Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1838(2): 665-73, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23680626

RESUMO

The cytoskeleton and cardiac ion channel expression are closely linked. From the time that newly synthesized channels exit the endoplasmic reticulum, they are either traveling along the microtubule or actin cytoskeletons or likely anchored in the plasma membrane or in internal vesicular pools by those scaffolds. Molecular motors, small GTPases and even the dynamics of the cytoskeletons themselves influence the trafficking and expression of the channels. In some cases, the functioning of the channels themselves has profound influences on the cytoskeleton. Here we provide an overview of the current state of knowledge on the involvement of the actin and microtubule cytoskeletons in the trafficking, targeting and expression of cardiac ion channels and a few channels expressed elsewhere. We highlight, also, some of the many questions that remain about these processes. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Assuntos
Citoesqueleto/metabolismo , Coração/fisiologia , Canais Iônicos/metabolismo , Microtúbulos/metabolismo , Animais , Humanos
2.
Methods Mol Biol ; 940: 145-55, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23104340

RESUMO

Transfection of mammalian cells has long been an extremely powerful approach for the study of the effects of specific gene expression on cell function. Until recently, however, this approach has been unavailable for the study of gene function in adult cardiac myocytes. Here, an adaptation of the biolistic method to the transfection of adult cardiac myocytes is described. DNA is precipitated onto gold particles in the absence of PVP and the particles are biolistically delivered to freshly isolated adult rat cardiomyocytes via a Bio-Rad Helios System gene gun. The myocytes are cultured in the absence of bovine serum albumin and expression of the introduced genes, in phenotypically intact myocytes, is robust within 12-24 h.


Assuntos
Biolística/instrumentação , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Transfecção/instrumentação , Animais , Separação Celular , DNA/administração & dosagem , DNA/química , DNA/genética , Microesferas , Ratos
3.
Am J Physiol Cell Physiol ; 303(9): C963-76, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22914645

RESUMO

The roles of several small GTPases in the expression of an endogenous potassium current, I(to,f), in adult rat ventricular myocytes have been investigated. The results indicate that forward trafficking of newly synthesized Kv4.2, which underlies I(to,f) in these cells, requires both Rab1 and Sar1 function. Expression of a Rab1 dominant negative (DN) reduced I(to,f) current density by roughly one-half relative to control, mCherry-transfected myocytes. Similarly, expression of a Sar1DN nearly halved I(to,f) current density. Rab11 is not essential to trafficking of Kv4.2, as expression of a Rab11DN had no effect on I(to,f) over the time frames investigated here. In a process dependent on intact endoplasmic reticulum (ER)-to-Golgi transport, however, overexpression of wild-type Rab11 resulted in a doubling of I(to,f) density; block of ER-to-Golgi traffic by Brefeldin A completely abrogated the effect. Also implicated in the trafficking of Kv4.2 are Rab5 and Rab4. Rab5DN expression increased endogenous I(to,f) by two- to threefold, nonadditively with inhibition of dynamin-dependent endocytosis. And, in a phenomenon similar to that previously reported for myoblast-expressed Kv1.5, Rab4DN expression roughly doubled endogenous peak transient currents. Colocalization experiments confirmed the involvement of Rab4 in postinternalization trafficking of Kv4.2. There was little role evident for the lysosome in the degradation of internalized Kv4.2, as overexpression of neither wild-type nor DN isoforms of Rab7 had any effect on I(to,f). Instead, degradation may depend largely on the proteasome; the proteasome inhibitor MG132 significantly increased I(to,f) density.


Assuntos
Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Transporte Proteico/fisiologia , Canais de Potássio Shal/metabolismo , Animais , Brefeldina A/farmacologia , Células Cultivadas , Retículo Endoplasmático/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Leupeptinas/farmacologia , Masculino , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteassoma/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar , Proteínas rab1 de Ligação ao GTP/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
4.
Physiol Rev ; 92(3): 1317-58, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22811429

RESUMO

Cardiac myocytes are characterized by distinct structural and functional entities involved in the generation and transmission of the action potential and the excitation-contraction coupling process. Key to their function is the specific organization of ion channels and transporters to and within distinct membrane domains, which supports the anisotropic propagation of the depolarization wave. This review addresses the current knowledge on the molecular actors regulating the distinct trafficking and targeting mechanisms of ion channels in the highly polarized cardiac myocyte. In addition to ubiquitous mechanisms shared by other excitable cells, cardiac myocytes show unique specialization, illustrated by the molecular organization of myocyte-myocyte contacts, e.g., the intercalated disc and the gap junction. Many factors contribute to the specialization of the cardiac sarcolemma and the functional expression of cardiac ion channels, including various anchoring proteins, motors, small GTPases, membrane lipids, and cholesterol. The discovery of genetic defects in some of these actors, leading to complex cardiac disorders, emphasizes the importance of trafficking and targeting of ion channels to cardiac function. A major challenge in the field is to understand how these and other actors work together in intact myocytes to fine-tune ion channel expression and control cardiac excitability.


Assuntos
Comunicação Celular , Membrana Celular/metabolismo , Canais Iônicos/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Potenciais de Ação , Animais , Comunicação Celular/genética , Acoplamento Excitação-Contração , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Canais Iônicos/genética , Cinética , Metabolismo dos Lipídeos , Mutação , Transporte Proteico , Sarcolema/metabolismo , Transdução de Sinais/genética
5.
Circ Res ; 110(6): 802-4, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22427322
6.
Trends Pharmacol Sci ; 31(12): 587-95, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20951446

RESUMO

The K(2P) potassium channels are responsible for the background conductance observed in several tissues. Their ubiquitous localization and thus their potential implications in diseases have led to increased research on these channels over the last few years. In this review, we outline different aspects of the research on K(2P) channels and highlight some of the latest discoveries in this area. We focus on research into K(2P) channels as potential therapeutic targets in ischemia/hypoxia, depression, memory disorders, pain, cardiovascular disease and disorders of the immune system. We address the challenge of developing novel pharmacological compounds to target these channels. We also discuss the regulation of expression of the K(2P) gene in health and disease, as well as the value of assessing the expression of K(2P) channels as potential biomarkers of disease.


Assuntos
Doenças Cardiovasculares/metabolismo , Infertilidade Masculina/metabolismo , Transtornos Mentais/metabolismo , Neoplasias/metabolismo , Bloqueadores dos Canais de Potássio/uso terapêutico , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Animais , Sistemas de Liberação de Medicamentos/métodos , Dosagem de Genes , Humanos , Masculino , Camundongos , Camundongos Knockout , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/genética , Canais de Potássio/metabolismo
7.
Expert Rev Cardiovasc Ther ; 8(7): 965-79, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20602558

RESUMO

Cardiac arrhythmias are a leading cause of morbidity and mortality in the Western world. Ventricular arrhythmias are reportedly responsible for the majority of sudden cardiac deaths and atrial fibrillation is responsible for 15% of all strokes in the USA. Recent evidence suggests a role for cholesterol in the development of these arrhythmias. In addition to its association with atherosclerotic plaques, high cholesterol has been shown to cause changes in membrane properties, including the function of hormone receptors, ion channels and pumps. These effects are mediated through direct interactions between cholesterol and the membrane proteins, through changes in membrane fluidity and/or an association with lipid rafts. Cholesterol-lowering therapy, therefore, may prove an effective method for the treatment of cardiac arrhythmias. Statins, a class of cholesterol-lowering drugs, have been frequently shown to protect against ventricular arrhythmias and atrial fibrillation. Some of this protection may stem from their cholesterol-lowering activities.


Assuntos
Arritmias Cardíacas/etiologia , Colesterol/sangue , Animais , Anticolesterolemiantes/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/prevenção & controle , Fibrilação Atrial/prevenção & controle , Membrana Celular/fisiologia , Colesterol/biossíntese , Colesterol/metabolismo , Ventrículos do Coração , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Canais Iônicos/metabolismo , Bombas de Íon/metabolismo , Fluidez de Membrana , Microdomínios da Membrana/fisiologia , Miocárdio/metabolismo , Transporte Proteico , Receptores de Superfície Celular/metabolismo
8.
Am J Physiol Cell Physiol ; 298(6): C1343-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20357183

RESUMO

The transfection of cardiac myocytes is difficult, and so most of the data regarding the regulation of trafficking and targeting of cardiac ion channels have been obtained using heterologous expression systems. Here we apply the fast biolistic transfection procedure to adult cardiomyocytes to show that biolistically introduced exogenous voltage-gated potassium channel, Kv1.5, is functional and, like endogenous Kv1.5, localizes to the intercalated disc, where it is expressed at the surface of that structure. Transfection efficiency averages 28.2 +/- 5.7% of surviving myocytes at 24 h postbombardment. Ventricular myocytes transfected with a tagged Kv1.5 exhibit an increased sustained current component that is approximately 40% sensitive to 100 microM 4-aminopyridine and which is absent in myocytes transfected with a fluorescent protein-encoding construct alone. Kv1.5 deletion mutations known to reduce the surface expression of the channel in heterologous cells similarly reduce the surface expression in transfected ventricular myocytes, although targeting to the intercalated disc per se is generally unaffected by both NH(2)- and COOH-terminal deletion mutants. Expressed current levels in wild-type Kv1.5, Kv1.5DeltaSH3(1), Kv1.5DeltaN209, and Kv1.5DeltaN135 mutants were well correlated with apparent surface expression of the channel at the intercalated disc. Our results conclusively demonstrate functionality of channels present at the intercalated disc in native myocytes and identify determinants of trafficking and surface targeting in intact cells. Clearly, biolistic transfection of adult cardiac myocytes will be a valuable method to study the regulation of surface expression of channels in their native environment.


Assuntos
Biolística , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/metabolismo , Potássio/metabolismo , Transfecção/métodos , 4-Aminopiridina/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Humanos , Junções Intercelulares/metabolismo , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Lipídeos , Masculino , Potenciais da Membrana , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Transporte Proteico , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
9.
J Physiol ; 587(Pt 19): 4565-74, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19675065

RESUMO

We have investigated the role of the kinesin I isoform Kif5b in the trafficking of a cardiac voltage-gated potassium channel, Kv1.5. In Kv1.5-expressing HEK293 cells and H9c2 cardiomyoblasts, current densities were increased from control levels of 389 +/- 50.0 and 317 +/- 50.3 pA pF(1), respectively, to 614 +/- 74.3 and 580 +/- 90.9 pA pF(1) in cells overexpressing the Kif5b motor. Overexpression of the Kif5b motor increased Kv1.5 expression additively with several manipulations that reduce channel internalization, suggesting that it is involved in the delivery of the channel to the cell surface. In contrast, expression of a Kif5b dominant negative (Kif5bDN) construct increased Kv1.5 expression non-additively with these manipulations. Thus, the dominant negative acts by indirectly inhibiting endocytosis. The increase in Kv1.5 currents induced by wild-type Kif5b was dependent on Golgi function; a 6 h treatment with Brefeldin A reduced Kv1.5 currents to control levels in Kif5b-overexpressing cells but had little effect on the increase associated with Kif5bDN expression. Finally, expression of the Kif5bDN prior to induction of Kv1.5 in a tetracycline inducible system blocked surface expression of the channel in both HEK293 cells and H9c2 cardiomyoblasts. Thus, Kif5b is essential to anterograde trafficking of a cardiac voltage-gated potassium channel.


Assuntos
Cinesinas/metabolismo , Canal de Potássio Kv1.5/metabolismo , Mioblastos Cardíacos/metabolismo , Brefeldina A/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Expressão Gênica , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinesinas/genética , Canal de Potássio Kv1.5/genética , Potenciais da Membrana , Microtúbulos/metabolismo , Mioblastos Cardíacos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Transfecção
10.
Am J Physiol Heart Circ Physiol ; 296(1): H71-83, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18978193

RESUMO

Potassium channels at the cardiomyocyte surface must eventually be internalized and degraded, and changes in cardiac potassium channel expression are known to occur during myocardial disease. It is not known which trafficking pathways are involved in the control of cardiac potassium channel surface expression, and it is not clear whether all cardiac potassium channels follow a common pathway or many pathways. In the present study we have surveyed the role of retrograde microtubule-dependent transport in modulating the surface expression of several cardiac potassium channels in ventricular myocytes and heterologous cells. The disruption of microtubule transport in rat ventricular myocytes with nocodazole resulted in significant changes in potassium currents. A-type currents were enhanced 1.6-fold at +90 mV, rising from control densities of 20.9 +/- 2.8 to 34.0 +/- 5.4 pA/pF in the nocodazole-treated cells, whereas inward rectifier currents were reduced by one-third, perhaps due to a higher nocodazole sensitivity of Kir channel forward trafficking. These changes in potassium currents were associated with a significant decrease in action potential duration. When expressed in heterologous human embryonic kidney (HEK-293) cells, surface expression of Kv4.2, known to substantially underlie A-type currents in rat myocytes, was increased by nocodazole, by the dynein inhibitor erythro-9-(2-hydroxy-3-nonyl) adenine hydrochloride, and by p50 overexpression, which specifically interferes with dynein motor function. Peak current density was 360 +/- 61.0 pA/pF in control cells and 658 +/- 94.5 pA/pF in cells overexpressing p50. The expression levels of Kv2.1, Kv3.1, human ether-a-go-go-related gene, and Kir2.1 were similarly increased by p50 overexpression in this system. Thus the regulation of potassium channel expression involves a common dynein-dependent process operating similarly on the various channels.


Assuntos
Citoesqueleto/fisiologia , Dineínas/fisiologia , Coração/fisiologia , Microtúbulos/fisiologia , Canais de Potássio/biossíntese , Adenina/análogos & derivados , Adenina/farmacologia , Antineoplásicos/farmacologia , Transporte Biológico Ativo/efeitos dos fármacos , Biotinilação , Canais de Cloro CLC-2 , Linhagem Celular , Canais de Cloreto/biossíntese , Canais de Cloreto/genética , Eletrofisiologia , Endopeptidase K/metabolismo , Ventrículos do Coração , Humanos , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Miócitos Cardíacos/fisiologia , Subunidade p50 de NF-kappa B/biossíntese , Nocodazol/farmacologia , Plasmídeos/genética , Receptores de Superfície Celular/metabolismo , Transfecção
11.
J Physiol ; 586(20): 4793-813, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18755741

RESUMO

Little is known about the postinternalization trafficking of surface-expressed voltage-gated potassium channels. Here, for the first time, we investigate into which of four major trafficking pathways a voltage-gated potassium channel is targeted after internalization. In both a cardiac myoblast cell line and in HEK293 cells, channels were found to internalize and to recycle quickly. Upon internalization, Kv1.5 rapidly associated with Rab5-and Rab4-positive endosomes, suggesting that the channel is internalized via a Rab5-dependent pathway and rapidly targeted for recycling to the plasma membrane. Nevertheless, as indicated by colocalization with Rab7, a fraction of the channels are targeted for degradation. Recycling through perinuclear endosomes is limited; colocalization with Rab11 was evident only after 24 h postsurface labelling. Expression of dominant negative (DN) Rab constructs significantly increased Kv1.5 functional expression. In the myoblast line, Rab5DN increased Kv1.5 current densities to 1305 +/- 213 pA pF(-1) from control 675 +/- 81.6 pA pF(-1). Rab4DN similarly increased Kv1.5 currents to 1382 +/- 155 pA pF(-1) from the control 522 +/- 82.7 pA pF(-1) at +80 mV. Expression of the Rab7DN increased Kv1.5 currents 2.5-fold in HEK293 cells but had no significant effect in H9c2 myoblasts, and, unlike the other Rab GTPases tested, over-expression of wild-type Rab7 decreased Kv1.5 currents in the myoblast line. Densities fell to 573 +/- 96.3 pA pF(-1) from the control 869 +/- 135.5 pA pF(-1). The Rab11DN was slow to affect Kv1.5 currents but had comparable effects to other dominant negative constructs after 48 h. With the exception of Rab11DN and nocodazole, the effects of interference with microtubule-dependent trafficking by nocodazole or p50 overexpression were not additive with the Rab dominant negatives. The Rab GTPases thus constitute dynamic targets by which cells may modulate Kv1.5 functional expression.


Assuntos
Ativação do Canal Iônico/fisiologia , Rim/metabolismo , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Linhagem Celular , Humanos , Transporte Proteico/fisiologia
12.
J Physiol ; 582(Pt 1): 17-26, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17412767

RESUMO

The regulation of ion channels involves more than just modulation of their synthesis and kinetics, as controls on their trafficking and localization are also important. Although the body of knowledge is fairly large, the entire trafficking pathway is not known for any one channel. This review summarizes current knowledge on the trafficking of potassium channels that are expressed in the heart. Our knowledge of channel assembly, trafficking through the Golgi apparatus and on to the surface is covered, as are controls on channel surface retention and endocytosis.


Assuntos
Endocitose , Complexo de Golgi/metabolismo , Miocárdio/metabolismo , Canais de Potássio/metabolismo , Sarcolema/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Retículo Endoplasmático/metabolismo , Guanilato Quinases/metabolismo , Humanos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Miocárdio/enzimologia , Canais de Potássio/química , Conformação Proteica , Transporte Proteico , Sarcolema/enzimologia
13.
Biochem Biophys Res Commun ; 342(1): 1-8, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16466689

RESUMO

We have previously reported that SAP97 enhancement of hKv1.5 currents requires an intact Kv1.5 N-terminus and is independent of the PDZ-binding motif at the C-terminus of the channel [J. Eldstrom, W.S. Choi, D.F. Steele, D. Fedida, SAP97 increases Kv1.5 currents through an indirect N-terminal mechanism, FEBS Lett. 547 (2003) 205-211]. Here, we report that an interaction between the two proteins can be detected under certain conditions but their interaction is irrelevant to the enhancement of channel expression. Instead, a threonine residue at position 15 in the hKv1.5 N-terminus is critically important. Mutation of this residue, which lies within a consensus site for phosphorylation by protein kinase C, to an alanine, completely abrogated the effect of SAP97 on channel expression. Although we were unable to detect phosphorylation of this residue, specific inhibition of kinase C by Calphostin C eliminated the increase in wild-type hKv1.5 currents associated with SAP97 overexpression suggesting a role for this kinase in the response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/metabolismo , Proteínas de Membrana/metabolismo , Regulação para Cima , Sequência de Aminoácidos , Linhagem Celular , Proteína 1 Homóloga a Discs-Large , Eletrofisiologia , Humanos , Canal de Potássio Kv1.5/genética , Dados de Sequência Molecular , Mutação/genética , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Tetraciclina/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
Circ Res ; 97(4): 363-71, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-16051887

RESUMO

In this article we have investigated the mechanisms by which retrograde trafficking regulates the surface expression of the voltage-gated potassium channel, Kv1.5. Overexpression of p50/dynamitin, known to disrupt the dynein-dynactin complex responsible for carrying vesicle cargo, substantially increased outward K+ currents in HEK293 cells stably expressing Kv1.5 (0.57+/-0.07 nA/pF, n=12; to 1.18+/-0.2 nA/pF, n=12, P<0.01), as did treatment of the cells with a dynamin inhibitory peptide, which blocks endocytosis. Nocodazole pretreatment, which depolymerizes the microtubule cytoskeleton along which dynein tracks, also doubled Kv1.5 currents in HEK cells and sustained K+ currents in isolated rat atrial myocytes. These increased currents were blocked by 1 mmol/L 4-aminopyridine, and the specific Kv1.5 antagonist, DMM (100 nM). Confocal imaging of both HEK cells and myocytes, as well as experiments testing the sensitivity of the channel in living cells to external Proteinase K, showed that this increase of K+ current density was caused by a redistribution of channels toward the plasma membrane. Coimmunoprecipitation experiments demonstrated a direct interaction between Kv1.5 and the dynein motor complex in both heterologous cells and rat cardiac myocytes, supporting the role of this complex in Kv1.5 trafficking, which required an intact SH3-binding domain in the Kv1.5 N terminus to occur. These experiments highlight a pathway for Kv1.5 internalization from the cell surface involving early endosomes, followed by later trafficking by the dynein motor along microtubules. This work has significant implications for understanding the way Kv channel surface expression is regulated.


Assuntos
Dineínas/fisiologia , Endocitose , Miócitos Cardíacos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Sítios de Ligação , Células CHO , Linhagem Celular , Cricetinae , Endossomos/metabolismo , Humanos , Imunoprecipitação , Canal de Potássio Kv1.5 , Nocodazol/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Transporte Proteico , Ratos , Domínios de Homologia de src
15.
Circ Res ; 93(8): 744-51, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-14500335

RESUMO

Although the canine atrium has proven useful in several experimental models of atrial fibrillation and for studying the effects of rapid atrial pacing on atrial electrical remodeling, it may not fully represent the human condition because of reported differences in functional ionic currents and ion channel subunit expression. In this study, we reassessed the molecular components underlying one current, the ultrarapid delayed rectifier current in canine atrium [IKur(d)], by evaluating the mRNA, protein, immunofluorescence, and currents of the candidate channels. Using reverse transcriptase-polymerase chain reaction, we found that Kv1.5 mRNA was expressed in canine atrium whereas message for Kv3.1 was not detected. Western analysis on cytosolic and membrane fractions of canine tissues, using selective antibodies, showed that Kv3.1 was only detectable in the brain preparations, whereas Kv1.5 was expressed at high levels in both atrial and ventricular membrane fractions. Confocal imaging performed on isolated canine atrial myocytes clearly demonstrated the presence of Kv1.5 immunostaining, whereas that of Kv3.1 was equivocal. Voltage- and current-clamp studies showed that 0.5 mmol/L tetraethylammonium had variable effects on sustained K+ currents, whereas a compound with demonstrated selectivity for hKv1.5 versus Kv3.1, hERG or the sodium channel, fully suppressed canine atrial IKur tail currents and depressed sustained outward K+ current. This agent also increased action potential plateau potentials and action potential duration at 20% and 50% repolarization. These results suggest that in canine atria, as in other species including human, Kv1.5 protein is highly expressed and contributes to IKur.


Assuntos
Função Atrial , Miócitos Cardíacos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Potenciais de Ação , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cães , Condutividade Elétrica , Átrios do Coração/química , Átrios do Coração/citologia , Humanos , Canal de Potássio Kv1.5 , Miócitos Cardíacos/química , Miócitos Cardíacos/efeitos dos fármacos , Neuropeptídeos/análise , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/análise , Canais de Potássio Shaw
16.
FEBS Lett ; 547(1-3): 205-11, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12860415

RESUMO

The functional interaction of the voltage-gated potassium channel hKv1.5 with the PDZ domain containing protein SAP97 has been investigated. In marked contrast with the known dependence of SAP97-induced Kv1 potassium current down-regulation on the channel C-termini, SAP97 increased hKv1.5 current through an indirect interaction with the Kv1.5 N-terminus. Deletion of the Kv1.5 N-terminus eliminated the SAP97-mediated increase in potassium currents whereas deletion of the channel's C-terminal PDZ binding motif had no effect. In contrast with other Kv1-SAP97 interactions, no physical interaction could be detected in vivo or in vitro between the two proteins. The proteins did not co-localize in cardiac myocytes nor did they co-immunoprecipitate from transfected HEK cells. Yeast two-hybrid experiments also failed to detect any interaction between the two proteins, but in one experiment of six, Kv1.5 co-immunoprecipitated very inefficiently with SAP97 from rat ventricular myocytes. Thus, we conclude that the influence of SAP97 on Kv1.5 potassium current levels is dependent upon a novel regulatory mechanism.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Sítios de Ligação , Linhagem Celular , Proteína 1 Homóloga a Discs-Large , Coração/fisiologia , Átrios do Coração , Ventrículos do Coração , Humanos , Canal de Potássio Kv1.5 , Proteínas de Membrana , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/isolamento & purificação , Fragmentos de Peptídeos/isolamento & purificação , Fragmentos de Peptídeos/metabolismo , Canais de Potássio/isolamento & purificação , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transfecção , Domínios de Homologia de src
17.
FEBS Lett ; 531(3): 529-37, 2002 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-12435606

RESUMO

We have investigated the interactions of prototypical PDZ domains with both the C- and N-termini of Kv1.5 and other Kv channels. A combination of in vitro binding and yeast two-hybrid assays unexpectedly showed that PDZ domains derived from PSD95 bind both the C- and N-termini of the channels with comparable avidity. From doubly transfected HEK293 cells, Kv1.5 was found to co-immunoprecipitate with the PDZ protein, irrespective of the presence of the canonical C-terminal PDZ-binding motif in Kv1.5. Imaging analysis of the same HEK cell lines demonstrated that co-localization of Kv1.5 with PSD95 at the cell surface is similarly independent of the canonical PDZ-binding motif. Deletion analysis localized the N-terminal PDZ-binding site in Kv1.5 to the T1 region of the channel. Co-expression of PSD95 with Kv1.5 N- and C-terminal deletions in HEK cells had contrasting effects on the magnitudes of the potassium currents across the membranes of these cells. These findings may have important implications for the regulation of channel expression and function by PDZ proteins like PSD95.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Proteína 4 Homóloga a Disks-Large , Eletroforese em Gel de Poliacrilamida , Glicosilação , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Canal de Potássio Kv1.5 , Proteínas de Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio/química , Canais de Potássio/genética , Testes de Precipitina , Proteínas Recombinantes de Fusão/metabolismo , Técnicas do Sistema de Duplo-Híbrido
18.
J Biol Chem ; 277(32): 29045-53, 2002 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-12021261

RESUMO

The T1 domain is a cytosolic NH2-terminal domain present in all Kv (voltage-dependent potassium) channels, and is highly conserved between Kv channel subfamilies. Our characterization of a truncated form of Kv1.5 (Kv1.5deltaN209) expressed in myocardium demonstrated that deletion of the NH2 terminus of Kv1.5 imparts a U-shaped inactivation-voltage relationship to the channel, and prompted us to investigate the NH2 terminus as a regulatory site for slow inactivation of Kv channels. We examined the macroscopic inactivation properties of several NH2-terminal deletion mutants of Kv1.5 expressed in HEK 293 cells, demonstrating that deletion of residues up to the T1 boundary (Kv1.5deltaN19, Kv1.5deltaN91, and Kv1.5deltaN119) did not alter Kv1.5 inactivation, however, deletion mutants that disrupted the T1 structure consistently exhibited inactivation phenotypes resembling Kv1.5deltaN209. Chimeric constructs between Kv1.5 and the NH2 termini of Kv1.1 and Kv1.3 preserved the inactivation kinetics observed in full-length Kv1.5, again suggesting that the Kv1 T1 domain influences slow inactivation. Furthermore, disruption of intersubunit T1 contacts by mutation of residues Glu(131) and Thr(132) to alanines resulted in channels exhibiting a U-shaped inactivation-voltage relationship. Fusion of the NH2 terminus of Kv2.1 to the transmembrane segments of Kv1.5 imparted a U-shaped inactivation-voltage relationship to Kv1.5, whereas fusion of the NH2 terminus of Kv1.5 to the transmembrane core of Kv2.1 decelerated Kv2.1 inactivation and abolished the U-shaped voltage dependence of inactivation normally observed in Kv2.1. These data suggest that intersubunit T1 domain interactions influence U-type inactivation in Kv1 channels, and suggest a generalized influence of the T1 domain on U-type inactivation between Kv channel subfamilies.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/química , Sequência de Aminoácidos , Linhagem Celular , Canais de Potássio de Retificação Tardia , Eletrofisiologia , Deleção de Genes , Humanos , Cinética , Canal de Potássio Kv1.5 , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fenótipo , Mutação Puntual , Potássio/química , Canais de Potássio/genética , Canais de Potássio/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Canais de Potássio Shab , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...