Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Mol Cell Cardiol ; 51(5): 722-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21803046

RESUMO

The transient outward current I(to) is an important determinant of the early repolarization phase. I(to) and its molecular basis Kv4.3 are regulated by adrenergic pathways including protein kinase C. However, the exact regulatory mechanisms have not been analyzed yet. We here analyzed isoenzyme specific regulation of Kv4.3 and I(to) by PKC. Kv4.3 channels were expressed in Xenopus oocytes and currents were measured with double electrode voltage clamp technique. Patch clamp experiments were performed in isolated rat cardiomyocytes. Unspecific PKC stimulation with PMA resulted in a reduction of Kv4.3 current. Similar effects could be observed after activation of conventional PKC isoforms by TMX. Both effects were reversible by pharmacological inhibition of the conventional PKC isoenzymes (Gö6976). In contrast, activation of the novel PKC isoforms (ingenol) did not significantly affect Kv4.3 current. Whereas TMX-induced PKC activation was not attenuated inhibition of PKCß, inhibition of PKCα with HBDDE prevented inhibitory effects of both PMA and TMX. Accordingly, stimulatory effects of PMA and TMX could be mimicked by the α-isoenzyme selective PKC activator iripallidal. Further evidence for the central role of PKCα was provided with the use of siRNAs. We found that PKCα siRNA but not PKCß siRNA abolished the TMX induced effect. In isolated rat cardiomyocytes, PMA dependent I(to) reduction could be completely abolished by pharmacologic inhibition of PKCα. In summary we show that PKCα plays a central role in protein kinase C dependent regulation of Kv4.3 current and native I(to). These results add to the current understanding of isoenzyme selective ion channel regulation by protein kinases.


Assuntos
Potenciais da Membrana/fisiologia , Miócitos Cardíacos/metabolismo , Oócitos/metabolismo , Proteína Quinase C-alfa/metabolismo , Canais de Potássio Shal/metabolismo , Transdução de Sinais , Animais , Carbazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Isoenzimas/genética , Isoenzimas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Técnicas de Patch-Clamp , Plasmídeos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Proteína Quinase C-alfa/genética , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Potássio Shal/genética , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Xenopus
2.
Br J Pharmacol ; 154(8): 1680-90, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18516069

RESUMO

BACKGROUND AND PURPOSE: Two-pore-domain potassium (K2P) channels mediate potassium background (or 'leak') currents, controlling excitability by stabilizing membrane potential below firing threshold and expediting repolarization. Inhibition of K2P currents permits membrane potential depolarization and excitation. As expected for key regulators of excitability, leak channels are under tight control from a plethora of stimuli. Recently, signalling via protein tyrosine kinases (TKs) has been implicated in ion channel modulation. The objective of this study was to investigate TK regulation of K2P channels. EXPERIMENTAL APPROACH: The two-electrode voltage clamp technique was used to record K2P currents in Xenopus oocytes. In addition, K2P channels were studied in Chinese hamster ovary (CHO) cells using the whole-cell patch clamp technique. KEY RESULTS: Here, we report inhibition of human K2P3.1 (TASK-1) currents by the TK antagonist, genistein, in Xenopus oocytes (IC50=10.7 microM) and in CHO cells (IC50=12.3 microM). The underlying molecular mechanism was studied in detail. hK2P3.1 was not affected by genistin, an inactive analogue of genistein. Perorthovanadate, an inhibitor of tyrosine phosphatase activity, reduced the inhibitory effect of genistein. Current reduction was voltage independent and did not require channel protonation at position H98 or phosphorylation at the single TK phosphorylation site, Y323. Among functional hK2P family members, genistein also reduced K2P6.1 (TWIK-2), K2P9.1 (TASK-3) and K2P13.1 (THIK-1) currents, respectively. CONCLUSIONS AND IMPLICATIONS: Modulation of K2P channels by the TK inhibitor, genistein, represents a novel molecular mechanism to alter background K+ currents.


Assuntos
Genisteína/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Eletrofisiologia , Genisteína/administração & dosagem , Humanos , Concentração Inibidora 50 , Proteínas do Tecido Nervoso/metabolismo , Oócitos , Técnicas de Patch-Clamp , Fosforilação , Canais de Potássio de Domínios Poros em Tandem/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais , Xenopus laevis
3.
Curr Pharm Des ; 12(18): 2271-83, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16787254

RESUMO

Human ether-a-go-go-related gene (hERG) potassium channels conduct the rapid component of the delayed rectifier potassium current, IKr, which is crucial for repolarization of cardiac action potentials. Moderate hERG blockade may produce a beneficial class III antiarrhythmic effect. In contrast, a reduction in hERG currents due to either genetic defects or adverse drug effects can lead to hereditary or acquired long QT syndromes characterized by action potential prolongation, lengthening of the QT interval on the surface ECG, and an increased risk for "torsade de pointes" arrhythmias and sudden death. This undesirable side effect of non-antiarrhythmic compounds has prompted the withdrawal of several blockbuster drugs from the market. Studies on mechanisms of hERG channel inhibition provide significant insights into the molecular factors that determine state-, voltage-, and use-dependency of hERG current block. In addition, crucial properties of the high-affinity drug binding site in hERG and its interaction with drug molecules have been identified, providing the basis for more refined approaches in drug design, safety pharmacology and in silico modeling. Recently, mutations in hERG have been shown to cause current increase and hereditary short QT syndrome with a high risk for life-threatening arrhythmias. Finally, the discovery of adrenergic mechanisms of hERG channel regulation as well as the development of strategies to enhance hERG currents and to modify intracellular hERG protein processing may provide novel antiarrhythmic options in repolarization disorders. In conclusion, the increasing understanding of hERG channel function and molecular mechanisms of hERG current regulation could improve prevention and treatment of hERG-associated cardiac repolarization disorders.


Assuntos
Antiarrítmicos/efeitos adversos , Antidepressivos Tricíclicos/efeitos adversos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Coração/efeitos dos fármacos , Síndrome do QT Longo/etiologia , Bloqueadores dos Canais de Potássio/efeitos adversos , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Coração/inervação , Humanos , Ativação do Canal Iônico , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Modelos Moleculares , Miocárdio/metabolismo , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/uso terapêutico , Conformação Proteica , Receptores Adrenérgicos/efeitos dos fármacos , Receptores Adrenérgicos/metabolismo , Estresse Fisiológico/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo
4.
Biochim Biophys Acta ; 1690(3): 185-92, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15511625

RESUMO

Romano-Ward syndrome (RWS), the autosomal dominant form of the congenital long QT syndrome, is characterised by prolongation of the cardiac repolarisation process associated with ventricular tachyarrhythmias of the torsades de pointes type. Genetic studies have identified mutations in six ion channel genes, KCNQ1, KCNH2, SCN5A, KCNE1 and KCNE2 and the accessory protein Ankyrin-B gene, to be responsible for this disorder. Single-strand conformation polymorphism (SSCP) analysis and subsequent DNA sequence analysis have identified a KCNQ1 mutation in a family that were clinically conspicuous due to several syncopes and prolonged QTc intervals in the ECG. The mutant subunit was expressed and functionally characterised in the Xenopus oocyte expression system. A novel heterozygous missense mutation with a C to T transition at the first position of codon 343 (CCA) of the KCNQ1 gene was identified in three concerned family members (QTc intervals: 500, 510 and 530 ms, respectively). As a result, proline 343 localised within the highly conserved transmembrane segment S6 of the KCNQ1 channel is replaced by a serine. Co-expression of mutant (KCNQ1-P343S) and wild-type (KCNQ1) cRNA in Xenopus oocytes produced potassium currents reduced by approximately 92%, while IKs reconstitution experiments with a combination of KCNQ1 mutant, wild-type and KCNE1 subunits yielded currents reduced by approximately 60%. A novel mutation (P343S) identified in the KCNQ1 subunit gene of three members of a RWS family showed a dominant-negative effect on native IKs currents leading to prolongation of the heart repolarisation and possibly increases the risk of malign arrhythmias with sudden cardiac death.


Assuntos
Mutação/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Síndrome de Romano-Ward/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Análise Mutacional de DNA , Eletrofisiologia , Feminino , Expressão Gênica , Humanos , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Masculino , Dados de Sequência Molecular , Oócitos/metabolismo , Técnicas de Patch-Clamp , Linhagem , Fenótipo , Polimorfismo Conformacional de Fita Simples , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Prolina/genética , Alinhamento de Sequência , Xenopus laevis
5.
Herzschrittmacherther Elektrophysiol ; 16(4): 229-38, 2005 Dec.
Artigo em Alemão | MEDLINE | ID: mdl-16362729

RESUMO

The last decade has seen rapid progress in our understanding of the molecular basis of arrhythmias, particularly concerning hereditary arrhythmia syndromes. This has led to significant improvement regarding differentiation, risk stratification and therapy in these patients and their families. However, there is mounting evidence that the knowledge obtained by studying these rare monogenic disorders will also enable us to dissect the molecular mechanisms underlying polygenetic and multi-factorial arrhythmias that are by far more common in clinical practice. The goal of this review is to give a brief overview of current knowledge on the molecular basis of primary electrical heart diseases. A focus is on the long QT syndrome.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Modelos Cardiovasculares , Canais de Potássio/metabolismo , Animais , Humanos , Biologia Molecular/métodos
6.
Cardiovasc Res ; 49(2): 361-70, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11164846

RESUMO

OBJECTIVE: The aryloxypropanolamine carvedilol is a multiple action cardiovascular drug with blocking effects on alpha-receptors, beta-receptors, Ca(2+)-channels, Na(+)-channels and various native cardiac K(+) channels, thereby prolonging the cardiac action potential. In a number of clinical trials with patients suffering from congestive heart failure, carvedilol appeared to be superior to other beta-blocking agents in reducing total mortality. Given the multiple pharmacological actions of carvedilol, this may be due to specific channel blockade rather than beta-antagonistic activity. Since human ether-a-go-go related gene (HERG) K(+)channels play a critical role in the pathogenesis of cardiac arrhythmias and sudden cardiac death, the effects of carvedilol on HERG K(+)channels were investigated. METHODS: Double-electrode voltage-clamp experiments were performed on HERG potassium channels which were expressed heterologously in Xenopus oocytes. RESULTS: Carvedilol at a concentration of 10 microM blocked HERG potassium tail currents by 47%. The electrophysiological characteristics of HERG, i.e. activation, steady-state inactivation and recovery from inactivation were not affected by carvedilol. Inhibition of current gradually increased from 0% immediately after the test pulse to about 80% at 600 ms with subsequent marginal changes of current kinetics during the resting 29 s, indicating a very fast open channel block by carvedilol as the major blocking mechanism. CONCLUSION: This is the first study demonstrating that carvedilol blocks HERG potassium channels. The biophysical data presented in this study with a potentially antiarrhythmic effect may contribute to the positive outcome of clinical trials with carvedilol.


Assuntos
Antiarrítmicos/farmacologia , Carbazóis/farmacologia , Proteínas de Transporte de Cátions , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Propanolaminas/farmacologia , Animais , Carvedilol , Depressão Química , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go , Feminino , Técnicas In Vitro , Ativação do Canal Iônico/efeitos dos fármacos , Técnicas de Patch-Clamp , Xenopus
7.
Herzschrittmacherther Elektrophysiol ; 26(2): 129-33, 2015 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-25939989

RESUMO

While the implantable cardioverter-defibrillator (ICD) has been proven to be the best choice for patients with long-term risk for sudden cardiac arrest/sudden cardiac death (SCA/SCD), the question is how to manage patients with only temporary risk, e.g., during the guidelines-recommended waiting period until the decision for an ICD can be made. These patient groups should be monitored around the clock to guarantee a lifesaving shock within a few minutes, if necessary.These conditions can be accomplished by the wearable cardioverter-defibrillator (WCD) in the outpatient sector. The WCD is worn on the skin and consists of four nonadhesive ECG electrodes as well as three defibrillation electrodes-two at the back and one at the front-embedded in a garment. The defibrillation unit is connected via a cord and can be worn over the shoulder or on a belt. Cardiac events can be recorded and retrospectively analyzed by the treating physician.The WCD is a safe and effective measure to terminate potentially lethal ventricular tachycardia and ventricular fibrillation. It may be used early after myocardial infarction with reduced left ventricular ejection fraction (LVEF), as well as for patients with acute heart failure in nonischemic cardiomyopathy with uncertain cause and prognosis. In addition, it may be used for patients waiting for heart transplantation, for patients who cannot be implanted an ICD due to comorbidities, and for patients after explantation of their ICD, e.g., because of infection until reimplantation.One may expect that risk stratification of patients with the WCD will lead to even better selection for ICD use.


Assuntos
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/prevenção & controle , Morte Súbita Cardíaca/prevenção & controle , Desfibriladores , Cardioversão Elétrica/instrumentação , Eletrocardiografia Ambulatorial/instrumentação , Desenho de Equipamento , Análise de Falha de Equipamento , Medicina Baseada em Evidências , Humanos , Resultado do Tratamento
8.
Naunyn Schmiedebergs Arch Pharmacol ; 358(3): 374-81, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9774226

RESUMO

Single channel cell-attached patch and whole-cell clamp experiments on the mode of action of the K+ channel opener (KCO), levcromakalim, were performed in guinea pig isolated portal vein cells. At +20 mV (135/23 mM K+ in bath/pipette), 10 microM levcromakalim activated K+ channels with a chord conductance of 23.2 pS (K(KCO)), which were sensitive to the blocker of ATP-dependent K+ channels (K(ATP)), glibenclamide. Voltage steps from -80 mV to +20 mV activated 4-aminopyridine-sensitive K+ channels of 6.5 pS with properties of delayed rectifier K+ channels (Kv). In patches which upon a previous voltage step had revealed the existence of Kv, levcromakalim reduced the open-probability of Kv, but it did not concomitantly activate K(KCO). During the course of the experiments, but unrelated to the presence of levcromakalim, large conductance K+ channels (BK(Ca)) appeared which could be inhibited by iberiotoxin, a selective blocker of BK(Ca), and by the membrane-permeant calcium buffer, BAPTA/AM, but not by glibenclamide. Whole-cell current-voltage (i-V) relations were established in response to voltage ramps from +50 mV to -100 mV; on subtraction of control i-V curves from i-V curves obtained in the presence of 10 microM levcromakalim, the KCO-induced K+ current remained which was proportional to voltage. This is not compatible with the upward-bent curvature predicted by the GHK current equation for purely resistive channels at high [K+]i versus low [K+]o. In conclusion, in the guinea pig portal vein cells, no evidence could be established for the hypotheses that KCOs may act via conversion of Kv to K(ATP) (Beech and Bolton 1989; Edwards et al. 1993) or by activation of BK(Ca) (Balwierczak et al. 1995). In these cells, mild inward rectification of the levcromakalim-induced current was observed which underlines their relationship to K(ATP) in other tissues.


Assuntos
Cromakalim/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Cálcio/farmacologia , Cromakalim/antagonistas & inibidores , Interações Medicamentosas , Glibureto/farmacologia , Cobaias , Hipoglicemiantes/farmacologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Veia Porta/efeitos dos fármacos
9.
Naunyn Schmiedebergs Arch Pharmacol ; 359(3): 212-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10208308

RESUMO

The human ether-a-go-go-related gene (HERG) encodes a K+ channel with biophysical properties nearly identical to the rapid component of the cardiac-delayed rectifier K+ current (I(Kr)). HERG channels are one primary target for the pharmacological management of arrhythmias. In this study, we investigated the acute effects of the class III antiarrhythmic drug amiodarone on HERG channels expressed heterologously in Xenopus oocytes by use of the two-microelectrode voltage clamp technique. Amiodarone blocked HERG channels with an IC50 of 9.8 microM with a maximum outward tail current reduction of 62.8%. The block consisted of two main components, a closed channel block that could not be reversed within the time of experiments and an open channel block with a slow unblock, having a recovery time constant of 73 s at -80 mV. Inactivation of the HERG channel at very positive potentials could not prevent amiodarone block. These results indicate that HERG channels can be blocked by amiodarone in closed, open and inactivated states. The block of open channels was cumulative, use-dependent and voltage-dependent. In summary, our data suggest that the strong class III antiarrhythmic action of amiodarone is at least partially based upon its acute inhibitory effects on HERG potassium channels.


Assuntos
Amiodarona/farmacologia , Antiarrítmicos/farmacologia , Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Oócitos/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Transativadores , Xenopus/genética , Animais , Clonagem Molecular , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Humanos , Técnicas In Vitro , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Fatores de Tempo , Regulador Transcricional ERG
10.
Br J Pharmacol ; 165(5): 1467-75, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21838752

RESUMO

BACKGROUND AND PURPOSE: TASK1 (K(2P)3.1) two-pore-domain K(+) channels contribute substantially to the resting membrane potential in human pulmonary artery smooth muscle cells (hPASMC), modulating vascular tone and diameter. The endothelin-1 (ET-1) pathway mediates vasoconstriction and is an established target of pulmonary arterial hypertension (PAH) therapy. ET-1-mediated inhibition of TASK1 currents in hPASMC is implicated in the pathophysiology of PAH. This study was designed to elucidate molecular mechanisms underlying inhibition of TASK1 channels by ET-1. EXPERIMENTAL APPROACH: Two-electrode voltage clamp and whole-cell patch clamp electrophysiology was used to record TASK1 currents from hPASMC and Xenopus oocytes. KEY RESULTS: ET-1 inhibited TASK1-mediated I(KN) currents in hPASMC, an effect attenuated by Rho kinase inhibition with Y-27632. In Xenopus oocytes, TASK1 current reduction by ET-1 was mediated by endothelin receptors ET(A) (IC(50) = 0.08 nM) and ET(B) (IC(50) = 0.23 nM) via Rho kinase signalling. TASK1 channels contain two putative Rho kinase phosphorylation sites, Ser(336) and Ser(393) . Mutation of Ser(393) rendered TASK1 channels insensitive to ET(A) - or ET(B)-mediated current inhibition. In contrast, removal of Ser(336) selectively attenuated ET(A) -dependent TASK1 regulation without affecting the ET(B) pathway. CONCLUSIONS AND IMPLICATIONS: ET-1 regulated vascular TASK1 currents through ET(A) and ET(B) receptors mediated by downstream activation of Rho kinase and direct channel phosphorylation. The Rho kinase pathway in PASMC may provide a more specific therapeutic target in pulmonary arterial hypertension treatment.


Assuntos
Endotelina-1/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Animais , Células Cultivadas , Feminino , GTP Fosfo-Hidrolases/metabolismo , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Mutação , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiologia , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Transdução de Sinais , Vasoconstrição/genética , Vasoconstrição/fisiologia , Xenopus laevis , Quinases Associadas a rho/antagonistas & inibidores
11.
Kidney Int ; 70(2): 321-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16760913

RESUMO

Treatment of organ donors with catecholamines reduces acute rejection episodes and improves long-term graft survival after renal transplantation. The aim of this study was to investigate the effect of catecholamine pre-treatment on ischemia/reperfusion (I/R)- and cold preservation injury in rat kidneys. I/R-injury was induced by clamping the left kidney vessels for 60 min along with a contralateral nephrectomy. Cold preservation injury was induced by storage of the kidneys for 24 h at +4 degrees Celsius in University of Wisconsin solution, followed by syngeneic transplantation. Rats were pre-treated with either dopamine (DA), dobutamine (DB), or norepinephrine (2, 5, and 10 microg/kg/min, each group) intravenously via an osmotic minipump for 24 h before I/R- and cold preservation injury. Pre-treatment with DA (2 or 5 microg/kg/min) and DB (5 microg/kg/min) improved recovery of renal function after I/R-injury and dose dependently reduced mononuclear and major histocompatibility complex class II-positive cells infiltrating the kidney after I/R-injury. One day after I/R-injury, upregulation of transforming growth factor (TGF)-beta 1 and 2 and phosphorylation of p42/p44 mitogen-activated protein kinases was observed in kidneys of animals treated with DA or DB. DA (5 microg/kg/min) and DB (5 microg/kg/min) pre-treatment reduced endothelial cell damage after 24 h of cold preservation. Only DA pre-treatment improved renal function and reduced renal inflammation after 24 h of cold preservation and syngeneic transplantation. Our results demonstrate a protective effect of pre-treatment with catecholamines on renal inflammation and function after I/R- or cold preservation injury. This could help to explain the potent organoprotective effects of catecholamine pre-treatment observed in human kidney transplantation.


Assuntos
Catecolaminas/farmacologia , Criopreservação/métodos , Precondicionamento Isquêmico/métodos , Transplante de Rim , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Temperatura Baixa , Dobutamina/farmacologia , Dopamina/farmacologia , Dopaminérgicos/farmacologia , Sobrevivência de Enxerto , Rim/efeitos dos fármacos , Rim/fisiologia , Rim/cirurgia , Masculino , Nefrectomia/métodos , Norepinefrina/farmacologia , Ratos , Ratos Endogâmicos Lew , Traumatismo por Reperfusão/prevenção & controle , Simpatomiméticos/farmacologia
12.
J Biol Chem ; 275(35): 26743-7, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10823835

RESUMO

The slow component of the delayed rectifier potassium current (IKs) plays an important role during repolarization in the human heart. Life-threatening arrhythmias can be triggered by sympathetic stimulation, presumably acting on IKs. The ion channel responsible for the IKs current is made of two proteins, the KvLQT1 protein and the MinK protein. In this study, we investigated the effects of adrenergic stimulation on the KvLQT1/MinK channel by coexpressing KvLQT1/MinK channels with the human beta(3)-adrenoreceptor subunit heterologously in Xenopus oocytes. Western blot experiments revealed that beta(3)-adrenoreceptor proteins appear in the cell membrane of Xenopus oocytes, when the corresponding cRNA was injected. In electrophysiological measurements we found that stimulation with the beta-adrenergic agonist isoproterenol increased the current amplitude of the beta(3)/KvLQT1/MinK complex up to 237% with an ED(50) of 8 nm, a value similar to that found on IKs in guinea pig cardiomyocytes. When oocytes with beta(3)/KvLQT1/MinK were preincubated with cholera toxin (2 microg/ml), an activator of G(S) proteins, the basal current amplitude of the beta(3)/KvLQT1/MinK complex was increased 3.1-fold, and the current amplitude increase by isoproterenol was drastically reduced, indicating that the signal transduction cascade was mediated via G(s) proteins. The knowledge about functional coupling of the human beta(3)-adrenoreceptor to KvLQT1/MinK channels reveals interesting aspects about the genesis and therapy of arrhythmias.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Humanos , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Síndrome do QT Longo/metabolismo , Ligação Proteica , Receptores Adrenérgicos beta 3 , Xenopus laevis
13.
J Biol Chem ; 273(39): 25285-91, 1998 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-9737994

RESUMO

We investigated the effects of the phorbol ester phorbol 12-myristate 13-acetate (PMA) on the rapid component of the delayed rectifier potassium current, IKr, in guinea pig cardiomyocytes and found that the IKr current amplitude was reduced by 20% with 10 nM PMA and 44% with 100 nM PMA. The ether-a-go-go-related gene (HERG) encodes IKr in human heart. We expressed HERG heterologously in Xenopus oocytes and investigated the effects of PMA on the delayed rectifier potassium current. Upon application of PMA in a concentration of 100 nM, we found a similar reduction of HERG outward current amplitude by 59%. This reduction was due to a shift in the HERG activation curve by 37 mV. The ED50 for the PMA-induced shift was 9.0 nM. The inactive 4alpha-phorbol 12-myristate 13-acetate (4alpha-PMA) had no effect. PMA is known to act by stimulating distinct protein kinase cascades. Additional application of the specific protein kinase C inhibitors chelerythrine (10 microM) or bisindolylmaleimide (1 microM) could not attenuate the PMA-induced shift. In contrast, the shift by PMA was reduced significantly when the specific protein kinase A (PKA) inhibitors H89 (50 microM) or KT5720 (2.5 microM) were applied. Forskolin (400 microM), an activator of the adenylate cyclase that results in PKA activation, shifted the HERG activation curve by 14 mV. Moreover the specific protein kinase C activator 1-stearoyl-2-arachidonylglycerol (10 microM) showed no effect. Our data suggest that mainly PKA is mediating the shift of the HERG activation kinetics.


Assuntos
Proteínas de Transporte de Cátions , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Síndrome do QT Longo/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Alcaloides , Animais , Benzofenantridinas , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Canais de Potássio Éter-A-Go-Go , Cobaias , Indóis/farmacologia , Maleimidas/farmacologia , Potenciais da Membrana , Miocárdio/metabolismo , Miocárdio/patologia , Técnicas de Patch-Clamp , Fenantridinas/farmacologia , Canais de Potássio/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Acetato de Tetradecanoilforbol/farmacologia
14.
Langenbecks Arch Chir ; 366: 579-83, 1985.
Artigo em Alemão | MEDLINE | ID: mdl-2414622

RESUMO

Between 1979 and 1984 a total of 233 patients underwent surgical treatment of small bowel obstruction. In 43 patients (18.4%) the obstruction was caused by an advanced tumor disease (peritoneal carcinosis, local recurrence). In patients with benign obstruction the operative mortality was 5.2%; in the tumor patients it was 14%. Wound infection and cardio-pulmonary disturbances were the most frequent complications. The mean survival time of the tumor patients was 159 days. In 65% of these patients the operation had a significant palliative effect.


Assuntos
Neoplasias Gastrointestinais/complicações , Obstrução Intestinal/cirurgia , Intestino Delgado/cirurgia , Neoplasias Gastrointestinais/cirurgia , Humanos , Recidiva Local de Neoplasia/cirurgia , Cuidados Paliativos , Complicações Pós-Operatórias/mortalidade , Prognóstico , Qualidade de Vida
15.
Pacing Clin Electrophysiol ; 24(8 Pt 1): 1240-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11523610

RESUMO

Initial experience with the Medtronic Jewel 7250, the ICD designed to detect and treat ventricular and supraventricular tachyarrhythmias, is very promising. Its effectiveness, however, depends on sensing performance, which has not yet been systematically examined. The aim of the study was to determine the incidence of, predisposing factors for, and practical implications of far-field R wave oversensing (FFRWOS) in this dual chamber ICD. During a total follow-up of 797 months in 48 patients who had the Jewel 7250, follow-up strip charts, 12-channel Holter recordings and, in particular cases, Holter recordings with intracardiac markers were analyzed for the presence of FFRWOS. FFRWOS was documented in ten (21.3%) patients. Compared to other lead locations, the right atrial appendage lead position was most frequently associated with FFRWOS (7/27 vs 3/21, P < 0.05). Patients with FFRWOS had significantly more treated and nontreated atrial episodes, many of which were judged to have been detected inappropriately. In one case, inappropriate atrial antitachycardia pacing due to R wave oversensing triggered sustained ventricular tachycardia, terminated eventually with a high energy shock. In dual chamber ICDs, FFRWOS may represent a frequent phenomenon possibly leading to serious consequences. For atrial leads, a lateral atrial wall position seems to be preferable. In most cases, FFRWOS can be eliminated by optimization of atrial sensing parameters. Given the possibility of ventricular proarrhythmia with atrial pacing therapy, the capability of ventricular backup defibrillation in respective devices is at least reassuring.


Assuntos
Arritmias Cardíacas/diagnóstico , Desfibriladores Implantáveis/efeitos adversos , Adulto , Idoso , Algoritmos , Arritmias Cardíacas/etiologia , Eletrocardiografia , Eletrocardiografia Ambulatorial , Eletrodos Implantados , Falha de Equipamento , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
J Biol Chem ; 274(39): 27457-62, 1999 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-10488078

RESUMO

We investigated the role of protein kinase A (PKA) in regulation of the human ether-a-go-go-related gene (HERG) potassium channel activation. HERG clones with single mutations destroying one of four consensus PKA phosphorylation sites (S283A, S890A, T895A, S1137A), as well as one clone carrying all mutations with no PKA phosphorylation sites (HERG 4M) were constructed. These clones were expressed heterologously in Xenopus oocytes, and HERG potassium currents were measured with the two microelectrode voltage clamp technique. Application of the cAMP-specific phosphodiesterase (PDE IV) inhibitor Ro-20-1724 (100 microM), which results in an increased cAMP level and PKA stimulation, induced a reduction of HERG wild type outward currents by 19.1% due to a shift in the activation curve of 12.4 mV. When 100 microM Ro-20-1724 was applied to the HERG 4M channel, missing all PKA sites, there was no significant shift in the activation curve, and the current amplitude was not reduced. Furthermore, the adenylate cyclase activator forskolin that leads to PKA activation (400 microM, 60 min), shifted HERG wild type channel activation by 14.1 mV and reduced currents by 39.9%, whereas HERG 4M channels showed only a small shift of 4.3 mV and a weaker current reduction of 22.3%. We conclude that PKA regulates HERG channel activation, and direct phosphorylation of the HERG channel protein has a functional role that may be important in regulation of cardiac repolarization.


Assuntos
Proteínas de Transporte de Cátions , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação a DNA , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Transativadores , 4-(3-Butoxi-4-metoxibenzil)-2-imidazolidinona/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Colforsina/farmacologia , Sequência Consenso , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Feminino , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Moleculares , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Fosforilação , Canais de Potássio/química , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Regulador Transcricional ERG , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA