases of the test. Moreover, the local L-arginine and diazoxide before the sub-effective dose of montelukast (3 µg/paw) produced an analgesic effect. Also, local GW-9662 blocked antinociception induced by montelukast plus pioglitazone (10 µg/paw). CONCLUSION: In conclusion, montelukast produced peripheral analgesia through PPARγ receptors and activation of the L-arginine/NO/cGMP/KATP channel pathway, with potential for a new topical analgesic drug.


Assuntos
Acetatos/farmacologia , Analgesia , Analgésicos/farmacologia , Arginina/efeitos dos fármacos , GMP Cíclico/metabolismo , Ciclopropanos/farmacologia , Canais KATP/efeitos dos fármacos , Antagonistas de Leucotrienos/farmacologia , Óxido Nítrico/metabolismo , Dor Nociceptiva/tratamento farmacológico , PPAR gama/efeitos dos fármacos , Quinolinas/farmacologia , Sulfetos/farmacologia , Acetatos/administração & dosagem , Analgésicos/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Ciclopropanos/administração & dosagem , Modelos Animais de Doenças , Antagonistas de Leucotrienos/administração & dosagem , Masculino , Quinolinas/administração & dosagem , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Sulfetos/administração & dosagem
9.
J Cereb Blood Flow Metab ; 41(6): 1328-1337, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33028147

RESUMO

Glibenclamide inhibits sulfonylurea receptor (SUR), which regulates several ion channels including SUR1-transient receptor potential melastatin 4 (SUR1-TRPM4) channel and ATP-sensitive potassium (KATP) channel. Stroke upregulates SURl-TRPM4 channel, which causes a rapid edema formation and brain swelling. Glibenclamide may antagonize the formation of cerebral edema during stroke. Preclinical studies showed that glibenclamide inhibits KATP channel-induced vasodilation without altering the basal vascular tone. The in vivo human cerebrovascular effects of glibenclamide have not previously been investigated.In a randomized, double-blind, placebo-controlled, three-way cross-over study, we used advanced 3 T MRI methods to investigate the effects of glibenclamide and KATP channel opener levcromakalim on mean global cerebral blood flow (CBF) and intra- and extracranial artery circumferences in 15 healthy volunteers. Glibenclamide administration did not alter the mean global CBF and the basal vascular tone. Following levcromakalim infusion, we observed a 14% increase of the mean global CBF and an 8% increase of middle cerebral artery (MCA) circumference, and glibenclamide did not attenuate levcromakalim-induced vascular changes. Collectively, the findings demonstrate the vital role of KATP channels in cerebrovascular hemodynamic and indicate that glibenclamide does not inhibit the protective effects of KATP channel activation during hypoxia and ischemia-induced brain injury.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Cromakalim/farmacologia , Glibureto/farmacologia , Canais KATP/metabolismo , Adulto , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Estudos Cross-Over , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Canais KATP/efeitos dos fármacos , Imageamento por Ressonância Magnética/métodos , Masculino , Adulto Jovem
10.
Toxicol Lett ; 332: 164-170, 2020 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-32659473

RESUMO

Manganese (Mn) is an environmental pollutant having a toxic effect on Parkinson's disease, with significant damage seen in the neurons of basal ganglia. Hence, Mn pollution is a public health concern. A Sprague-Dawley rat model was used to determine the damage to basal nuclei, and the effect of Mn intake was detected using the Morris water maze test and transmission electron microscopy. The SH-SY5Y cell line was exposed to Mn, and downstream signaling was assessed to determine the mechanism of toxicity. Mn exposure injured neurons, repressing GABAAR receptors and inducing GABABR receptors. The synergistic effect of the GABABR receptor and Kir6.1-SUR1 or Kir6.2-SUR1 was found to be one of the potential factors for the secretion of α-synuclein. The accumulation of α-synuclein regulated downstream factors calmodulin (CAM) cAMP response element-binding protein (CREB), thereby impairing learning and memory. Other genes downstream of CREB, rather than the feedback regulation of CREB, and brain-derived neurotrophic factor might also be involved.


Assuntos
Canais KATP/efeitos dos fármacos , Intoxicação por Manganês/metabolismo , Receptores de GABA/efeitos dos fármacos , alfa-Sinucleína/metabolismo , Animais , Gânglios da Base/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Masculino , Intoxicação por Manganês/psicologia , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/psicologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-B/efeitos dos fármacos
11.
Biomed Pharmacother ; 127: 110210, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32407991

RESUMO

Paclitaxel-induced neuropathic pain (PINP) is a dose-limiting side effect that largely affects the patient's quality of life and may limit the use of the drug as a chemotherapeutic agent for treating metastatic breast cancer and other solid tumors. Recently, a putative role for the gaseous mediator hydrogen sulfide (H2S) in nociception modulation has been suggested. The aim of the present study was to investigate the potential efficacy of the slow release H2S donor GYY4137 to alleviate and prevent PINP. Female BALB/c mice that were intraperitoneally (i.p.) injected with paclitaxel (2 mg/kg) for 5 consecutive days developed thermal hyperalgesia, cold and mechanical allodynia and had reduced of H2S, generation in the spinal cord and paw skin. Treatment of mice with established thermal hyperalgesia with GYY4137 or the analgesic positive control drug gabapentin produced antihyperalgesic activities. The antihyperalgesic activity of GYY4137 was antagonized by the ATP sensitive potassium channels (KATP channels) blocker glibenclamide. Co-treatment with GYY4137 and paclitaxel prevented the paclitaxel-induced decrease in H2S, generation as well as the paclitaxel-induced thermal hyperalgesia, cold allodynia and mechanical allodynia. GYY4137 enhanced paclitaxel's anti-proliferative effects against the breast cancer cell line MCF-7. The present results suggest that GYY4137 alleviates paclitaxel-induced thermal hyperalgesia, via KATP channels. GYY4137 prevents PINP possibly by blocking the paclitaxel-induced reduction in the generation of H2S, in the tissues, while enhancing the anti-cancer activity of paclitaxel, and therefore warrants further research as a candidate for prevention of PINP in clinical settings.


Assuntos
Hiperalgesia/prevenção & controle , Morfolinas/farmacologia , Neuralgia/prevenção & controle , Compostos Organotiofosforados/farmacologia , Paclitaxel/toxicidade , Analgésicos/farmacologia , Animais , Antineoplásicos Fitogênicos/toxicidade , Modelos Animais de Doenças , Feminino , Gabapentina/farmacologia , Sulfeto de Hidrogênio/metabolismo , Hiperalgesia/induzido quimicamente , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neuralgia/induzido quimicamente
12.
J Cereb Blood Flow Metab ; 40(10): 1987-1996, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31594422

RESUMO

We investigated the effects of sulforaphane (SFN), an isothiocyanate from cruciferous vegetables, in the regulation of cerebral blood flow using cranial windows in newborn pigs. SFN administered topically (10 µM-1 mM) or systemically (0.4 mg/kg ip) caused immediate and sustained dilation of pial arterioles concomitantly with elevated H2S in periarachnoid cortical cerebrospinal fluid. H2S is a potent vasodilator of cerebral arterioles. SFN is not a H2S donor but it acts via stimulating H2S generation in the brain catalyzed by cystathionine γ-lyase (CSE) and cystathionine ß-synthase (CBS). CSE/CBS inhibitors propargylglycine, ß-cyano-L-alanine, and aminooxyacetic acid blocked brain H2S generation and cerebral vasodilation caused by SFN. The SFN-elicited vasodilation requires activation of potassium channels in cerebral arterioles. The inhibitors of KATP and BK channels glibenclamide, paxilline, and iberiotoxin blocked the vasodilator effects of topical and systemic SFN, supporting the concept that H2S is the mediator of the vasodilator properties of SFN in cerebral circulation. Overall, we provide first evidence that SFN is a brain permeable compound that increases cerebral blood flow via a non-genomic mechanism that is mediated via activation of CSE/CBS-catalyzed H2S formation in neurovascular cells followed by H2S-induced activation of KATP and BK channels in arteriolar smooth muscle.


Assuntos
Arteríolas/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Sulfeto de Hidrogênio/metabolismo , Isotiocianatos/farmacologia , Canais KATP/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Músculo Liso Vascular/metabolismo , Vasodilatadores/farmacologia , Animais , Animais Recém-Nascidos , Arteríolas/efeitos dos fármacos , Encéfalo/metabolismo , Cistationina beta-Sintase/antagonistas & inibidores , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/antagonistas & inibidores , Cistationina gama-Liase/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Isotiocianatos/antagonistas & inibidores , Canais KATP/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Sulfóxidos , Suínos
13.
Fundam Clin Pharmacol ; 34(1): 148-155, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31197879

RESUMO

Melatonin is a neurohormone secreted principally by the pineal gland. This molecule has various pharmacological properties including improving immune system, prevent cancer, anti-aging, and anti-oxidant effects. The anticonvulsant effects of melatonin have been proved by previous studies. Adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels are considered as an important target in the seizure modulation. The aim of the present study was to investigate the anticonvulsant effect of melatonin in pentylenetetrazole (PTZ)-induced seizures in mice, focusing on its ability to regulate KATP channels. Acute intraperitoneal administration of melatonin (40 and 80 mg/kg) increased clonic seizure threshold induced by intravenous administration of PTZ. Melatonin (40 and 80 mg/kg) increased the latency of clonic seizure and reduced its frequency in mice receiving an intraperitoneal injection of PTZ. Administration of glibenclamide, a KATP channels blocker, before intravenous injection of PTZ reduced melatonin anticonvulsant effect. Diazoxide and cromakalim, as KATP channels openers, increased antiseizure effect of melatonin in PTZ model of seizures. These findings suggest that the antiseizure effect of melatonin probably is gained through increasing the opening of KATP channels.


Assuntos
Anticonvulsivantes/farmacologia , Canais KATP/efeitos dos fármacos , Melatonina/farmacologia , Convulsões/tratamento farmacológico , Animais , Anticonvulsivantes/administração & dosagem , Cromakalim/farmacologia , Diazóxido/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glibureto/farmacologia , Injeções Intraperitoneais , Canais KATP/metabolismo , Masculino , Melatonina/administração & dosagem , Camundongos , Pentilenotetrazol , Convulsões/fisiopatologia
14.
Indian J Pharmacol ; 51(5): 296-301, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31831918

RESUMO

Nicorandil is a well-known antianginal agent, which has been recommended as one of the second-line treatments for chronic stable angina as justified by the European guidelines. It shows an efficacy equivalent to that of classic antianginal agents. Nicorandil has also been applied clinically in various cardiovascular diseases such as variant or unstable angina and reperfusion-induced damage following coronary angioplasty or thrombolysis. Different mechanisms have been involved in the protective effects of nicorandil in various diseases through either opening of adenosine triphosphate-sensitive potassium (KATP) channel or donation of nitric oxide (NO). The predominance or participation of any of these proposed mechanisms depends on the dose of nicorandil used, the location of diseased conditions, and if this mechanism is still functioning or not. The protection afforded by nicorandil has been shown to be mainly attributed to KATP channel opening in experimental models of myocardial and pulmonary fibrosis as well as renal injury or glomerulonephritis, whereas NO donation predominates as a mechanism of protection in hepatic fibrosis and inflammatory bowel diseases. Therefore, in different diseased conditions, it is important to know which mechanism plays the major role in nicorandil-induced curative or protective effects. This can bring new insights into the proper use of selected medication and its recommended dose for targeting certain disease.


Assuntos
Antiarrítmicos/administração & dosagem , Nicorandil/administração & dosagem , Angina Estável/tratamento farmacológico , Animais , Antiarrítmicos/farmacologia , Relação Dose-Resposta a Droga , Humanos , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Nicorandil/farmacologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/administração & dosagem , Doadores de Óxido Nítrico/farmacologia
15.
J Clin Neurosci ; 63: 196-201, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30795876

RESUMO

The present study was examined the antioxidant effect of tamoxifen, a synthetic non-steroidal antiestrogen, on cromakalim or nicorandil (ATP-sensitive K+ (KATP) channels opener)-enhanced hydroxyl radical (OH) generation induced by 1-methyl-4-phenylpyridinium ion (MPP+) in extracellular fluid of rat striatum. Rats were anesthetized, and sodium salicylate in Ringer's solution (0.5 mM or 0.5 nmol/µl/min) was infused through a microdialysis probe to detect the generation of OH as reflected by the non-enzymatic formation of 2,3-dihydroxybenzoic acid (DHBA) in the striatum. Cromakalim (100 µM) or nicorandil (1 mM) enhanced the formation of OH trapped as DHBA induced by MPP+ (5 mM). Concomitantly, these drugs enhanced dopamine (DA) efflux induced by MPP+. Tamoxifen (30 µM) significantly decreased the level of DA enhanced by cromakalim or nicorandil. Tamoxifen suppressed DHBA formation induced by MPP+ and cromakalim or nicorandil. When iron(II) was administered to cromakalim treated animals, a marked elevation of DHBA was observed, compared with the tamoxifen-treated rats These results indicated that the effects of tamoxifen on opening of KATP channels enhances OH generation in the extracellular space of striatum during of DA release by MPP+. These results indicated that estrogen protects against neuronal degeneration by as an anti-oxidant.


Assuntos
Antioxidantes/farmacologia , Corpo Estriado/efeitos dos fármacos , Radical Hidroxila/metabolismo , Canais KATP/efeitos dos fármacos , Tamoxifeno/farmacologia , 1-Metil-4-fenilpiridínio/metabolismo , Animais , Corpo Estriado/metabolismo , Dopamina/metabolismo , Masculino , Ratos , Ratos Wistar
16.
BMC Res Notes ; 11(1): 614, 2018 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-30144824

RESUMO

OBJECTIVE: Memantine, a drug for Alzheimer's disease, is considered to suppress excessive stimulation of N-methyl-D-aspartic acid receptors and to prevent neuronal death. However, a recent report indicated that the neuronal KATP channel also can become a target of memantine. The KATP channel is a key regulator of insulin secretion in pancreatic ß cells. Therefore, if memantine could inhibit the KATP channel in pancreatic ß cells, it would be an effective drug for both Alzheimer's disease and diabetes. However, there is no report on the effect of memantine on the KATP channel in pancreatic ß cells. Therefore, we investigated whether memantine affect the blood glucose level, insulin secretion and KATP channel activity in pancreatic ß cells. RESULTS: An intraperitoneal glucose tolerance test was performed with or without memantine (1 mg/kg) injection in intact mice. Insulin secretion from isolated islets was measured under low (2 mM) and high (20 mM) glucose concentrations with or without memantine (1 µM). The effect of memantine (1 µM) on KATP channel currents in isolated pancreatic ß cells was recorded using the whole-cell patch-clamp technique. Memantine had no effect on the blood glucose level, insulin secretion from isolated islets or KATP channel current in pancreatic ß cells.


Assuntos
Dopaminérgicos/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Canais KATP/efeitos dos fármacos , Memantina/farmacologia , Animais , Glucose , Insulina , Ilhotas Pancreáticas , Japão , Canais KATP/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
17.
Semin Perinatol ; 42(4): 221-227, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29880312

RESUMO

Forty years ago, non-steroidal anti-inflammatory drugs were first reported to decrease systemic prostaglandin levels and promote ductus arteriosus (DA) closure. And yet, prolonged patency of the DA (PDA) remains a significant clinical problem, complicated by imperfect therapies and wide variations in treatment strategy. There are few pharmacology-based tools available for treating PDA (indomethacin, ibuprofen, and acetaminophen), or for maintaining DA patency (PGE1) as is needed to facilitate corrective surgery for ductus-dependent congenital heart defects. Unfortunately, all of these treatments are inefficient and are associated with concerning adverse effects. This review highlights novel potential DA drug targets that may expand our therapeutic repertoire beyond the prostaglandin pathway.


Assuntos
Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Canais KATP/efeitos dos fármacos , Grau de Desobstrução Vascular/efeitos dos fármacos , Acetaminofen/farmacologia , Acetaminofen/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Canal Arterial/fisiopatologia , Permeabilidade do Canal Arterial/fisiopatologia , Humanos , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Indometacina/farmacologia , Indometacina/uso terapêutico , Recém-Nascido , Recém-Nascido Prematuro , Canais KATP/fisiologia , Modelos Animais , Estudo de Prova de Conceito , Grau de Desobstrução Vascular/fisiologia
18.
J Neurosci ; 38(22): 5053-5066, 2018 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-29703787

RESUMO

The onset of the headache phase during attacks of migraine with aura, which occur in ∼30% of migraineurs, is believed to involve cortical spreading depression (CSD) and the ensuing activation and sensitization of primary afferent neurons that innervate the intracranial meninges, and their related large vessels. The mechanism by which CSD enhances the activity and mechanosensitivity of meningeal afferents remains poorly understood, but may involve cortical metabolic perturbations. We used extracellular single-unit recording of meningeal afferent activity and monitored changes in cortical blood flow and tissue partial pressure of oxygen (tpO2) in anesthetized male rats to test whether the prolonged cortical hypoperfusion and reduction in tissue oxygenation that occur in the wake of CSD contribute to meningeal nociception. Suppression of CSD-evoked cortical hypoperfusion with the cyclooxygenase inhibitor naproxen blocked the reduction in cortical tpO2, but had no effect on the activation of meningeal afferents. Naproxen, however, distinctly prevented CSD-induced afferent mechanical sensitization. Counteracting the CSD-evoked persistent hypoperfusion and reduced tpO2 by preemptively increasing cortical blood flow using the ATP-sensitive potassium [K(ATP)] channel opener levcromakalim did not inhibit the sensitization of meningeal afferents, but prevented their activation. Our data show that the cortical hypoperfusion and reduction in tpO2 that occur in the wake of CSD can be dissociated from the activation and mechanical sensitization of meningeal afferent responses, suggesting that the metabolic changes do not contribute directly to these neuronal nociceptive responses.SIGNIFICANCE STATEMENT Cortical spreading depression (CSD)-evoked activation and mechanical sensitization of meningeal afferents is thought to mediate the headache phase in migraine with aura. We report that blocking the CSD-evoked cortical hypoperfusion and reduced tissue partial pressure of oxygen by cyclooxygenase inhibition is associated with the inhibition of the afferent sensitization, but not their activation. Normalization of these CSD-evoked metabolic perturbations by activating K(ATP) channels is, however, associated with the inhibition of afferent activation but not sensitization. These results question the contribution of cortical metabolic perturbations to the triggering mechanism underlying meningeal nociception and the ensuing headache in migraine with aura, further point to distinct mechanisms underlying the activation and sensitization of meningeal afferents in migraine, and highlight the need to target both processes for an effective migraine therapy.


Assuntos
Vias Aferentes/fisiopatologia , Depressão Alastrante da Atividade Elétrica Cortical , Meninges/fisiopatologia , Transtornos de Enxaqueca/fisiopatologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Encéfalo/diagnóstico por imagem , Circulação Cerebrovascular , Cromakalim/farmacologia , Canais KATP/agonistas , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Masculino , Meninges/diagnóstico por imagem , Transtornos de Enxaqueca/diagnóstico por imagem , Naproxeno/farmacologia , Nociceptividade/efeitos dos fármacos , Nociceptores , Consumo de Oxigênio , Ratos , Ratos Sprague-Dawley , Ultrassonografia Doppler
19.
Behav Pharmacol ; 29(4): 336-343, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29239973

RESUMO

Studies have shown that hydrogen sulfide (H2S) exerts a neuroprotective effect and may have a therapeutic value for treating neurodegenerative diseases including Parkinson's disease. However, little is known about the mechanisms underlying the neuroprotective activity of H2S in vivo. Here, we evaluated the effect of glibenclamide, an ATP-sensitive potassium channel blocker, on the neuroprotective activity of H2S in the 6-hydroxydopamine (6-OHDA) animal model of Parkinson's disease. 6-OHDA was administered by stereotaxic surgery into the medial forebrain bundle. Sodium hydrosulfate (NaHS, 3 and 5.6 mg/kg), as a donor of H2S, alone or in combination with glibenclamide (5 mg/kg), was daily injected for 7 days starting 1-2 h before the stereotaxic surgery. After an apomorphine-induced rotational test, the number of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta was determined by immunofluorescence. The striatal dopamine level and oxidative stress markers were also measured in brain homogenates. Pretreatment with NaHS significantly attenuated 6-OHDA-induced motor asymmetry in the rotational test. Histological and biochemical evaluations demonstrated that NaHS, especially at high dose, increased the survival of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta and reduced the decreasing effect of 6-OHDA on striatal dopamine levels. However, co-administration of glibenclamide reversed the antiparkinsonian and neuroprotective effects of NaHS. However, glibenclamide did not change the reducing effect of NaHS on 6-OHDA-induced overproduction of malondialdehyde. Our data show that ATP-sensitive potassium channels are involved in the antiparkinsonian and neuroprotective effects of H2S in the 6-OHDA animal model of Parkinson's disease.


Assuntos
Sulfeto de Hidrogênio/farmacologia , Canais KATP/fisiologia , Doença de Parkinson/tratamento farmacológico , Trifosfato de Adenosina/fisiologia , Animais , Apomorfina/farmacologia , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina , Agonistas de Dopamina/farmacologia , Sulfeto de Hidrogênio/uso terapêutico , Canais KATP/efeitos dos fármacos , Masculino , Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Wistar , Substância Negra/efeitos dos fármacos
20.
J Neuroinflammation ; 14(1): 228, 2017 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-29178967

RESUMO

BACKGROUND: Long-term use of morphine induces analgesic tolerance, which limits its clinical efficacy. Evidence indicated morphine-evoked neuroinflammation mediated by toll-like receptor 4 (TLR4) - NOD-like receptor protein 3 (NLRP3) inflammasome was important for morphine tolerance. In our study, we investigated whether other existing alternative pathways caused morphine-induced activation of TLR4 in microglia. We focused on heat shock protein 70 (HSP70), a damage-associated molecular pattern (DAMP), which was released from various cells upon stimulations under the control of KATP channel and bound with TLR4-inducing inflammation. Glibenclamide, a classic KATP channel blocker, can improve neuroinflammation by inhibiting the activation of NLRP3 inflammasome. Our present study investigated the effect and possible mechanism of glibenclamide in improving morphine tolerance via its specific inhibition on the release of HSP70 and activation of NLRP3 inflammasome induced by morphine. METHODS: CD-1 mice were used for tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 and neural cell line SH-SY5Y were used to investigate the pharmacological effects and the mechanism of glibenclamide on morphine-induced neuroinflammation. The activation of microglia was accessed by immunofluorescence staining. Neuroinflammation-related cytokines were measured by western blot and real-time PCR. The level of HSP70 and related signaling pathway were evaluated by western blot and immunofluorescence staining. RESULTS: Morphine induced the release of HSP70 from neurons. The released HSP70 activated microglia and triggered TLR4-mediated inflammatory response, leading to the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) p65 and the activation of NLRP3 inflammasome. Moreover, anti-HSP70 neutralizing antibody partly attenuated chronic morphine tolerance. The secretion of HSP70 was under the control of MOR/AKT/KATP/ERK signal pathway. Glibenclamide as a classic KATP channel blocker markedly inhibited the release of HSP70 induced by morphine and suppressed HSP70-TLR4-NLRP3 inflammasome-mediated neuroinflammation, which consequently attenuated morphine tolerance. CONCLUSIONS: Our study indicated that morphine-induced extracellular HSP70 was an alternative way for the activation of TLR4-NLRP3 in analgesic tolerance. The release of HSP70 was regulated by MOR/AKT/KATP/ERK pathway. Our study suggested a promising target, KATP channel and a new leading compound, glibenclamide, for treating morphine tolerance.


Assuntos
Tolerância a Medicamentos/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Canais KATP/antagonistas & inibidores , Morfina , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptor 4 Toll-Like/imunologia , Animais , Glibureto/farmacologia , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Canais KATP/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA