Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Biol Chem ; 286(8): 6500-9, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21156805

RESUMO

Protein kinase D (PKD) exists as a family of structurally related enzymes that are activated through similar phosphorylation-dependent mechanisms involving protein kinase C (PKC). While individual PKD isoforms could in theory mediate distinct biological functions, previous studies identify a high level of functional redundancy for PKD1 and PKD2 in various cellular contexts. This study shows that PKD1 and PKD2 are activated in a stimulus-specific manner in neonatal cardiomyocytes. The α(1)-adrenergic receptor agonist norepinephrine selectively activates PKD1, thrombin and PDGF selectively activate PKD2, and endothelin-1 and PMA activate both PKD1 and PKD2. PKC activity is implicated in the α(1)-adrenergic receptor pathway that activates PKD1 and the thrombin- and PDGF-dependent pathways that activate PKD2. Endothelin-1 activates PKD via both rapid PKC-dependent and more sustained PKC-independent mechanisms. The functional consequences of PKD activation were assessed by tracking phosphorylation of CREB and cardiac troponin I (cTnI), two physiologically relevant PKD substrates in cardiomyocytes. We show that overexpression of an activated PKD1-S744E/S748E transgene increases CREB-Ser(133) and cTnI-Ser(23)/Ser(24) phosphorylation, but agonist-dependent pathways that activate native PKD1 or PKD2 selectively increase CREB-Ser(133) phosphorylation; there is no associated increase in cTnI-Ser(23)/Ser(24) phosphorylation. Gene silencing studies provide unanticipated evidence that PKD1 down-regulation leads to a compensatory increase in PKD2 activity and that down-regulation of PKD1 (alone or in combination with PKD2) leads to an increase in CREB-Ser(133) phosphorylation. Collectively, these studies identify distinct roles for native PKD1 and PKD2 enzymes in stress-dependent pathways that influence cardiac remodeling and the progression of heart failure.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Proteínas Musculares/metabolismo , Miócitos Cardíacos/enzimologia , Norepinefrina/farmacologia , Proteína Quinase C/metabolismo , Substituição de Aminoácidos , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Proteínas Musculares/genética , Mutação de Sentido Incorreto , Miócitos Cardíacos/patologia , Proteína Quinase C/genética , Ratos , Ratos Wistar
2.
Circ Res ; 104(5): 660-9, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19168439

RESUMO

p66Shc is an adapter protein that is induced by hypertrophic stimuli and has been implicated as a major regulator of reactive oxygen species (ROS) production and cardiovascular oxidative stress responses. This study implicates p66Shc in an alpha(1)-adrenergtic receptor (alpha(1)-AR) pathway that requires the cooperative effects of protein kinase (PK)Cepsilon and PKCdelta and leads to AKT-FOXO3a phosphorylation in cardiomyocytes. alpha(1)-ARs promote p66Shc-YY(239/240) phosphorylation via a ROS-dependent mechanism that is localized to caveolae and requires epidermal growth factor receptor (EGFR) and PKCepsilon activity. alpha(1)-ARs also increase p66Shc-S(36) phosphorylation via an EGFR transactivation pathway involving PKCdelta. p66Shc links alpha(1)-ARs to an AKT signaling pathway that selectively phosphorylates/inactivates FOXO transcription factors and downregulates the ROS-scavenging protein manganese superoxide dismutase (MnSOD); the alpha(1)-AR-p66Shc-dependent pathway involving AKT does not regulate GSK3. Additional studies show that RNA interference-mediated downregulation of endogenous p66Shc leads to the derepression of FOXO3a-regulated genes such as MnSOD, p27Kip1, and BIM-1. p66Shc downregulation also increases proliferating cell nuclear antigen expression and induces cardiomyocyte hypertrophy, suggesting that p66Shc exerts an antihypertrophic action in neonatal cardiomyocytes. The novel alpha(1)-AR- and ROS-dependent pathway involving p66Shc identified in this study is likely to contribute to cardiomyocyte remodeling and the evolution of heart failure.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Antibióticos Antineoplásicos/toxicidade , Apoptose , Cardiomegalia/metabolismo , Cavéolas/metabolismo , Crescimento Celular , Células Cultivadas , Doxorrubicina/toxicidade , Receptores ErbB/metabolismo , Proteína Forkhead Box O3 , Quinase 3 da Glicogênio Sintase/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Norepinefrina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Proteínas Adaptadoras da Sinalização Shc/genética , Transdução de Sinais/efeitos dos fármacos , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Fatores de Tempo , Transdução Genética
3.
Circ Res ; 104(12): 1382-9, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19461043

RESUMO

Complex modulation of voltage-gated Ca2+ currents through the interplay among Ca2+ channels and various Ca(2+)-binding proteins is increasingly being recognized. The K+ channel interacting protein 2 (KChIP2), originally identified as an auxiliary subunit for K(V)4.2 and a component of the transient outward K+ channel (I(to)), is a Ca(2+)-binding protein whose regulatory functions do not appear restricted to K(V)4.2. Consequently, we hypothesized that KChIP2 is a direct regulator of the cardiac L-type Ca2+ current (I(Ca,L)). We found that I(Ca,L) density from KChIP2(-/-) myocytes is reduced by 28% compared to I(Ca,L) recorded from wild-type myocytes (P<0.05). This reduction in current density results from loss of a direct effect on the Ca2+ channel current, as shown in a transfected cell line devoid of confounding cardiac ion currents. I(Ca,L) regulation by KChIP2 was independent of Ca2+ binding to KChIP2. Biochemical analysis suggested a direct interaction between KChIP2 and the Ca(V)1.2 alpha(1C) subunit N terminus. We found that KChIP2 binds to the N-terminal inhibitory module of alpha(1C) and augments I(Ca,L) current density without increasing Ca(V)1.2 protein expression or trafficking to the plasma membrane. We propose a model in which KChIP2 impedes the N-terminal inhibitory module of Ca(V)1.2, resulting in increased I(Ca,L). In the context of recent reports that KChIP2 modulates multiple K(V) and Na(V) currents, these results suggest that KChIP2 is a multimodal regulator of cardiac ionic currents.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Potenciais da Membrana/fisiologia , Proteínas Musculares/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Membrana Celular/genética , Células Cultivadas , Transporte de Íons/fisiologia , Proteínas Interatuantes com Canais de Kv/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Estrutura Terciária de Proteína/fisiologia , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo
4.
Mol Pharmacol ; 76(4): 896-902, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620255

RESUMO

Reactive oxygen species (ROS) exert pleiotropic effects on a wide array of signaling proteins that regulate cellular growth and apoptosis. This study shows that long-term treatment with a low concentration of H2O2 leads to the activation of signaling pathways involving extracellular signal-regulated kinase, ribosomal protein S6 kinase, and protein kinase D (PKD) that increase cAMP binding response element protein (CREB) phosphorylation at Ser(133) in cardiomyocytes. Although CREB-Ser(133) phosphorylation typically mediates cAMP-dependent increases in CREB target gene expression, the H2O2-dependent increase in CREB-Ser(133) phosphorylation is accompanied by a decrease in CREB protein abundance and no change in Cre-luciferase reporter activity. Mutagenesis studies indicate that H2O2 decreases CREB protein abundance via a mechanism that does not require CREB-Ser(133) phosphorylation. Rather, the H2O2-dependent decrease in CREB protein is prevented by the proteasome inhibitor lactacystin, by inhibitors of mitogen-activated protein kinase kinase or protein kinase C activity, or by adenoviral-mediated delivery of a small interfering RNA that decreases PKD1 expression. A PKD1-dependent mechanism that links oxidative stress to decreased CREB protein abundance is predicted to contribute to the pathogenesis of heart failure by influencing cardiac growth and apoptosis responses.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Miocárdio/metabolismo , Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo , Animais , Western Blotting , Regulação para Baixo/efeitos dos fármacos , Coração/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Fosforilação , Proteína Quinase C , Proteínas Quinases/química , Ratos , Ratos Wistar , Transdução de Sinais
5.
Cardiovasc Res ; 75(4): 758-69, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17588552

RESUMO

OBJECTIVE: Atrial fibrillation is often initiated by bursts of ectopic activity arising in the pulmonary veins. We have previously shown that a 3-h intermittent burst pacing protocol (BPP), mimicking ectopic pulmonary vein foci, shortens action potential duration (APD) locally at the pulmonary vein-atrial interface (PV) while having no effect elsewhere in rabbit atrium. This shortening is Ca(2+) dependent and is prevented by apamin, which blocks small conductance Ca(2+)-activated K(+) channels (SK(Ca)). The present study investigates the ionic and molecular mechanisms whereby two apamin-sensitive SK(Ca) channels, SK2 and SK3, might contribute to the regional APD changes. METHODS: Microelectrode and patch clamp techniques were used to record APDs and apamin-sensitive currents in isolated rabbit left atria and cells dispersed from PV and Bachmann's bundle (BB) regions. SK2 and SK3 mRNA and protein levels were quantified, and immunofluorescence was used to observe channel protein distribution. RESULTS: There was a direct relationship between APD shortening and apamin-sensitive current in burst-paced but not sham-paced PV. Moreover, apamin-sensitive current density increased in PV but not BB after BPP. SK2 mRNA, protein, and current were increased in PV after BPP, while SK2 immunostaining shifted from a perinuclear pattern in sham atria to predominance at sites near or at the PV membrane. CONCLUSIONS: BPP-induced acceleration of repolarization in PV results from SK2 channel trafficking to the membrane, leading to increased apamin-sensitive outward current. This is the first indication of involvement of Ca(2+)-activated K(+) currents in atrial remodeling and provides a possible basis for evolution of an arrhythmogenic substrate.


Assuntos
Potenciais de Ação/fisiologia , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Veias Pulmonares/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Apamina/farmacologia , Transporte Biológico , Membrana Celular/metabolismo , Átrios do Coração , Imuno-Histoquímica , Masculino , Microeletrodos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Veias Pulmonares/metabolismo , RNA Mensageiro/análise , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Potássio Ativados por Cálcio de Condutância Baixa/análise , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
6.
Heart Rhythm ; 4(4): 499-507, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17399641

RESUMO

BACKGROUND: Pulmonary veins (PVs) and the coronary sinus (CS) play pivotal roles in triggering some episodes of atrial fibrillation. In isolated rabbit right or left atrial preparations, a 3-hour intermittent burst pacing protocol shortens action potential duration (APD) in CS and PV, but not in sinus node (SN) and left Bachmann bundle (BB) regions. OBJECTIVE: The purpose of this study was to use patch clamp techniques to study the rapidly inactivating (I(to)) and sustained (I(sus)) K(+) currents as well as Ca(2+) currents (I(Ca)) in cells dispersed from intermittent burst pacing and sham PV, BB, CS, and SN regions to determine whether changes in these currents contributed to APD shortening. METHODS: Real-time polymerase chain reaction was performed for transient outward K(+) and Ca(2+) channel subunit mRNAs to determine if intermittent burst pacing affected expression levels. RESULTS: I(to) densities were unaffected by intermittent burst pacing in PV and Bachmann bundle cells. mRNA levels of K(V)4.3, K(V)4.2, K(V)1.4, and KChIP2 subunits of I(to) in both regions were stable. In CS cells, I(to) densities in intermittent burst pacing were greater than in sham (P <.05), but there were no parallel mRNA changes. I(Ca) density of PV cells was reduced from 14.27 +/- 2.08 pA/pF (at -5 mV) in sham to 7.52 +/- 1.65 pA/pF in intermittent burst pacing PV cells (P <.05) due to a significant shift in voltage dependence of activation. These results were seen in the absence of mRNA changes in alpha(1C) and alpha(1D) Ca(2+) channel subunits. In contrast, intermittent burst pacing had no effect on Ca(2+) current densities and kinetics of CS cells, but decreased alpha(1)C and alpha(1)D mRNA levels. CONCLUSION: There is region-specific remodeling of I(to) and I(Ca) by intermittent burst pacing protocols in rabbit atrium. Increased I(to) in CS cells could account for the APD shortening observed with intermittent burst pacing, whereas an intermittent burst pacing-induced shift in voltage dependence of activation may contribute to APD shortening in PV cells.


Assuntos
Potenciais de Ação , Função Atrial , Canais de Cálcio/metabolismo , Estimulação Cardíaca Artificial , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/terapia , Função Atrial/efeitos dos fármacos , Fascículo Atrioventricular/citologia , Fascículo Atrioventricular/metabolismo , Fascículo Atrioventricular/fisiopatologia , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Estimulação Elétrica , Eletrodos Implantados , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Átrios do Coração/fisiopatologia , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Bloqueadores dos Canais de Potássio/farmacologia , Veias Pulmonares/citologia , Veias Pulmonares/metabolismo , Veias Pulmonares/fisiopatologia , RNA Mensageiro/metabolismo , Coelhos , Tempo de Reação/efeitos dos fármacos , Projetos de Pesquisa , Nó Sinoatrial/citologia , Nó Sinoatrial/metabolismo , Nó Sinoatrial/fisiopatologia
7.
Cardiovasc Res ; 71(1): 88-96, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16626671

RESUMO

OBJECTIVE: Cardiac memory (CM) is characterized by an altered T-wave morphology, which reflects altered repolarization gradients. We hypothesized that the delayed rectifier currents, I(Kr) and I(Ks), might contribute to these repolarization changes. METHODS: We studied conscious, chronically instrumented dogs paced from the postero-lateral left ventricular (LV) wall at rates 5-10% faster than sinus rate for 3 weeks. ECGs during sinus rhythm were recorded on days 0, 7, 14 and 21 of pacing. Within 3 weeks, CM achieved steady state, hearts were excised, and epicardial and endocardial tissues and myocytes were studied. RESULTS: In unpaced controls, action potential duration to 50% and 90% repolarization (APD) in epicardium was shorter than in endocardium (P < 0.05); in CM epicardial APD increased at CL > or = 500 ms, while endocardial APD was either unchanged or decreased such that the transmural gradient seen in controls diminished (P < 0.05). A transmural I(Kr) gradient occurred in controls (epicardium>endocardium, P < 0.05) and was reversed in CM. No I(Ks) transmural gradient was found in controls, while in CM endocardial I(Ks) was greater than epicardial at greater than +50 mV. Canine ERG (cERG) mRNA and protein in epicardium > endocardium in controls (P < 0.05), and this difference was lost in CM. Expression levels of KCNQ1 and KCNE1 protein were similar in all groups. CONCLUSIONS: A transcriptionally induced change in epicardial I(Kr) contributes to the altered ventricular repolarization that characterizes CM.


Assuntos
Potenciais de Ação/fisiologia , Miócitos Cardíacos/metabolismo , Pericárdio/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Animais , Western Blotting/métodos , Estimulação Cardíaca Artificial , Cães , Eletrocardiografia , Endocárdio/metabolismo , Endocárdio/fisiologia , Canais de Potássio Éter-A-Go-Go/análise , Canais de Potássio Éter-A-Go-Go/genética , Ventrículos do Coração , Canal de Potássio KCNQ1/análise , Canal de Potássio KCNQ1/genética , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Pericárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/análise , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Remodelação Ventricular
8.
J Am Coll Cardiol ; 61(11): 1192-201, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23395072

RESUMO

OBJECTIVES: This study sought to test the hypothesis that hyperpolarization-activated cyclic nucleotide-gated (HCN)-based biological pacing might be improved significantly by hyperpolarizing the action potential (AP) threshold via coexpression of the skeletal muscle sodium channel 1 (SkM1). BACKGROUND: Gene-based biological pacemakers display effective in vivo pacemaker function. However, approaches used to date have failed to manifest optimal pacemaker properties, defined as basal beating rates of 60 to 90 beats/min, a brisk autonomic response achieving maximal rates of 130 to 160 beats/min, and low to absent electronic backup pacing. METHODS: We implanted adenoviral SkM1, HCN2, or HCN2/SkM1 constructs into left bundle branches (LBB) or left ventricular (LV) epicardium of atrioventricular-blocked dogs. RESULTS: During stable peak gene expression on days 5 to 7, HCN2/SkM1 LBB-injected dogs showed highly stable in vivo pacemaker activity superior to SkM1 or HCN2 alone and superior to LV-implanted dogs with regard to beating rates (resting approximately 80 beats/min; maximum approximately 130 beats/min), no dependence on electronic backup pacing, and enhanced modulation of pacemaker function during circadian rhythm or epinephrine infusion. In vitro isolated LV of dogs overexpressing SkM1 manifested a significantly more negative AP threshold. CONCLUSIONS: LBB-injected HCN2/SkM1 potentially provides a more clinically suitable biological pacemaker strategy than other reported constructs. This superiority is attributable to the more negative AP threshold and injection into the LBB.


Assuntos
Técnicas de Transferência de Genes , Sistema de Condução Cardíaco , Frequência Cardíaca/fisiologia , Canais Iônicos/genética , Proteínas Musculares/genética , Animais , Cães , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização
9.
Heart Rhythm ; 9(11): 1865-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22820054

RESUMO

BACKGROUND: Left ventricular pacing (LVP) in canine heart alters ventricular activation, leading to reduced transient outward potassium current (I(to)), loss of the epicardial action potential notch, and T-wave vector displacement. These repolarization changes, referred to as cardiac memory, are initiated by locally increased angiotensin II (AngII) levels. In HEK293 cells in which Kv4.3 and KChIP2, the channel subunits contributing to I(to), are overexpressed with the AngII receptor 1 (AT1R), AngII induces a decrease in I(to) as the result of internalization of a Kv4.3/KChIP2/AT1R macromolecular complex. OBJECTIVE: To test the hypothesis that in canine heart in situ, 2h LVP-induced decreases in membrane KChIP2, AT1R, and I(to) are prevented by blocking subunit trafficking. METHODS: We used standard electrophysiological, biophysical, and biochemical methods to study 4 groups of dogs: (1) Sham, (2) 2h LVP, (3) LVP + colchicine (microtubule-disrupting agent), and (4) LVP + losartan (AT1R blocker). RESULTS: The T-wave vector displacement was significantly greater in LVP than in Sham and was inhibited by colchicine or losartan. Epicardial biopsies showed significant decreases in KChIP2 and AT1R proteins in the membrane fraction after LVP but not after sham treatment, and these decreases were prevented by colchicine or losartan. Colchicine but not losartan significantly reduced microtubular polymerization. In isolated ventricular myocytes, AngII-induced I(to) reduction and loss of action potential notch were blocked by colchicine. CONCLUSIONS: LVP-induced reduction of KChIP2 in plasma light membranes depends on an AngII-mediated pathway and intact microtubular status. Loss of I(to) and the action potential notch appear to derive from AngII-initiated trafficking of channel subunits.


Assuntos
Estimulação Cardíaca Artificial , Sistema de Condução Cardíaco/fisiologia , Losartan/farmacologia , Microtúbulos/metabolismo , Canais de Potássio/fisiologia , Receptores de Angiotensina/metabolismo , Adaptação Fisiológica/fisiologia , Análise de Variância , Animais , Biópsia , Western Blotting , Colchicina/farmacologia , Cães , Sistema de Condução Cardíaco/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos
10.
Cardiovasc Res ; 94(3): 450-9, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22374989

RESUMO

AIMS: Reentry accounts for most life-threatening arrhythmias, complicating myocardial infarction, and therapies that consistently prevent reentry from occurring are lacking. In this study, we compare antiarrhythmic effects of gene transfer of green fluorescent protein (GFP; sham), the skeletal muscle sodium channel (SkM1), the liver-specific connexin (Cx32), and SkM1/Cx32 in the subacute canine infarct. METHODS AND RESULTS: Immediately after ligation of the left anterior descending artery, viral constructs were implanted in the epicardial border zone (EBZ). Five to 7 days later, efficient restoration of impulse propagation (narrow QRS and local electrogram duration) occurred in SkM1, Cx32, and SkM1/Cx32 groups (P< 0.05 vs. GFP). Programmed electrical stimulation from the EBZ induced sustained ventricular tachycardia (VT)/ventricular fibrillation (VF) in 15/22 GFP dogs vs. 2/12 SkM1, 6/14 Cx32, and 8/10 SkM1/Cx32 (P< 0.05 SkM1 vs. GFP). GFP, SkM1, and SkM1/Cx32 had predominantly polymorphic VT/VF, whereas in Cx32 dogs, monomorphic VT predominated (P< 0.05 for Cx32 vs. GFP). Tetrazolium red staining showed significantly larger infarcts in Cx32- vs. GFP-treated animals (P< 0.05). CONCLUSION: Whereas SkM1 gene transfer reduces the incidence of inducible VT/VF, Cx32 therapy to improve gap junctional conductance results in larger infarct size, a different VT morphology, and no antiarrhythmic efficacy.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Conexinas/metabolismo , Junções Comunicantes/efeitos dos fármacos , Proteínas Musculares/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Canais de Sódio/metabolismo , Fibrilação Ventricular/tratamento farmacológico , Animais , Antiarrítmicos/uso terapêutico , Conexinas/genética , Cães , Estimulação Elétrica , Eletrocardiografia , Masculino , Camundongos , Proteínas Musculares/genética , Ratos , Canais de Sódio/genética , Fibrilação Ventricular/fisiopatologia , Proteína beta-1 de Junções Comunicantes
11.
Circ Arrhythm Electrophysiol ; 5(4): 831-40, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22722661

RESUMO

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel is largely inactivated, contributing to slow conduction and reentry. We have demonstrated that adenoviral delivery of the skeletal muscle Na(+) channel (SkM1) to epicardial border zones normalizes conduction and reduces induction of ventricular tachycardia/ventricular fibrillation. We now studied the impact of canine mesenchymal stem cells (cMSCs) in delivering SkM1. METHODS AND RESULTS: cMSCs were isolated and transfected with SkM1. Coculture experiments showed cMSC/SkM1 but not cMSC alone and maintained fast conduction at depolarized potentials. We studied 3 groups in the canine 7d infarct: sham, cMSC, and cMSC/SkM1. In vivo epicardial border zones electrograms were broad and fragmented in sham, narrower in cMSCs, and narrow and unfragmented in cMSC/SkM1 (P<0.05). During programmed electrical stimulation of epicardial border zones, QRS duration in cMSC/SkM1 was shorter than in cMSC and sham (P<0.05). Programmed electrical stimulation-induced ventricular tachycardia/ventricular fibrillation was equivalent in all groups (P>0.05). CONCLUSION: cMSCs provide efficient delivery of SkM1 current. The interventions performed (cMSCs or cMSC/SkM1) were neither antiarrhythmic nor proarrhythmic. Comparing outcomes with cMSC/SkM1 and viral gene delivery highlights the criticality of the delivery platform to SkM1 antiarrhythmic efficacy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Proteínas Musculares/metabolismo , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Taquicardia Ventricular/prevenção & controle , Fibrilação Ventricular/prevenção & controle , Potenciais de Ação , Animais , Animais Recém-Nascidos , Estimulação Cardíaca Artificial , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Cães , Técnicas Eletrofisiológicas Cardíacas , Humanos , Proteínas Musculares/genética , Infarto do Miocárdio/complicações , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Ratos , Ratos Sprague-Dawley , Canais de Sódio/genética , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Transfecção , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/genética , Fibrilação Ventricular/metabolismo , Fibrilação Ventricular/fisiopatologia
12.
Cardiovasc Res ; 89(1): 41-50, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20823275

RESUMO

AIMS: acute myocardial ischaemia induces a decrease in resting membrane potential [which leads to reduction of action potential (AP) V(max)] and intracellular acidification (which closes gap junctions). Both contribute to conduction slowing. We hypothesized that ventricular expression of the skeletal muscle Na(+) channel, Nav1.4 (which activates fully at low membrane potentials), or connexin32 (Cx32, which is less pH-sensitive than connexin43) would support conduction and be antiarrhythmic. We tested this hypothesis in a murine model of ischaemia and reperfusion arrhythmias. METHODS AND RESULTS: empty adenovirus (Sham) or adenoviral constructs expressing either SkM1 (gene encoding Nav1.4) or Cx32 genes were injected into the left ventricular wall. Four days later, ventricular tachycardia (VT) occurred during reperfusion following a 5 min coronary occlusion. In Nav1.4- and Cx32-expressing mice, VT incidence and duration were lower than in Sham (P < 0.05). In vitro multisite microelectrode mapping was performed in the superfused right ventricular wall. To simulate ischaemic conditions, [K(+)] in solution was increased to 10 mmol/L and/or pH was decreased to 6.0. Western blots revealed Cx32 and Nav1.4 expression in both ventricles. Nav1.4 APs showed higher V(max) and conduction velocity (CV) than Shams at normal and elevated [K(+)]. Exposure of tissue to acid solution reduced intracellular pH to 6.4. There was no difference in CV between Sham and Cx32 groups in control solution. Acid solution slowed CV in Sham (P < 0.05) but not in Cx32. CONCLUSION: Nav1.4 or Cx32 expression preserved normal conduction in murine hearts and decreased the incidence of reperfusion VT.


Assuntos
Arritmias Cardíacas/prevenção & controle , Conexinas/genética , Conexinas/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Canais de Sódio/genética , Canais de Sódio/fisiologia , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Modelos Animais de Doenças , Eletrocardiografia , Expressão Gênica , Sistema de Condução Cardíaco/fisiopatologia , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Potássio/metabolismo , Ratos , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/prevenção & controle , Proteína beta-1 de Junções Comunicantes
13.
Heart Rhythm ; 7(7): 964-70, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20346417

RESUMO

BACKGROUND: Left ventricular pacing (LVP) to induce cardiac memory (CM) in dogs results in a decreased transient outward K current (I(to)) and reduced mRNA and protein of the I(to) channel accessory subunit, KChIP2. The KChIP2 decrease is attributed to a decrease in its transcription factor, cyclic adenosine monophosphate response element binding protein (CREB). OBJECTIVE: This study sought to determine the mechanisms responsible for the CREB decrease that is initiated by LVP. METHODS: CM was quantified as T-wave vector displacement in 18 LVP dogs. In 5 dogs, angiotensin II receptor blocker, saralasin, was infused before and during pacing. In 3 dogs, proteasomal inhibitor, lactacystin, was injected into the left anterior descending artery before LVP. Epicardial biopsy samples were taken before and after LVP. Neonatal rat cardiomyocytes (NRCM) were incubated with H(2)O(2) (50 micromol/l) for 1 hour with or without lactacystin. RESULTS: LVP significantly displaced the T-wave vector and was associated with increased lipid peroxidation and increased tissue angiotensin II levels. Saralasin prevented T-vector displacement and lipid peroxidation. CREB was significantly decreased after 2 hours of LVP and was comparably decreased in H(2)O(2)-treated NRCM. Lactacystin inhibited the CREB decrease in LVP dogs and H(2)O(2)-treated NRCM. LVP and H(2)O(2) both induced CREB ubiquitination, and the H(2)O(2)-induced CREB decrease was prevented by knocking down ubiquitin. CONCLUSION: LVP initiates myocardial angiotensin II production and reactive oxygen species synthesis, leading to CREB ubiquitination and its proteasomal degradation. This sequence of events would explain the pacing-induced reduction in KChIP2, and contribute to altered repolarization and the T-wave changes of cardiac memory.


Assuntos
Estimulação Cardíaca Artificial , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Sistema de Condução Cardíaco/metabolismo , Proteínas Interatuantes com Canais de Kv/análise , Função Ventricular Esquerda/fisiologia , Potenciais de Ação/fisiologia , Angiotensina II/fisiologia , Animais , Arritmias Cardíacas/metabolismo , Western Blotting , Células Cultivadas , Cães , Canais Iônicos/fisiologia , Peroxidação de Lipídeos , Masculino , Modelos Animais , Modelos Cardiovasculares , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Estresse Oxidativo/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina/fisiologia , Ubiquitinação , Remodelação Ventricular/fisiologia
14.
Heart Rhythm ; 7(8): 1104-10, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20385252

RESUMO

BACKGROUND: Skeletal muscle sodium channel (Nav1.4) expression in border zone myocardium increases action potential upstroke velocity in depolarized isolated tissue. Because resting membrane potential in the 1-week canine infarct is reduced, we hypothesized that conduction velocity (CV) is greater in Nav1.4 dogs compared with in control dogs. OBJECTIVE: The purpose of this study was to measure CV in the infarct border zone border in dogs with and without Nav1.4 expression. METHODS: Adenovirus was injected in the infarct border zone in 34 dogs. The adenovirus incorporated the Nav1.4- and a green fluorescent protein (GFP) gene (Nav1.4 group, n = 16) or only GFP (n = 18). After 1 week, upstroke velocity and CV were measured by sequential microelectrode recordings at 4 and 7 mM [K(+)] in superfused epicardial slabs. High-density in vivo epicardial activation mapping was performed in a subgroup (8 Nav1.4, 6 GFP) at three to four locations in the border zone. Microscopy and antibody staining confirmed GFP or Nav1.4 expression. RESULTS: Infarct sizes were similar between groups (30.6% +/- 3% of left ventricle mass, mean +/- standard error of the mean). Longitudinal CV was greater in Nav1.4 than in GFP sites (58.5 +/- 1.8 vs. 53.3 +/- 1.2 cm/s, 20 and 15 sites, respectively; P <.05). Transverse CV was not different between the groups. In tissue slabs, dV/dt(max) was higher and CV was greater in Nav1.4 than in control at 7 mM [K(+)] (P <.05). Immunohistochemical Nav1.4 staining was seen at the longitudinal ends of the myocytes. CONCLUSION: Nav1.4 channels in myocardium surviving 1 week infarction increases longitudinal but not transverse CV, consistent with the increased dV/dt(max) and with the cellular localization of Nav1.4.


Assuntos
Sistema de Condução Cardíaco/fisiopatologia , Músculo Esquelético/fisiologia , Infarto do Miocárdio/fisiopatologia , Canais de Sódio/biossíntese , Potenciais de Ação , Animais , Modelos Animais de Doenças , Cães
15.
Heart Rhythm ; 6(4): 564-70, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19324320

RESUMO

Cardiac memory is a form of electrophysiological remodeling generally considered benign, although it shares transduction pathways with factors that may be pathological, such as angiotensin II and reactive oxygen species. When induced by electrical pacing, memory provides a window into the mechanisms engaged during cardiac device therapy. Emphasis is placed on the complexity of signaling processes occurring downstream to the simple intervention of cardiac pacing and the relationship of resultant ion channel changes to their expression in action potentials and body surface recordings.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/fisiopatologia , Proteína de Ligação a CREB/fisiologia , Sistema de Condução Cardíaco/fisiologia , Coração/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Mapeamento Potencial de Superfície Corporal , Estimulação Cardíaca Artificial , Conexinas/farmacologia , Eletrocardiografia , Canais Iônicos/fisiologia , Modelos Cardiovasculares , Contração Miocárdica/fisiologia , Vetorcardiografia
16.
Heart Rhythm ; 6(3): 370-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19251214

RESUMO

BACKGROUND: Four voltage-gated potassium currents, I(to,f) (K(V)4.2), I(to,s) (K(V)1.4), I(K,slow) (K(V)1.5+K(V)2.1), and I(SS) (TASK1), govern murine ventricular repolarization. Although the accessory subunit KChIP2 influences I(to,f) expression, in preliminary experiments we found that action potential duration (APD) is maintained in KChIP2 knockout mice. OBJECTIVE: We tested the role of KChIP2 in regulating APD and studied the underlying ionic currents. METHODS: We used microelectrode techniques, whole-cell patch clamp studies, and real-time polymerase chain reaction amplification to characterize ventricular repolarization and its determinants in wild-type and KChIP2(-/-) mice. RESULTS: Despite comparable baseline action potentials, APD was more markedly prolonged by 4-aminopyridine (4-AP) in KChIP2(-/-) preparations. Peak K(+) current densities were similar in wild-type and KChIP2(-/-) cells (mean +/- SEM I(P): 28.3 +/- 2 (n = 27) vs. 29.2 +/- 2 pA/pF (n = 24), respectively; P > .05). Heteropodatoxin-2 (HpTx-2, 1 microM) had no effect on current amplitude in KChIP2(-/-) myocytes. The current fractions sensitive to 4-AP (50 microM and 1 mM) were larger in KChIP2(-/-) than wild-type (P < .05). Real-time polymerase chain reaction showed absence of KChIP2 and increased K(V)1.5 expression in KChIP2(-/-) ventricular myocardium. CONCLUSION: KChIP2 deficiency eliminated HpTx-2-sensitive I(to,f), but had little impact on total APD, secondary to upregulation of 4-AP-sensitive I(K,slow) in association with increased K(V)1.5 expression. There is increased sensitivity to 4-AP-mediated APD prolongation in KChIP2(-/-). Thus, KChIP2 seems important for murine repolarization in circumstances of reduced repolarization reserve.


Assuntos
Potenciais de Ação/fisiologia , Proteínas Interatuantes com Canais de Kv/fisiologia , Miócitos Cardíacos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Técnicas Eletrofisiológicas Cardíacas , Ventrículos do Coração , Técnicas In Vitro , Proteínas Interatuantes com Canais de Kv/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microeletrodos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Biol Chem ; 283(25): 17009-19, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18378685

RESUMO

Many growth regulatory stimuli promote cAMP response element-binding protein (CREB) Ser(133) phosphorylation, but the physiologically relevant CREB-Ser(133) kinase(s) in the heart remains uncertain. This study identifies a novel role for protein kinase D (PKD) as an in vivo cardiac CREB-Ser(133) kinase. We show that thrombin activates a PKCdelta-PKD pathway leading to CREB-Ser(133) phosphorylation in cardiomyocytes and cardiac fibroblasts. alpha(1)-Adrenergic receptors also activate a PKCdelta-PKD-CREB-Ser(133) phosphorylation pathway in cardiomyocytes. Of note, while the epidermal growth factor (EGF) promotes CREB-Ser(133) phosphorylation via an ERK-RSK pathway in cardiac fibroblasts, the thrombin-dependent EGFR transactivation pathway leading to ERK-RSK activation does not lead to CREB-Ser(133) phosphorylation in this cell type. Adenoviral-mediated overexpression of PKCdelta (but not PKCepsilon or PKCalpha) activates PKD; PKCdelta and PKD1-S744E/S748E overexpression both promote CREB-Ser(133) phosphorylation. Pasteuralla multocida toxin (PMT), a direct Galpha(q) agonist that induces robust cardiomyocyte hypertrophy, also activates the PKD-CREB-Ser(133) phosphorylation pathway, leading to the accumulation of active PKD and Ser(133)-phosphorylated CREB in the nucleus, activation of a CRE-responsive promoter, and increased Bcl-2 (CREB target gene) expression in cardiomyocyte cultures. Cardiac-specific Galpha(q) overexpression also leads to an increase in PKD-Ser(744)/Ser(748) and CREB-Ser(133) phosphorylation as well as increased Bcl-2 protein expression in the hearts of transgenic mice. Collectively, these studies identify a novel Galpha(q)-PKCdelta-PKD-CREB-Ser(133) phosphorylation pathway that is predicted to contribute to cardiac remodeling and could be targeted for therapeutic advantage in the setting of heart failure phenotypes.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Miocárdio/metabolismo , Proteína Quinase C/metabolismo , Animais , Ativação Enzimática , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Pasteurella multocida/metabolismo , Fosforilação , Ratos , Ratos Wistar , Ativação Transcricional
18.
Nephrol Dial Transplant ; 19(9): 2198-207, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15266032

RESUMO

BACKGROUND: Haemodialysis therapy does not provide renal tubule function, such as active fluid and solute transport, nor metabolic or endocrine action. Moreover, this treatment is usually associated with serious complications and high mortality. We constructed a bioartificial renal tubule device by using renal tubule epithelial cells in an artificial membrane, and evaluated transport properties of the device for 2 weeks. METHODS: A renal epithelial cell line, LLC-PK(1) (Lewis-lung cancer porcine kidney), was seeded on polysulfone hollow fibres in small and large modules. We studied perfusion and leakage of urea nitrogen (UN) and creatinine (Cr), as well as reabsorption of water, glucose and sodium for a period of 2 weeks. RESULTS: Cell-lined hollow fibre membranes significantly reduced the leakage of UN and Cr throughout the 2 week period. Reabsorption of water, glucose and sodium were adequate from days 3 to 10 and gradually decreased thereafter. LLC-PK(1) cells actively transported these substances. Scanning electron microscopy revealed that cells in the hollow fibres on day 8 became completely confluent. However, they became multi-layered and almost obstructed the hollow fibres on day 13. CONCLUSIONS: This bioartificial renal tubule device functioned to reabsorb water, glucose and sodium for approximately 10 days. This is the first report of successful long-term evaluation of a bioartificial renal tubule device. This device, in combination with continuous haemofiltration, may provide treatment to prevent complications of dialysis and raise the quality of life in chronic renal failure patients.


Assuntos
Rins Artificiais , Membranas Artificiais , Animais , Transporte Biológico/fisiologia , Glucose/farmacocinética , Túbulos Renais/fisiologia , Células LLC-PK1 , Sódio/farmacocinética , Suínos , Água/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa