Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 524(4): 791-797, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32019676

RESUMO

Increased granulosa cell (GC) proliferation may contribute to abnormal folliculogenesis in patients with polycystic ovary syndrome (PCOS), which affects approximately 10% reproductive aged women. However, the mechanisms underlying increased GC proliferation in PCOS remain incompletely understood. In this study, we identified miR-3940-5p as a hub miRNA in GC from PCOS using weighted gene co-expression network analysis (WGCNA), and real-time polymerase chain reaction (RT-PCR) analysis confirmed that miR-3940-5p was significantly increased in GC from PCOS. Enrichment analysis of predicted target genes of miR-3940-5p indicated potential roles of miR-3940-5p in follicular development and cell proliferation regulation. Consistently, functional study confirmed that miR-3940-5p overexpression promoted granulosa cell proliferation. Integrated analysis of mRNA expression profiling data and predicted target genes of miR-3940-5p identified potassium voltage-gated channel subfamily A member 5 (KCNA5) as a potential target of miR-3940-5p, and was validated by luciferase reporter assay. Finally, functional analysis suggested that miR-3940-5p promoted GC proliferation in a KCNA5 dependent manner. In conclusion, miR-3940-5p was a hub miRNA upregulated in GC from PCOS, and promoted GC proliferation by targeting KCNA5.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células da Granulosa/metabolismo , Canal de Potássio Kv1.5/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Síndrome do Ovário Policístico/genética , Adulto , Antagomirs/genética , Antagomirs/metabolismo , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Genes Reporter , Células da Granulosa/patologia , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/metabolismo , Luciferases/genética , Luciferases/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Anotação de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
2.
Br J Pharmacol ; 171(1): 92-106, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24102184

RESUMO

BACKGROUND AND PURPOSE: Atrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with an increased risk for stroke, heart failure and cardiovascular-related mortality. Candidate targets for anti-AF drugs include a potassium channel K(v)1.5, and the ionic currents I(KACh) and late I(Na), along with increased oxidative stress and activation of NFAT-mediated gene transcription. As pharmacological management of AF is currently suboptimal, we have designed and characterized a multifunctional small molecule, compound 1 (C1), to target these ion channels and pathways. EXPERIMENTAL APPROACH: We made whole-cell patch-clamp recordings of recombinant ion channels, human atrial I(Kur), rat atrial I(KACh), cellular recordings of contractility and calcium transient measurements in tsA201 cells, human atrial samples and rat myocytes. We also used a model of inducible AF in dogs. KEY RESULTS: C1 inhibited human peak and late K(v)1.5 currents, frequency-dependently, with IC50 of 0.36 and 0.11 µmol·L(-1) respectively. C1 inhibited I(KACh)(IC50 of 1.9 µmol·L(-1)) and the Na(v)1.5 sodium channel current (IC50s of 3 and 1 µmol·L(-1) for peak and late components respectively). C1 (1 µmol·L(-1)) significantly delayed contractile and calcium dysfunction in rat ventricular myocytes treated with 3 nmol·L(-1) sea anemone toxin (ATX-II). C1 weakly inhibited the hERG channel and maintained antioxidant and NFAT-inhibitory properties comparable to the parent molecule, resveratrol. In a model of inducible AF in conscious dogs, C1 (1 mg·kg(-1)) reduced the average and total AF duration. CONCLUSION AND IMPLICATIONS: C1 behaved as a promising multifunctional small molecule targeting a number of key pathways involved in AF.


Assuntos
Antiarrítmicos/farmacologia , Fibrilação Atrial/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Estilbenos/farmacologia , Potenciais de Ação , Adulto , Idoso , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Acoplamento Excitação-Contração/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Células HEK293 , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Masculino , Pessoa de Meia-Idade , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Resveratrol , Bloqueadores dos Canais de Sódio/farmacologia , Transfecção
3.
J Lipid Res ; 54(1): 34-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23099443

RESUMO

Cholesterol-metabolism-associated molecules, including scavenger receptor class A (SR-A), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), CD36, ACAT1, ABCA1, ABCG1, and scavenger receptor class B type I, can modulate cholesterol metabolism in the transformation from macrophages to foam cells. Voltage-gated potassium channel Kv1.3 has increasingly been demonstrated to play an important role in the modulation of macrophage function. Here, we investigate the role of Kv1.3 in modulating cholesterol-metabolism-associated molecules in human acute monocytic leukemia cell-derived macrophages (THP-1 macrophages) and human monocyte-derived macrophages exposed to oxidized LDL (ox-LDL). Human Kv1.3 and Kv1.5 channels (hKv1.3 and hKv1.5) are expressed in macrophages and form a heteromultimeric channel. The hKv1.3-E314 antibody that we had generated as a specific hKv1.3 blocker inhibited outward delayed rectifier potassium currents, whereas the hKv1.5-E313 antibody that we had generated as a specific hKv1.5 blocker failed. Accordingly, the hKv1.3-E314 antibody reduced percentage of cholesterol ester and enhanced apoA-I-mediated cholesterol efflux in THP-1 macrophages and human monocyte-derived macrophages exposed to ox-LDL. The hKv1.3-E314 antibody downregulated SR-A, LOX-1, and ACAT1 expression and upregulated ABCA1 expression in THP-1 macrophages and human monocyte-derived macrophages. Our results reveal that specific Kv1.3 blockade represents a novel strategy modulating cholesterol metabolism in macrophages, which benefits the treatment of atherosclerotic lesions.


Assuntos
Especificidade de Anticorpos , Colesterol/metabolismo , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/imunologia , Lipoproteínas LDL/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Acetil-CoA C-Acetiltransferase/metabolismo , Apolipoproteína A-I/metabolismo , Transporte Biológico/efeitos dos fármacos , Antígenos CD36/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/imunologia , Canal de Potássio Kv1.5/metabolismo , Macrófagos/citologia , Monócitos/citologia , Potássio/metabolismo , Receptores Depuradores Classe A/metabolismo , Receptores Depuradores Classe E/metabolismo
4.
Curr Med Chem ; 19(31): 5394-404, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22856664

RESUMO

We have previously shown that the mitochondrial potassium channel Kv1.3 (mtKv1.3) in T lymphocytes is a novel target of Bax. Mutation of Bax at lysine 128 (BaxK128E) abrogates its inhibitory effects on mtKv1.3 and prevents apoptosis. The importance of mtKv1.3 inhibition was underscored by the finding that membrane-permeant Kv1.3 inhibitors induced Bax/Bak-independent cell death and reduced the volume of an mtKv1.3-expressing tumor by 90% in a mouse model. However, the possible involvement of other Kv channels in apoptosis has not been clarified. Here we report that, like Kv1.3, Kv1.1 and Kv1.5 also interact with Bax. Transfection of Kvdeficient lymphocytes with Kv1.1 restores sensitivity to cell death in apoptosis-resistant CTLL-2 lymphocytes. SiRNA down-regulation of Kv1.3 and Kv1.5 expression in macrophages confers resistance to apoptosis. We further report that J774 macrophages express Kv1.3 and Kv1.5 in their mitochondria and that inhibition of both channels with specific membrane-permeant drugs can efficiently induce apoptosis in a macrophage cell line. Thus, our results indicate that the mechanism proposed for Kv1.3 can be extended to other Kv channels and suggest that membrane-permeant drugs may be a novel pharmacological tool for inducing apoptosis in macrophages, important players in the immune system. This result could be exploited for the depletion of tumor-associated macrophages, which have been shown to foster tumor growth.


Assuntos
Apoptose , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Macrófagos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estaurosporina/farmacologia , Transfecção , Proteína X Associada a bcl-2/metabolismo
5.
Biochem Pharmacol ; 84(4): 498-506, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22583923

RESUMO

The natural flavones and polymethylflavone have been reported to have cardiovascular protective effects. In the present study, we determined whether quecertin, apigenin and their methylated compounds (3,7,3',4'-tetramethylquecertin, 3,5,7,3',4'-pentamethylquecertin, 7,4'-dimethylapigenin, and 5,7,4'-trimethylapigenin) would block the atrial specific potassium channel hKv1.5 using a whole-cell patch voltage-clamp technique. We found that only trimethylapigenin showed a strong inhibitory effect on hKv1.5 channel current. This compound suppressed hKv1.5 current in HEK 293 cell line (IC50=6.4 µM), and the ultra-rapid delayed rectify K⁺ current I(Kur) in human atrial myocytes (IC50=8.0 µM) by binding to the open channels and showed a use- and frequency-dependent manner. In addition, trimethylapigenin decreased transient outward potassium current (I(to)) in human atrial myocytes, inhibited acetylcholine-activated K⁺ current (IC50=6.8µM) in rat atrial myocytes. Interestingly, trimethylapigenin had a weak inhibition of hERG channel current. Our results indicate that trimethyapigenin significantly inhibits the atrial potassium currents hKv1.5/I(Kur) and I(KACh), which suggests that trimethylapigenin may be a potential candidate for anti-atrial fibrillation.


Assuntos
Apigenina/farmacologia , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Feminino , Células HEK293 , Átrios do Coração/citologia , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/fisiologia , Masculino , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Quercetina/análogos & derivados , Quercetina/farmacologia , Ratos , Ratos Sprague-Dawley
6.
Eur J Pharmacol ; 649(1-3): 14-22, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20826138

RESUMO

Repolarization of cardiac action potentials is regulated by several types of K(+) currents. The present study examined the presence and functional significance of rapid delayed rectifier (I(Kr)) in left and right atrial myocytes of mouse heart, using whole-cell patch-clamp method. The functional role of ultrarapid delayed rectifier (I(Kur)) in the repolarization was also examined by blocking with 4-aminopyridine (50 µM). The presence of I(Kr) was detected in left and right atrial myocytes as an E-4031 (5 µM)-sensitive current that exhibited relatively rapid activation during depolarization and half activation voltage of -17.5 and -17.4 mV for left and right atrial myocytes, respectively. The current density of I(Kr) was similar between left and right atria. The prolongation of action potential measured at 50% repolarization evoked by 4-aminopyridine was significantly larger in left than in right atrium, which appears to be consistent with the larger amplitude of I(Kur) in left atrium. On the other hand, the prolongation of action potential measured at 90% repolarization caused by E-4031 was significantly larger in right than in left atrium. The longer action potential of right atrium, which may result at least partly from smaller amplitude of I(Kur), is likely to enhance the functional significance of I(Kr) in repolarization process of right atrium, despite of similar magnitude of I(Kr) in left and right atria. Our data thus identifies I(Kr) in mouse atria and indicates the presence of functional interaction between I(Kr) and I(Kur) that potentially contributes to repolarization heterogeneity in left and right atria of mouse heart.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Função Atrial/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/fisiologia , Canal de Potássio Kv1.5/fisiologia , Aminopiridinas/farmacologia , Animais , Células Cultivadas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Cinética , Canal de Potássio Kv1.5/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Especificidade de Órgãos , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia
7.
Br J Pharmacol ; 161(4): 872-84, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20860665

RESUMO

BACKGROUND AND PURPOSE: N-(6-aminohexyl)-5-chloro-1-naphthalene sulphonamide (W-7) is a well-known calmodulin inhibitor used to study calmodulin regulation of intracellular Ca(2+) signalling-related process. Here, we have determined whether W-7 would inhibit human ether gene (hERG or K(v) 11.1) potassium channels, hK(v) 1.5 channels or hK(IR) 2.1 channels expressed in human embryonic kidney (HEK) 293 cells. EXPERIMENTAL APPROACH: The hERG channel current, hK(v) 1.5 channel current or hK(IR) 2.1 channel current was recorded with a whole-cell patch clamp technique. KEY RESULTS: It was found that the calmodulin inhibitor W-7 blocked hERG, hK(v) 1.5 and hK(IR) 2.1 channels. W-7 decreased the hERG current (I(hERG) ) in a concentration-dependent manner (IC(50) : 3.5 µM), and the inhibition was more significant at depolarization potentials between +10 and +60 mV. The hERG mutations in the S6 region Y652A and F656V, and in the pore helix S631A, had the IC(50) s of 5.5, 9.8 and 25.4 µM respectively. In addition, the compound inhibited hK(v) 1.5 and hK(IR) 2.1 channels with IC(50) s of 6.5 and 13.4 µM respectively. CONCLUSION AND IMPLICATIONS: These results indicate that the calmodulin inhibitor W-7 exerts a direct channel-blocking effect on hERG, hK(v) 1.5 and hK(IR) 2.1 channels stably expressed in HEK 293 cells. Caution should be taken in the interpretation of calmodulin regulation of ion channels with W-7.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canal de Potássio Kv1.5/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Sulfonamidas/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Linhagem Celular , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Concentração Inibidora 50 , Rim/citologia , Mutação , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/farmacologia , Sulfonamidas/administração & dosagem
8.
Curr Opin Investig Drugs ; 11(9): 1048-58, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20730700

RESUMO

Despite being the most common arrhythmia currently treated by cardiologists, safe and effective treatments for atrial fibrillation (AF) remain elusive. To address this issue, Astellas Pharma Inc, Merck & Co Inc and Cardiome Pharma Corp are developing vernakalant (RSD-1235), a drug which dose-dependently inhibits sodium channels and several potassium repolarizing currents. Of particular note, vernakalant inhibits I(Kur) (K(v)1.5), a current that is more predominant in atrial than in ventricular tissue. Consistent with this observation, vernakalant produced increases in atrial refractory period with minimal actions on QTc interval or ventricular refractory period in both humans and animals. Intravenous vernakalant terminated recent-onset AF in several animal models, and also in patients with short-duration AF or AF following cardiac surgery enrolled in phase II and III clinical trials. Vernakalant was well tolerated and adverse reactions were transient and mild. Thus, vernakalant holds considerable promise for the treatment of recent-onset AF; however, given its relatively short half-life, continuous dosing may be required in order to maintain sinus rhythm following conversion from AF. The efficacy and safety of vernakalant for the long-term management of AF remains to be determined. Phase III clinical trials with intravenous vernakalant are ongoing, and phase II clinical trials are also being conducted with an oral formulation intended for chronic use.


Assuntos
Anisóis/uso terapêutico , Fibrilação Atrial/tratamento farmacológico , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/uso terapêutico , Pirrolidinas/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Animais , Anisóis/efeitos adversos , Anisóis/metabolismo , Anisóis/farmacocinética , Fibrilação Atrial/metabolismo , Ensaios Clínicos como Assunto , Cães , Avaliação Pré-Clínica de Medicamentos , Humanos , Macaca fascicularis , Masculino , Bloqueadores dos Canais de Potássio/efeitos adversos , Bloqueadores dos Canais de Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacocinética , Pirrolidinas/efeitos adversos , Pirrolidinas/metabolismo , Pirrolidinas/farmacocinética , Coelhos , Bloqueadores dos Canais de Sódio/efeitos adversos , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacocinética
9.
Pflugers Arch ; 460(5): 851-62, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20734202

RESUMO

The goal of the present study was to examine the effects of lobeline, an agonist at nicotinic receptors, on cloned Kv channels, Kv1.5, Kv3.1, Kv4.3, and human ether-a-gogo-related gene (HERG), which are stably expressed in Chinese hamster ovary (CHO) or human embryonic kidney 293 (HEK293) cells. Whole-cell patch-clamp experiments revealed that lobeline accelerated the decay rate of Kv1.5 inactivation, decreasing the current amplitude at the end of the pulse in a concentration-dependent manner with a half-maximal inhibitory concentration (IC(50)) value of 15.1 µM. Using a time constant for the time course of drug-channel interaction, the apparent association (k(+1)), and dissociation rate (k(-1)) constants were 2.4 µΜ(-1) s(-1) and 40.9 s(-1), respectively. The calculated K(D) was 17.0 µΜ. Lobeline slowed the decay rate of the tail current, resulting in a tail crossover phenomenon. The inhibition of Kv1.5 by lobeline steeply increased at potentials between -10 and +10 mV, which corresponds to the voltage range of channel activation. At more depolarized potentials a weaker voltage dependence was observed (δ=0.26). The voltage dependence of the steady-state activation curve was not affected by lobeline, but lobeline shifted the steady-state inactivation curve of Kv1.5 in the hyperpolarizing direction. Lobeline produced use-dependent inhibition of Kv1.5 at frequencies of 1 and 2 Hz, and slowed the recovery from inactivation. Lobeline also inhibited Kv3.1, Kv4.3, and HERG in a concentration-dependent manner, with IC(50) values of 21.7, 28.2, and 0.34 µM, respectively. These results indicate that lobeline produces a concentration-, time-, voltage-, and use-dependent inhibition of Kv1.5, which can be interpreted as an open-channel block mechanism.


Assuntos
Canal de Potássio Kv1.5/efeitos dos fármacos , Lobelina/farmacologia , Animais , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Técnicas de Patch-Clamp , Canais de Potássio Shal/antagonistas & inibidores , Canais de Potássio Shaw/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Regulador Transcricional ERG
10.
IDrugs ; 13(8): 517-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20721818

RESUMO

The 32nd National Medicinal Chemistry Symposium, held in Minneapolis, MN, USA, included topics covering new developments in the field of medicinal chemistry. This conference report highlights selected presentations on Hsp90 inhibitors and Hsp70 inducers, such as KU-32 and KU-174 (University of Kansas); natural products in drug design, such as minnelide (University of Minnesota) and tylocrebrine; novel compounds from Merck for metabolic and cardiovascular diseases, such as MK-7725, a series of DDP4 inhibitors and KV1.5 ion channel antagonists; and the discovery of the VEGFR2 kinase inhibitor AMG-429 (Amgen Inc).


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Drogas em Investigação/uso terapêutico , Transtornos Mentais/tratamento farmacológico , Doenças Metabólicas/tratamento farmacológico , Neoplasias/tratamento farmacológico , Animais , Desenho de Fármacos , Proteínas de Choque Térmico HSP70/agonistas , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
11.
J Ethnopharmacol ; 131(1): 187-95, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20600762

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aconitum has been widely used to treat various diseases in China for a long time. However, improper use of this drug results in severe intoxication. Aconitine (ACO), a diterpenoid alkaloid from aconitum, mainly contributes to cardio-toxic effects of aconitum and has also been commonly known to induce arrhythmias in animal models. However, its pro-arrhythmic mechanisms are not clear. AIM OF THE STUDY: The effects of ACO on HERG and Kv1.5 channels were investigated. MATERIALS AND METHODS: HERG and Kv1.5 channels were expressed in Xenopus laevis oocytes, and the resulting currents were recorded using a two-microelectrode voltage clamp technique. RESULTS: In HERG channels, ACO exhibited a blockade in a voltage- and time-dependent manner. The blockade was enhanced by further activation of currents, which were consistent with an open-channel blockade. In Kv1.5 channels, ACO produced a voltage-, time-, and frequency-dependent inhibition. The blockade was enhanced by higher rates of stimulation, consistent with preferential binding of the drug to the open state. In addition, ACO blocked Kv1.5 and HERG channels in a concentration-dependent manner with an IC(50) of 0.796+/-0.123 and 1.801+/-0.332 microM, respectively. CONCLUSIONS: ACO blocks HERG and Kv1.5 potassium channels in the open state. Blockade of potassium channels, particular the HERG channel, may be one of the important mechanisms of how ACO induces arrhythmias.


Assuntos
Aconitina/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Humanos , Canal de Potássio Kv1.5/fisiologia , Xenopus laevis
12.
J Gen Physiol ; 135(2): 135-47, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20100893

RESUMO

Voltage-dependent potassium (K(v)) channels play a pivotal role in the modulation of macrophage physiology. Macrophages are professional antigen-presenting cells and produce inflammatory and immunoactive substances that modulate the immune response. Blockage of K(v) channels by specific antagonists decreases macrophage cytokine production and inhibits proliferation. Numerous pharmacological agents exert their effects on specific target cells by modifying the activity of their plasma membrane ion channels. Investigation of the mechanisms involved in the regulation of potassium ion conduction is, therefore, essential to the understanding of potassium channel functions in the immune response to infection and inflammation. Here, we demonstrate that the biophysical properties of voltage-dependent K(+) currents are modified upon activation or immunosuppression in macrophages. This regulation is in accordance with changes in the molecular characteristics of the heterotetrameric K(v)1.3/K(v)1.5 channels, which generate the main K(v) in macrophages. An increase in K(+) current amplitude in lipopolysaccharide-activated macrophages is characterized by a faster C-type inactivation, a greater percentage of cumulative inactivation, and a more effective margatoxin (MgTx) inhibition than control cells. These biophysical parameters are related to an increase in K(v)1.3 subunits in the K(v)1.3/K(v)1.5 hybrid channel. In contrast, dexamethasone decreased the C-type inactivation, the cumulative inactivation, and the sensitivity to MgTx concomitantly with a decrease in K(v)1.3 expression. Neither of these treatments apparently altered the expression of K(v)1.5. Our results demonstrate that the immunomodulation of macrophages triggers molecular and biophysical consequences in K(v)1.3/K(v)1.5 hybrid channels by altering the subunit stoichiometry.


Assuntos
Imunomodulação , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Macrófagos/imunologia , Animais , Linhagem Celular , Dexametasona/farmacologia , Humanos , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.5/antagonistas & inibidores , Lipopolissacarídeos/imunologia , Macrófagos/efeitos dos fármacos , Potenciais da Membrana , Camundongos , Ratos , Venenos de Escorpião/farmacologia
13.
Acta Pharmacol Sin ; 29(8): 913-22, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18664324

RESUMO

AIM: The objectives of this study were to investigate the inhibitory action of telmisartan, a selective angiotensin II type 1 receptor antagonist, on hKv1.5 and human ether-a-go-go-related gene (HERG) channels expressed on Xenopus laevis oocytes. METHODS: hKv1.5 and HERG channels were expressed on Xenopus laevis oocytes and studied using the 2-microelectrode voltage clamp technique. RESULTS: In hKv1.5 channels, telmisartan produced a voltage- and concentration-dependent inhibition; the efficacies of blockade were different at peak and 1.5 s end-pulse currents, which were 7.75%+/-2.39% (half-maximal inhibition concentration [IC50]=2.25+/-0.97 micromol/L) and 52.64%+/-3.77% (IC50=0.82+/-0.39 micromol/L) at 1 micromol/L telmisartan, respectively. Meanwhile, telmisartan accelerated the inactivation of the channels. However, telmisartan exhibited a low affinity for HERG channels (IC50=24.35+/-5.06 micromol/L); the blockade was voltage- and concentration-dependent. Telmisartan preferentially blocked open-state HERG channels. The slow time constants of deactivation were accelerated (n=6, P<0.05), which was inconsistent with the "foot-in-the-door"effect. CONCLUSION: Telmisartan blocks hKv1.5 potassium channels involving open and inactivated states at plasma concentration levels of therapeutic doses; whereas the blockade of HERG channels occurs only at supra plasma concentration levels of therapeutic doses and preferentially in open and closed-state channels.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio , Animais , Relação Dose-Resposta a Droga , Eletrofisiologia , Feminino , Humanos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Telmisartan , Xenopus laevis
14.
Am J Physiol Heart Circ Physiol ; 294(4): H1851-61, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18245566

RESUMO

Membrane- associated guanylate kinase proteins (MAGUKs) are important determinants of localization and organization of ion channels into specific plasma membrane domains. However, their exact role in channel function and cardiac excitability is not known. We examined the effect of synapse-associated protein 97 (SAP97), a MAGUK abundantly expressed in the heart, on the function and localization of Kv1.5 subunits in cardiac myocytes. Recombinant SAP97 or Kv1.5 subunits tagged with green fluorescent protein (GFP) were overexpressed in rat neonatal cardiac myocytes and in Chinese hamster ovary (CHO) cells from adenoviral or plasmidic vectors. Immunocytochemistry, fluorescence recovery after photobleaching, and patch-clamp techniques were used to study the effects of SAP97 on the localization, mobility, and function of Kv1.5 subunits. Adenovirus-mediated SAP97 overexpression in cardiac myocytes resulted in the clustering of endogenous Kv1.5 subunits at myocyte-myocyte contacts and an increase in both the maintained component of the outward K(+) current, I(Kur) (5.64 +/- 0.57 pA/pF in SAP97 myocytes vs. 3.23 +/- 0.43 pA/pF in controls) and the number of 4-aminopyridine-sensitive potassium channels in cell-attached membrane patches. In live myocytes, GFP-Kv1.5 subunits were mobile and organized in clusters at the basal plasma membrane, whereas SAP97 overexpression reduced their mobility. In CHO cells, Kv1.5 channels were diffusely distributed throughout the cell body and freely mobile. When coexpressed with SAP97, Kv subunits were organized in plaquelike clusters and poorly mobile. In conclusion, SAP97 regulates the K(+) current in cardiac myocytes by retaining and immobilizing Kv1.5 subunits in the plasma membrane. This new regulatory mechanism may contribute to the targeting of Kv channels in cardiac myocytes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Membrana Celular/metabolismo , Canal de Potássio Kv1.5/metabolismo , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/metabolismo , Potássio/metabolismo , 4-Aminopiridina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Células CHO , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Proteína 1 Homóloga a Discs-Large , Recuperação de Fluorescência Após Fotodegradação , Vetores Genéticos , Imuno-Histoquímica , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Potenciais da Membrana , Proteínas de Membrana/genética , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Transporte Proteico , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Transfecção
15.
Bioorg Med Chem Lett ; 16(22): 5859-63, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16942874

RESUMO

A novel class of tetrahydroindolone-derived semicarbazones has been discovered as potent Kv1.5 blockers. In in vitro studies, several compounds exhibited very good potency for blockade of Kv1.5. Compound 8i showed good selectivity for blockade of Kv1.5 vs hERG and L-type calcium channels. In an anesthetized pig model, compounds 8i and 10c increased atrial ERP about 28%, 18%, respectively, in the right atrium without affecting ventricular ERP.


Assuntos
Potencial Evocado Motor/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/farmacologia , Semicarbazonas/química , Semicarbazonas/farmacologia , Animais , Canais de Cálcio Tipo L/farmacologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/farmacologia , Potencial Evocado Motor/fisiologia , Frequência Cardíaca/fisiologia , Humanos , Testes Neuropsicológicos , Relação Estrutura-Atividade , Suínos
16.
Bioorg Med Chem Lett ; 16(22): 5855-8, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16942878

RESUMO

A novel class of tetrahydroindolone-derived carbamates has been discovered whose members are potent Kv1.5 blockers. The in vitro data show that compounds 6 and 29 are quite potent. They are also very selective over hERG (>450-fold) and L-type calcium channels (>450-fold).


Assuntos
Carbamatos/química , Carbamatos/farmacologia , Indóis/química , Indóis/farmacologia , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Cálcio Tipo L/farmacologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/farmacologia , Humanos , Relação Estrutura-Atividade
17.
Ann Neurol ; 60(1): 118-27, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16729292

RESUMO

OBJECTIVE: Potassium (K(+)) channels on immune cells have gained attention recently as promising targets of therapy for immune-mediated neurological diseases such as multiple sclerosis (MS). We examined K(+) channels on dendritic cells (DCs), which infiltrate the brain in MS and may impact disease course. METHODS: We identified K(+) channels on blood-derived DCs by whole-cell patch-clamp analysis, confirmed by immunofluorescent staining. We also stained K(+) channels in brain sections from MS patients and control subjects. To test functionality, we blocked K(v)1.3 and K(v)1.5 in stimulated DCs with pharmacological blockers or with an inducible dominant-negative K(v)1.x adenovirus construct and analyzed changes in costimulatory molecule upregulation. RESULTS: Electrophysiological analysis of DCs showed an inward-rectifying K(+) current early after stimulation, replaced by a mix of voltage-gated K(v)1.3- and K(v)1.5-like channels at later stages of maturation. K(v)1.3 and K(v)1.5 were also highly expressed on DCs infiltrating MS brain tissue. Of note, we found that CD83, CD80, CD86, CD40, and interleukin-12 upregulation were significantly impaired on K(v)1.3 and K(v)1.5 blockade. INTERPRETATION: These data support a functional role of K(v)1.5 and K(v)1.3 on activated human DCs and further define the mechanisms by which K(+) channel blockade may act to suppress immune-mediated neurological diseases.


Assuntos
Encéfalo/imunologia , Doenças do Sistema Nervoso Central/imunologia , Células Dendríticas/fisiologia , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Adenoviridae/genética , Adulto , Idoso , Encéfalo/citologia , Doenças do Sistema Nervoso Central/patologia , Células Dendríticas/efeitos dos fármacos , Feminino , Imunofluorescência , Genes Dominantes , Humanos , Imuno-Histoquímica , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Lipopolissacarídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Mutagênese , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Transfecção
18.
Am J Physiol Cell Physiol ; 290(3): C907-16, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16236819

RESUMO

Acute hypoxia causes pulmonary vasoconstriction in part by inhibiting voltage-gated K(+) (Kv) channel activity in pulmonary artery smooth muscle cells (PASMC). The hypoxia-mediated decrease in Kv currents [I(K(V))] is selective to PASMC; hypoxia has little effect on I(K(V)) in mesenteric artery smooth muscle cells (MASMC). Functional Kv channels are homo- and/or heterotetramers of pore-forming alpha-subunits and regulatory beta-subunits. KCNA5 is a Kv channel alpha-subunit that forms functional Kv channels in PASMC and regulates resting membrane potential. We have shown that acute hypoxia selectively inhibits I(K(V)) through KCNA5 channels in PASMC. Overexpression of the human KCNA5 gene increased I(K(V)) and caused membrane hyperpolarization in HEK-293, COS-7, and rat MASMC and PASMC. Acute hypoxia did not affect I(K(V)) in KCNA5-transfected HEK-293 and COS-7 cells. However, overexpression of KCNA5 in PASMC conferred its sensitivity to hypoxia. Reduction of Po(2) from 145 to 35 mmHg reduced I(K(V)) by approximately 40% in rat PASMC transfected with human KCNA5 but had no effect on I(K(V)) in KCNA5-transfected rat MASMC (or HEK and COS cells). These results indicate that KCNA5 is an important Kv channel that regulates resting membrane potential and that acute hypoxia selectively reduces KCNA5 channel activity in PASMC relative to MASMC and other cell types. Because Kv channels (including KCNA5) are ubiquitously expressed in PASMC and MASMC, the observation from this study indicates that a hypoxia-sensitive mechanism essential for inhibiting KCNA5 channel activity is exclusively present in PASMC. The divergent effect of hypoxia on I(K(V)) in PASMC and MASMC also may be due to different expression levels of KCNA5 channels.


Assuntos
Hipóxia Celular/fisiologia , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/citologia , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Canal de Potássio Kv1.5/genética , Masculino , Potenciais da Membrana , Artérias Mesentéricas/citologia , Ratos , Ratos Sprague-Dawley
19.
Circulation ; 112(10): 1494-9, 2005 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16129789

RESUMO

BACKGROUND: Pergolide produces clinical benefit in Parkinson disease by stimulating dopamine D1 and D2 receptors. An increased incidence of carcinoid-like heart valve disease (CLHVD) has been noted in pergolide users, reminiscent of that induced by certain anorexigens used for weight reduction. Anorexigens that modulate serotonin release and reuptake, such as dexfenfluramine, were withdrawn from sale because of CLHVD. Interestingly, the anorexigens also caused pulmonary arterial hypertension (PAH). Anorexigens were shown to enhance hypoxic pulmonary vasoconstriction, in part by inhibiting voltage-gated K+ channels (Kv) in pulmonary artery smooth muscle cells (PASMCs). Although PAH has not been associated with pergolide use, we hypothesized that pergolide might have similar effects on hypoxic pulmonary vasoconstriction and Kv channels. METHODS AND RESULTS: Pergolide enhanced hypoxic pulmonary vasoconstriction in the isolated perfused rat lung compared with control lungs (mean pulmonary artery pressure 32+/-3 versus 21+/-2 mm Hg; P<0.01). Pergolide also caused vasoconstriction in rat pulmonary artery rings. Pergolide inhibited PASMC potassium current density, resulting in membrane depolarization (from -51+/-2 to -44+/-1 mV) and increased cytosolic calcium in both rat and human PASMCs. Pergolide directly inhibited heterologously expressed Kv1.5 and KCa channels. CONCLUSIONS: Pergolide causes Kv channel inhibition and, despite being from a different class of drugs, has pulmonary vascular effects reminiscent of dexfenfluramine. Coupled with their shared proclivity to induce CLHVD, these findings suggest that clinical monitoring for pergolide-induced PAH should be considered.


Assuntos
Canal de Potássio Kv1.5/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Pergolida/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Artéria Pulmonar/citologia , Vasoconstrição/efeitos dos fármacos , Animais , Células CHO , Cálcio/metabolismo , Cricetinae , Agonistas de Dopamina/farmacologia , Humanos , Técnicas In Vitro , Canal de Potássio Kv1.5/genética , Masculino , Músculo Liso Vascular/citologia , Técnicas de Patch-Clamp , Perfusão , Ratos , Ratos Sprague-Dawley , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA