Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; : e0045824, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38814067

RESUMO

Tryptophan metabolism plays a crucial role in facilitating various cellular processes essential for maintaining normal cellular function. Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the conversion of tryptophan (Trp) into kynurenine (Kyn), thereby initiating the degradation of Trp. The resulting Kyn metabolites have been implicated in the modulation of immune responses. Currently, the role of IDO1-mediated tryptophan metabolism in the process of viral infection remains relatively unknown. In this study, we discovered that classical swine fever virus (CSFV) infection of PK-15 cells can induce the expression of IDO1, thereby promoting tryptophan metabolism. IDO1 can negatively regulate the NF-κB signaling by mediating tryptophan metabolism, thereby facilitating CSFV replication. We found that silencing the IDO1 gene enhances the expression of IFN-α, IFN-ß, and IL-6 by activating the NF-κB signaling pathway. Furthermore, our observations indicate that both silencing the IDO1 gene and administering exogenous tryptophan can inhibit CSFV replication by counteracting the cellular autophagy induced by Rapamycin. This study reveals a novel mechanism of IDO1-mediated tryptophan metabolism in CSFV infection, providing new insights and a theoretical basis for the treatment and control of CSFV.IMPORTANCEIt is well known that due to the widespread use of vaccines, the prevalence of classical swine fever (CSF) is shifting towards atypical and invisible infections. CSF can disrupt host metabolism, leading to persistent immune suppression in the host and causing significant harm when co-infected with other diseases. Changes in the host's metabolic profiles, such as increased catabolic metabolism of amino acids and the production of immunoregulatory metabolites and their derivatives, can also influence virus replication. Mammals utilize various pathways to modulate immune responses through amino acid utilization, including increased catabolic metabolism of amino acids and the production of immunoregulatory metabolites and their derivatives, thereby limiting viral replication. Therefore, this study proposes that targeting the modulation of tryptophan metabolism may represent an effective approach to control the progression of CSF.

2.
Assay Drug Dev Technol ; 12(6): 352-60, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25147907

RESUMO

Human stem cell-derived cardiomyocytes provide a cellular model for the study of electrophysiology in the human heart and are finding a niche in the field of safety pharmacology for predicting proarrhythmia. The cardiac L-type Ca2+ channel is an important target for some of these safety studies. However, the pharmacology of this channel in these cells is altered compared to native cardiac tissue, specifically in its sensitivity to the Ca2+ channel activator S-(-)-Bay K 8644. Using patch clamp electrophysiology, we examined the effects of S-(-)-Bay K 8644 in three separate stem cell-derived cardiomyocyte cell lines under various conditions in an effort to detect more typical responses to the drug. S-(-)-Bay K 8644 failed to produce characteristically large increases in current when cells were held at -40 mV and Ca2+ was used as the charge carrier, although high-affinity binding and the effects of the antagonist isomer, R-(+)-Bay K 8644, were intact. Dephosphorylation of the channel with acetylcholine failed to restore the sensitivity of the channel to the drug. Only when the holding potential was shifted to a more hyperpolarized (-60 mV) level, and external Ca2+ was replaced by Ba2+, could large increases in current amplitude be observed. Even under these conditions, increases in current amplitude varied dramatically between different cell lines and channel kinetics following drug addition were generally atypical. The results indicate that the pharmacology of S-(-)-Bay K 8644 in stem cell-derived cardiomyocytes varies by cell type, is unusually dependent on holding potential and charge carrier, and is different from that observed in primary human heart cells.


Assuntos
Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L , Células-Tronco Embrionárias/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Canais de Cálcio Tipo L/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/fisiologia , Cobaias , Humanos , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/fisiologia
3.
J Pharmacol Exp Ther ; 341(2): 510-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22353878

RESUMO

Human stem cell-derived cardiomyocytes provide new models for studying the ion channel pharmacology of human cardiac cells for both drug discovery and safety pharmacology purposes. However, detailed pharmacological characterization of ion channels in stem cell-derived cardiomyocytes is lacking. Therefore, we used patch-clamp electrophysiology to perform a pharmacological survey of the L-type Ca²âº channel in induced pluripotent and embryonic stem cell-derived cardiomyocytes and compared the results with native guinea pig ventricular cells. Six structurally distinct antagonists [nifedipine, verapamil, diltiazem, lidoflazine, bepridil, and 2-[(cis-2-phenylcyclopentyl)imino]-azacyclotridecane hydrochloride (MDL 12330)] and two structurally distinct activators [methyl 2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-1,4-dihydropyridine-3-carboxylate (Bay K8644) and 2,5-dimethyl-4-[2-(phenylmethyl)benzoyl]-1H-pyrrole-3-carboxylic acid methyl ester (FPL 64176)] were used. The IC50 values for the six antagonists showed little variability between the three cell types. However, whereas Bay K8644 produced robust increases in Ca²âº channel current in guinea pig myocytes, it failed to enhance current in the two stem cell lines. Furthermore, Ca²âº channel current kinetics after addition of Bay K8644 differed in the stem cell-derived cardiomyocytes compared with native cells. FPL 64176 produced consistently large increases in Ca²âº channel current in guinea pig myocytes but had a variable effect on current amplitude in the stem cell-derived myocytes. The effects of FPL 64176 on current kinetics were similar in all three cell types. We conclude that, in the stem cell-derived myocytes tested, L-type Ca²âº channel antagonist pharmacology is preserved, but the pharmacology of activators is altered. The results highlight the need for extensive pharmacological characterization of ion channels in stem cell-derived cardiomyocytes because these complex proteins contain multiple sites of drug action.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Cobaias , Coração/efeitos dos fármacos , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo
4.
Toxicol Lett ; 208(1): 62-8, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-22037396

RESUMO

Mephedrone (4-methylmethcathinone) is a new and popular drug of abuse widely available on the Internet and still legal in some parts of the world. Clinical reports are now emerging suggesting that the drug displays sympathomimetic toxicity on the cardiovascular system but no studies have yet explored its cardiovascular effects. Therefore we examined the effects of mephedrone on the cardiovascular system using a combination of in vitro electrophysiology and in vivo hemodynamic and echocardiographic measurements. Patch clamp studies revealed that mephedrone, up to 30 µM, had little effect on the major voltage-dependent ion channels of the heart or on action potentials recorded in guinea pig myocytes. Subcutaneous administration of mephedrone (3 and 15 mg/kg) to conscious telemetry-implanted rats produced dose-dependent increases in heart rate and blood pressure which persisted after pre-treatment with reserpine. Echocardiographic analysis demonstrated that intravenous injection of mephedrone (0.3 and 1mg/kg) increased cardiac function, including cardiac output, ejection fraction, and stroke volume, similar to methamphetamine (0.3mg/kg). We conclude that mephedrone is not directly pro-arrhythmic, but induces substantial increases in heart rate, blood pressure and cardiac contractility and this activity contributes to the cardiovascular toxicity in people who abuse the drug.


Assuntos
Drogas Desenhadas/toxicidade , Hemodinâmica/efeitos dos fármacos , Drogas Ilícitas/toxicidade , Metanfetamina/análogos & derivados , Animais , Pressão Sanguínea/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ecocardiografia , Técnicas Eletrofisiológicas Cardíacas , Cobaias , Coração/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/induzido quimicamente , Masculino , Metanfetamina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Taquicardia/induzido quimicamente , Testes de Toxicidade Aguda
5.
J Pharmacol Exp Ther ; 334(2): 619-26, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20484151

RESUMO

Epigallocatechin-3-gallate (EGCG) is the major catechin found in green tea. EGCG is also available for consumption in the form of concentrated over-the-counter nutritional supplements. This compound is currently undergoing clinical trials for the treatment of a number of diseases including multiple sclerosis, and a variety of cancers. To date, few data exist regarding the effects of EGCG on the electrophysiology of the heart. Therefore, we examined the effects of EGCG on the electrocardiogram recorded from Langendorff-perfused guinea pig hearts and on cardiac ion channels using patch-clamp electrophysiology. EGCG had no significant effects on the electrocardiogram at concentrations of 3 and 10 microM. At 30 microM, EGCG prolonged PR and QRS intervals, slightly shortened the QT interval, and altered the shape of the ST-T-wave segment. The ST segment merged with the upstroke of the T wave, and we noted a prolongation in the time from the peak of the T wave until the end. Patch-clamp studies identified the KvLQT1/minK K(+) channel as a target for EGCG (IC(50) = 30.1 microM). In addition, EGCG inhibited the cloned human cardiac Na(+) channel Na(v)1.5 in a voltage-dependent fashion. The L-type Ca(2+) channel was inhibited by 20.8% at 30 microM, whereas the human ether-a-go-go-related gene and Kv4.3 cardiac K(+) channels were less sensitive to inhibition by EGCG. ECGC has a number of electrophysiological effects in the heart, and these effects may have clinical significance when multigram doses of this compound are used in human clinical trials or through self-ingestion of large amounts of over-the-counter products enriched in EGCG.


Assuntos
Catequina/análogos & derivados , Coração/efeitos dos fármacos , Canais Iônicos/fisiologia , Chá , Animais , Células CHO , Canais de Cálcio Tipo L/fisiologia , Catequina/química , Catequina/farmacologia , Cricetinae , Cricetulus , Eletrocardiografia , Cobaias , Coração/fisiologia , Humanos , Técnicas In Vitro , Ativação do Canal Iônico , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Canais de Sódio/fisiologia , Estereoisomerismo
6.
Clin Exp Pharmacol Physiol ; 36(11): 1104-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19413590

RESUMO

Summary 1. The non-selective K(+) channel blocker 4-aminopyridine (4-AP) has shown clinical efficacy in the treatment of neurological disorders such as multiple sclerosis. The clinical usefulness of 4-AP is hampered by its ability to produce seizures. Nerispirdine, an analogue of 4-AP, is currently under clinical investigation for the treatment of multiple sclerosis. In contrast with 4-AP, nerispirdine is not proconvulsant, suggesting mechanistic differences between the two drugs. 2. Using whole-cell patch-clamp electrophysiology, we compared the effects of 4-AP and nerispirdine on the cloned human K(+) channels K(v)1.1 and K(v)1.2, expressed in Chinese hamster ovary cells, and on voltage-dependent Na(+) channels recorded from human SH-SY5Y cells. 3. Nerispirdine inhibited K(v)1.1 and K(v)1.2 with IC(50) values of 3.6 and 3.7 micromol/L, respectively. 4-Aminopyridine was approximately 50-fold less potent at blocking these channels. Nerispirdine also inhibited voltage-dependent Na(+) channel currents recorded from human SH-SY5Y cells with an IC(50) of 11.9 micromol/L when measured from a -70 mV holding potential. In contrast, 4-AP had no effect on Na(+) channel currents. 4. The results demonstrate that nerispirdine, like 4-AP, can inhibit axonal K(+) channels and that this mechanism may underlie the ability of the drug to enhance neuronal conduction. Unlike 4-AP, nerispirdine can also inhibit neuronal Na(+) channels, a mechanism that may explain why nerispirdine lacks proconvulsant activity.


Assuntos
4-Aminopiridina/análogos & derivados , 4-Aminopiridina/farmacologia , Indóis/farmacologia , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.2/antagonistas & inibidores , Piridinas/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Proteínas de Drosophila , Feminino , Humanos , Potenciais da Membrana/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio , Bloqueadores dos Canais de Sódio/farmacologia
7.
Anesthesiology ; 104(5): 1015-22, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16645454

RESUMO

BACKGROUND: Sevoflurane prolongs ventricular repolarization in patients, but the mechanisms are not fully characterized. The effects of sevoflurane on many cloned human cardiac ion channels have not been studied, and the interactions between sevoflurane and other drugs that prolong cardiac repolarization have not been detailed. METHODS: The effects of sevoflurane on action potentials and L-type Ca channels in guinea pig myocytes were examined. Sevoflurane's effects on cloned human cardiac K channels and the cloned human cardiac Na channel were studied. The consequences of combining sevoflurane and the class III antiarrhythmic drugs sotalol or dofetilide on action potential duration were also examined. RESULTS: Sevoflurane produced an increase in action potential duration at concentrations of 0.3-1 mm. Contrary to most drugs that delay ventricular repolarization, sevoflurane was without effect on the human ether-a-go-go-related gene cardiac potassium channel but instead produced a reduction in KvLQT1/minK K channel currents and inhibited the Kv4.3 K channel by speeding its apparent rate of inactivation. Sevoflurane had little effect on Na and Ca channel currents at concentrations of 1 mm or less. When the authors coadministered sevoflurane with sotalol or dofetilide, synergistic effects on repolarization were observed, resulting in large increases in action potential duration (up to 66%). CONCLUSION: Prolonged ventricular repolarization observed with administration of sevoflurane results from inhibition of KvLQT1/minK and Kv4.3 cardiac K channels. Combining sevoflurane with class III antiarrhythmic drugs results in supra-additive effects on action potential duration. The results indicate that sevoflurane, when administered with this class of drug, could result in excessive delays in ventricular repolarization. The results suggest the need for further clinical studies.


Assuntos
Anestésicos Inalatórios/farmacologia , Síndrome do QT Longo/induzido quimicamente , Éteres Metílicos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Antagonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Células CHO , Separação Celular , Cricetinae , Interações Medicamentosas , Cobaias , Humanos , Técnicas In Vitro , Canais Iônicos/efeitos dos fármacos , Síndrome do QT Longo/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Sevoflurano , Sotalol/farmacologia , Sulfonamidas/farmacologia
8.
Mol Pharmacol ; 67(3): 827-36, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15548764

RESUMO

Many drugs inhibit the human ether-a-go-go-related gene (HERG) cardiac K+ channel. This leads to action potential prolongation on the cellular level, a prolongation of the QT interval on the electrocardiogram, and sometimes cardiac arrhythmia. To date, no activators of this channel have been reported. Here, we describe the in vitro electrophysiological effects of (3R,4R)-4-[3-(6-methoxyquinolin-4-yl)-3-oxo-propyl]-1-[3-(2,3,5-trifluoro-phenyl)-prop-2-ynyl]-piperidine-3-carboxylic acid (RPR260243), a novel activator of HERG. Using patch-clamp electrophysiology, we found that RPR260243 dramatically slowed current deactivation when applied to cells stably expressing HERG. The effects of RPR260243 on HERG channel deactivation were temperature- and voltage-dependent and occurred over the concentration range of 1 to 30 microM. RPR260243-modified HERG currents were inhibited by dofetilide (IC50 = 58 nM). RPR260243 had little effect on HERG current amplitude and no significant effects on steady-state activation parameters or on channel inactivation processes. RPR260243 displayed no activator-like effects on other voltage-dependent ion channels, including the closely related erg3 K+ channel. RPR260243 enhanced the delayed rectifier current in guinea pig myocytes but, when administered alone, had little effect on action potential parameters in these cells. However, RPR260243 completely reversed the action potential-prolonging effects of dofetilide in this preparation. Using the Langendorff heart method, we found that 5 microM RPR260243 increased T-wave amplitude, prolonged the PR interval, and shortened the QT interval. We believe RPR260243 represents the first known HERG channel activator and that the drug works primarily by inhibiting channel closure, leading to a persistent HERG channel current upon repolarization. Compounds like RPR260243 will be useful for studying the physiological role of HERG and may one day find use in treating cardiac disease.


Assuntos
Coração/fisiologia , Piperidinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Quinolinas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Células CHO , Cricetinae , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Cobaias , Coração/efeitos dos fármacos , Humanos , Técnicas In Vitro , Cinética , Síndrome do QT Longo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
9.
J Pharmacol Exp Ther ; 308(3): 935-40, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14711935

RESUMO

Tolterodine is a muscarinic antagonist widely used in the treatment of urinary incontinence. Although tolterodine has not been reported to alter cardiac repolarization, it is chemically related to other muscarinic antagonists known to prolong cardiac repolarization. For this reason, we studied the effects of tolterodine on cardiac ion channels and action potential recordings. Using patch-clamp electrophysiology, we found that tolterodine was a potent antagonist of the human ether-a-go-go-related gene (HERG) K(+) channel, displaying an IC(50) value of 17 nM. This potency was similar to that observed for the antiarrhythmic drug dofetilide (IC(50) of 11 nM). Tolterodine block of HERG displayed a positive voltage dependence, suggesting an interaction with an activated state. Tolterodine had little effect on the human cardiac Na(+) channel at concentrations of up to 1 microM. Inhibition of L-type Ca(2+) currents by tolterodine was frequency-dependent with IC(50) values measuring 143 and 1084 nM at 1 and 0.1 Hz, respectively. Both tolterodine and dofetilide prolonged action potential duration in single guinea pig myocytes over the concentration range of 3 to 100 nM. However, prolongation was significantly larger for dofetilide compared with tolterodine. Tolterodine seems to be an unusual drug in that it blocks HERG with high affinity, but produces little QT prolongation clinically. Low plasma levels after therapeutic doses combined with mixed ion channel effects, most notably Ca(2+) channel blockade, may serve to attenuate the QT prolonging effects of this potent HERG channel antagonist.


Assuntos
Compostos Benzidrílicos/farmacologia , Cresóis/farmacologia , Coração/efeitos dos fármacos , Canais Iônicos/metabolismo , Fenilpropanolamina , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Células CHO , Cricetinae , Eletrofisiologia , Coração/fisiologia , Tartarato de Tolterodina
10.
J Cardiovasc Pharmacol ; 43(2): 281-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14716218

RESUMO

Under basal conditions there is no observable nitric oxide synthase (NOS) activity in vascular smooth muscle (VSM). Pretreatment of endothelium-denuded aortic rings from Sprague-Dawley rats with 1-(5-isoquinolinylsulfonyl)-2-methylpiperazine (H-7), (0.1 micromol/L) significantly attenuated phenylephrine (PE)-induced contractile responses in a dose-dependent manner. In the presence of 10 micromol/L Nomega-nitro-L-arginine (L-NNA) or 0.1 mmol/L aminoguanidine (AG), the inhibition of contractions at 10 nmol/L PE by H-7 was blocked by 88% or 52%, respectively. The blockade by antagonists was completely reversed by l-arginine but not by d-arginine, and alone they did not significantly alter PE-induced contraction of endothelium-denuded aorta. Methylene blue (MB, 50 micromol/L) also inhibited the action of H-7. The inhibitory effect of H-7 occurred after 5 minutes and was reversible. PE-induced contraction was also inhibited by the selective protein kinase C inhibitors calphostin C (10 micromol/L), and bisindolylmaleimide IV (Bis-IV, 10 micromol/L), but not by the selective protein kinase A inhibitor H-89 (0.1 micromol/L). These results indicate protein kinase C inhibits NOS activity in VSM under basal conditions. Incubation of tissues with either H-7 or calphostin C stimulates NO production, and immunocytochemical studies reveal the presence of NOS in VSM under basal conditions.


Assuntos
Inibidores Enzimáticos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Nitroarginina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Animais , Masculino , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/metabolismo , Óxido Nítrico/biossíntese , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Vasoconstrição/efeitos dos fármacos
11.
Can J Physiol Pharmacol ; 82(11): 1006-17, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15644941

RESUMO

We hypothesized that regional differences in electrophysiological properties exist within the musculature of the feline lower esophageal sphincter (LES) and that they may potentially contribute to functional asymmetry within the LES. Freshly isolated esophageal smooth muscle cells (SMCs) from the circular muscle and sling regions within the LES were studied under a patch clamp. The resting membrane potential (RMP) of the circular SMCs was significantly more depolarized than was the RMP of the sling SMCs, resulting from a higher Na+ and Cl- permeability in circular muscle than in sling muscle. Large conductance Ca2+-activated K+ (BKCa) set the RMP at both levels, since specific BKCa inhibitors caused depolarization; however, BKCa density was greatest in the circular region. A significant portion of the outward current was due to non-BKCa, especially in sling muscle, and likely delayed rectifier K+ channels (KDR). There was a large reduction in outward current with 4-aminopyridine (4-AP) in sling muscle, while BKCa blockers had a limited effect on the voltage-activated outward current in sling muscle. Differences in BKCa:KDR channel ratios were also manifest by a leftward shift in the voltage-dependent activation curve in circular cells compared to sling cells. The electrophysiological differences seen between the circular and sling muscles provide a basis for their different contributions to LES activities such as resting tone and neurotransmitter responsiveness, and in turn could impart asymmetric drug responses and provide specific therapeutic targets.


Assuntos
Esfíncter Esofágico Inferior/fisiologia , Miócitos de Músculo Liso/fisiologia , Canais de Potássio/fisiologia , Animais , Gatos , Esfíncter Esofágico Inferior/citologia , Feminino , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia
12.
J Biol Chem ; 278(30): 27556-63, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12871971

RESUMO

The slower kinetics of insulin release from pancreatic islet beta cells, as compared with other regulated secretory processes such as chromaffin granule secretion, can in part be explained by the small number of the insulin granules that are docked to the plasma membrane and readily releasable. In type-2 diabetes, the kinetics of insulin secretion become grossly distorted, and, to therapeutically correct this, it is imperative to elucidate the mechanisms that regulate priming and secretion of insulin secretory granules. Munc13-1, a synaptic protein that regulates SNARE complex assembly, is the major protein determining the priming of synaptic vesicles. Here, we demonstrate the presence of Munc13-1 in human, rat, and mouse pancreatic islet beta cells. Expression of Munc13-1, along with its cognate partners, syntaxin 1a and Munc18a, is reduced in the pancreatic islets of type-2 diabetes non-obese Goto-Kakizaki and obese Zucker fa/fa rats. In insulinoma cells, overexpressed Munc13-1-enhanced green fluorescent protein is translocated to the plasma membrane in a temperature-dependent manner. This, in turn, greatly amplifies insulin exocytosis as determined by patch clamp capacitance measurements and radioimmunoassay of the insulin released. The potentiation of exocytosis by Munc13-1 is dependent on endogenously produced diacylglycerol acting on the overexpressed Munc13-1 because it is blocked by a phospholipase C inhibitor (U73122) and abrogated when the diacylglycerol binding-deficient Munc13-1H567K mutant is expressed instead of the wild type protein. Our data demonstrate that Munc13-mediated vesicle priming is not restricted to neurotransmitter release but is also functional in insulin secretion, where it is subject to regulation by the diacylglycerol second messenger pathway. In view of our findings, Munc13-1 is a potential drug target for therapeutic optimization of insulin secretion in diabetes.


Assuntos
Insulina/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Animais , Antígenos de Superfície/metabolismo , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Exocitose , Glucose/metabolismo , Proteínas de Fluorescência Verde , Humanos , Immunoblotting , Insulinoma/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/metabolismo , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Microscopia Confocal , Modelos Moleculares , Proteínas Munc18 , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Patch-Clamp , Proteína Quinase C/metabolismo , Transporte Proteico , Ratos , Ratos Zucker , Sintaxina 1 , Temperatura , Transfecção , Fosfolipases Tipo C/antagonistas & inibidores , Proteínas de Transporte Vesicular/metabolismo
13.
Biochem Biophys Res Commun ; 306(1): 298-302, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12788104

RESUMO

We recently reported that non-secretory gastrointestinal smooth muscle cells also possessed SNARE proteins, of which SNAP-25 regulated Ca(2+)-activated (K(Ca)) and delayed rectifier K(+) channels (K(V)). Voltage-gated, long lasting (L-type) calcium channels (L(Ca)) play an important role in excitation-contraction coupling of smooth muscle. Here, we show that SNAP-25 could also directly inhibit the L-type Ca(2+) channels in feline esophageal smooth muscle cells at the SNARE complex binding synprint site. SNARE proteins could therefore regulate additional cell actions other than membrane fusion and secretion, in particular, coordinated muscle membrane excitability and contraction, through their actions on membrane Ca(2+) and K(+) channels.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Esôfago/efeitos dos fármacos , Esôfago/metabolismo , Proteínas de Membrana/farmacologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/farmacologia , Proteínas de Transporte Vesicular , Animais , Sítios de Ligação , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Gatos , Esôfago/citologia , Técnicas In Vitro , Proteínas de Membrana/metabolismo , Músculo Liso/citologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas SNARE , Proteína 25 Associada a Sinaptossoma
14.
Diabetes ; 51(5): 1425-36, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11978639

RESUMO

Cognate soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins are now known to associate the secretory vesicle with both the target plasma membrane and Ca(2+) channels in order to mediate the sequence of events leading to exocytosis in neurons and neuroendocrine cells. Neuroendocrine cells, particularly insulin-secreting islet beta-cells, t-SNARE proteins, 25-kDa synaptosomal-associated protein (SNAP-25), and syntaxin 1A, independently inhibit the L-type Ca(2+) channel (L(Ca)). However, when both are present, they actually exhibit stimulatory actions on the L(Ca). This suggests that the positive regulation of the L(Ca) is conferred by a multi-SNARE protein complex. We hypothesized an alternate explanation, which is that each of these SNARE proteins possess distinct inhibitory and stimulatory domains that act on the L(Ca). These SNARE proteins were recently shown to bind the Lc(753-893) domain corresponding to the II and III intracellular loop of the alpha1C subunit of the L(Ca). In this study, using patch-clamp methods on primary pancreatic beta-cells and insulinoma HIT-T15 cells, we examined the functional interactions of the botulinum neurotoxin A (BoNT/A) cleavage products of SNAP-25, including NH(2)-terminal (1-197 amino acids) and COOH-terminal (amino acid 198-206) domains, on the L(Ca), particularly at the Lc(753-893) domain. Intracellular application of SNAP-25(1-206) in primary beta-cells decreased L(Ca) currents by approximately 15%. The reduction in L(Ca) currents was counteracted by coapplication of Lc(753-893). Overexpression or injection of wild-type SNAP-25 in HIT cells reduced L(Ca) currents by approximately 30%, and this inhibition was also blocked by the recombinant Lc(753-893) peptide. Expression of BoNT/A surprisingly caused an even greater reduction of L(Ca) currents (by 41%), suggesting that the BoNT/A cleavage products of SNAP-25 might possess distinct inhibitory and positive regulatory domains. Indeed, expression of SNAP-25(1-197) increased L(Ca) currents (by 19% at 10 mV), and these effects were blocked by the Lc(753-893) peptide. In contrast, injection of SNAP-25(198-206) peptide into untransfected cells inhibited L(Ca) currents (by 47%), and more remarkably, these inhibitory effects dominated over the stimulatory effects of SNAP-25(1-197) overexpression (by 34%). Therefore, the SNARE protein SNAP-25 possesses distinct inhibitory and stimulatory domains that act on the L(Ca). The COOH-terminal 197-206 domain of SNAP-25, whose inhibitory actions dominate over the opposing stimulatory NH(2)-terminal domain, likely confers the inhibitory actions of SNAP-25 on the L(Ca). We postulate that the eventual accelerated proteolysis of SNAP-25 brought about by BoNT/A cleavage allows the relatively intact NH(2)-terminal SNAP-25 domain to assert its stimulatory action on the L(Ca) to increase Ca(2+) influx, and this could in part explain the observed weak or inconsistent inhibitory effects of BoNT/A on insulin secretion. The present study suggests that distinct domains within SNAP-25 modulate L(C) subtype Ca(2+) channel activity in both primary beta-cells and insulinoma HIT-T15 cells.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Animais , Toxinas Botulínicas Tipo A , Cálcio/metabolismo , Linhagem Celular , Proteínas de Fluorescência Verde , Indicadores e Reagentes/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Ilhotas Pancreáticas/citologia , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Membrana/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Fármacos Neuromusculares , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Estrutura Terciária de Proteína , Proteína 25 Associada a Sinaptossoma , Transfecção
15.
J Biol Chem ; 277(23): 20195-204, 2002 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-11925439

RESUMO

Delayed-rectifier K(+) channels (K(DR)) are important regulators of membrane excitability in neurons and neuroendocrine cells. Opening of these voltage-dependent K(+) channels results in membrane repolarization, leading to the closure of the Ca(2+) channels and cessation of insulin secretion in neuroendocrine islet beta cells. Using patch clamp techniques, we have demonstrated that the activity of the K(DR) channel subtype, K(V)1.1, identified by its specific blocker dendrodotoxin-K, is inhibited by SNAP-25 in insulinoma HIT-T15 beta cells. A co-precipitation study of rat brain confirmed that SNAP-25 interacts with the K(V)1.1 protein. Cleavage of SNAP-25 by expression of botulinum neurotoxin A in HIT-T15 cells relieved this SNAP-25-mediated inhibition of K(DR). This inhibitory effect of SNAP-25 is mediated by the N terminus of K(V)1.1, likely by direct interactions with K(Valpha)1.1 and/or K(V)beta subunits, as revealed by co-immunoprecipitation performed in the Xenopus oocyte expression system and in vitro binding. Taken together we have concluded that SNAP-25 mediates secretion not only through its participation in the exocytotic SNARE complex but also by regulating membrane potential and calcium entry through its interaction with K(DR) channels.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Animais , Canais de Potássio de Retificação Tardia , Insulinoma/metabolismo , Insulinoma/patologia , Fusão de Membrana , Bloqueadores dos Canais de Potássio , Ligação Proteica , Ratos , Proteína 25 Associada a Sinaptossoma , Células Tumorais Cultivadas
16.
Gastroenterology ; 122(4): 994-1006, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11910352

RESUMO

BACKGROUND & AIMS: The plasma membrane-associated soluble N-ethylmaleimide-sensitive factors attachment protein receptors (SNAREs), synaptosome-associated protein of 25 kilodaltons (SNAP-25), and syntaxin 1A, have been found to physically interact with and functionally modify membrane-spanning ion channels. Studies were performed in cat esophageal body and lower esophageal sphincter (LES) smooth muscle to (1) show the presence of SNAP-25, and (2) determine whether SNAP-25 affects K+ channel activity. METHODS: Single circular muscle cells from the esophageal body and sphincter were studied. Cellular localization of SNAP-25 and K+ channel activity were assessed. RESULTS: SNAP-25 was found in the plasma membrane of all regions examined. Outward K+ currents in body circular muscle were mainly composed of large conductance Ca2+-activated channel currents (K(Ca), 40.1%) and delayed rectifier K+ channel currents (K(V), 54.2%). Microinjection of SNAP-25 into muscle cells caused a dose-dependent inhibition of both outward K+ currents, maximal 44% at 10(-8) mol/L. Cleavage of endogenous SNAP-25 by dialyzing botulinum neurotoxin A into the cell interior resulted in a 35% increase in outward currents. CONCLUSIONS: SNAP-25 protein is present in esophageal smooth muscle cells, and inhibits both K(V) and K(Ca) currents in circular muscle cells. The findings suggest a role for SNAP-25 in regulation of esophageal muscle cell excitability and contractility, and point to potential new targets for treatment of esophageal motor disorders.


Assuntos
Esôfago/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Proteínas de Transporte Vesicular , Animais , Toxinas Botulínicas Tipo A/farmacologia , Gatos , Membrana Celular/química , Canais de Potássio de Retificação Tardia , Relação Dose-Resposta a Droga , Técnicas In Vitro , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Membrana/análise , Proteínas de Membrana/farmacologia , Músculo Liso/citologia , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/farmacologia , Fármacos Neuromusculares/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Proteínas SNARE , Proteína 25 Associada a Sinaptossoma , Tetraetilamônio/farmacologia
17.
Am J Physiol Gastrointest Liver Physiol ; 282(2): G288-99, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11804850

RESUMO

We have characterized ion-channel identity and density differences along the feline smooth muscle esophagus using patch-clamp recording. Current clamp recording revealed that the resting membrane potential (RMP) of esophageal smooth muscle cells (SMC) from the circular layer at 4 cm above the lower esophageal sphincter (EBC4; LES) were more depolarized than at 2 cm above LES. Higher distal Na(+) permeability (but not Cl(-) permeability) contributes to this RMP difference. K(+) channels but not large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels contribute to RMP at both levels, because nonspecific K(+)-channel blockers depolarize all SMC. Depolarization of SMC under voltage clamp revealed that the density of voltage-dependent K(+) channels (K(V)) was greatest at EBC4 due to increased BK(Ca.) Delayed rectifier K(+) channels (K(DR)), compatible with subtype K(V)1.2, were present at both levels. Differences in K(Ca)-to-K(DR) channel ratios were also manifest by predictable shifts in voltage-dependent inactivation at EBC4 when BK(Ca) channels were blocked. We provide the first evidence for regional electrophysiological differences along the esophageal body resulting from SMC ion channel diversity, which could allow for differential muscular responses to innervation and varied muscular contribution to peristaltic contractions along the esophagus.


Assuntos
Ácido Egtázico/análogos & derivados , Esôfago/fisiologia , Músculo Liso/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , 4-Aminopiridina/farmacologia , Animais , Gatos , Quelantes/farmacologia , Canais de Potássio de Retificação Tardia , Ácido Egtázico/farmacologia , Venenos Elapídicos/farmacologia , Esôfago/química , Esôfago/citologia , Feminino , Canais de Potássio Ativados por Cálcio de Condutância Alta , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/fisiologia , Músculo Liso/química , Músculo Liso/citologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Potássio/farmacocinética , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/análise , Canais de Potássio Cálcio-Ativados/análise , Tetraetilamônio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...