Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 516(3): 777-783, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31253402

RESUMO

Mutations in gene SCN5A, which encodes cardiac voltage-gated sodium channel Nav1.5, are associated with multiple clinical phenotypes. Here we describe a novel A1294G genetic variant detected in a male patient with combined clinical phenotype including atrioventricular II block, Brugada-like ECG, septal fibrosis, right ventricular dilatation and decreased left ventricular contractility. Residue A1294 is located in the IIIS3-S4 extracellular loop, in proximity to several residues whose mutations are associated with sodium channelopathies. The wild-type channel Nav1.5 and mutant Nav1.5-A1294G were expressed in the CHO-K1 and HEK293T cells and whole-cell sodium currents were recorded using the patch-clamp method. The A1294G channels demonstrated a negative shift of steady-state inactivation, accelerated fast and slow inactivation and decelerated recovery from intermediate inactivation. Our study reveals biophysical mechanism of the Nav1.5-A1294G dysfunction, which may underlie the combined phenotypic manifestation observed in the patient.


Assuntos
Bloqueio Atrioventricular/genética , Síndrome de Brugada/genética , Predisposição Genética para Doença/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Mutação Puntual , Adulto , Animais , Bloqueio Atrioventricular/fisiopatologia , Síndrome de Brugada/fisiopatologia , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Técnicas de Patch-Clamp , Fenótipo
2.
Oncology ; 97(6): 373-382, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31430760

RESUMO

INTRODUCTION: Breast cancer is one of the leading causes of death worldwide and is the result of dysregulation of various signaling pathways in mammary epithelial cells. The mortality rate in patients suffering from breast cancer is high because the tumor cells have a prominent invasive capacity towards the surrounding tissues. Previous studies carried out in tumor cell models show that voltage-gated ion channels may be important molecular actors that contribute to the migratory and invasive capacity of the tumor cells. METHODS: In this study, by using an experimental strategy that combines cell and molecular biology assays with electrophysiological recording, we sought to determine whether the voltage-dependent sodium channel NaV1.5 regulates the migratory capacity of the human breast cancer cell line MDA-MB 231, when cells are maintained in the presence of epidermal growth factor (EGF), as an inductor of the epithelial-mesenchymal transition. RESULTS: Our data show that EGF stimulates the migratory capacity of MDA-MB 231 cells, by regulating the functional expression of NaV1.5 channels. Consistent with this, the stimulatory actions of the growth factor were prevented by the use of tetrodotoxin, an Na+ channel selective blocker, as well as by resveratrol, an antioxidant that can also affect Na+ channel activity. DISCUSSION: The understanding of molecular mechanisms, such as the EGF pathway in the progression of breast cancer is fundamental for the design of more effective therapeutic strategies for the disease.


Assuntos
Neoplasias da Mama/patologia , Fator de Crescimento Epidérmico/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal , Feminino , Humanos , Canal de Sódio Disparado por Voltagem NAV1.5/análise , Resveratrol/farmacologia
3.
Anesth Analg ; 127(3): 650-660, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29958221

RESUMO

BACKGROUND: Cardiotoxic effects of local anesthetics (LAs) involve inhibition of NaV1.5 voltage-gated Na channels. Metastatic breast and colon cancer cells also express NaV1.5, predominantly the neonatal splice variant (nNaV1.5) and their inhibition by LAs reduces invasion and migration. It may be advantageous to target cancer cells while sparing cardiac function through selective blockade of nNaV1.5 and/or by preferentially affecting inactivated NaV1.5, which predominate in cancer cells. We tested the hypotheses that lidocaine and levobupivacaine differentially affect (1) adult (aNaV1.5) and nNaV1.5 and (2) the resting and inactivated states of NaV1.5. METHODS: The whole-cell voltage-clamp technique was used to evaluate the actions of lidocaine and levobupivacaine on recombinant NaV1.5 channels expressed in HEK-293 cells. Cells were transiently transfected with cDNAs encoding either aNaV1.5 or nNaV1.5. Voltage protocols were applied to determine depolarizing potentials that either activated or inactivated 50% of maximum conductance (V½ activation and V½ inactivation, respectively). RESULTS: Lidocaine and levobupivacaine potently inhibited aNaV1.5 (IC50 mean [SD]: 20 [22] and 1 [0.6] µM, respectively) and nNaV1.5 (IC50 mean [SD]: 17 [10] and 3 [1.6] µM, respectively) at a holding potential of -80 mV. IC50s differed significantly between lidocaine and levobupivacaine with no influence of splice variant. Levobupivacaine induced a statistically significant depolarizing shift in the V½ activation for aNaV1.5 (mean [SD] from -32 [4.6] mV to -26 [8.1] mV) but had no effect on the voltage dependence of activation of nNaV1.5. Lidocaine had no effect on V½ activation of either variant but caused a significantly greater depression of maximum current mediated by nNaV1.5 compared to aNaV1.5. Similar statistically significant shifts in the V½ inactivation (approximately -10 mV) occurred for both LAs and NaV1.5 variants. Levobupivacaine (1 µM) caused a significantly greater slowing of recovery from inactivation of both variants than did lidocaine (10 µM). Both LAs caused approximately 50% tonic inhibition of aNaV1.5 or nNaV1.5 when holding at -80 mV. Neither LA caused tonic block at a holding potential of either -90 or -120 mV, voltages at which there was little steady-state inactivation. Higher concentrations of either lidocaine (300 µM) or levobupivacaine (100 µM) caused significantly more tonic block at -120 mV. CONCLUSIONS: These data demonstrate that low concentrations of the LAs exhibit inactivation-dependent block of NaV1.5, which may provide a rationale for their use to safely inhibit migration and invasion by metastatic cancer cells without cardiotoxicity.


Assuntos
Anestésicos Locais/farmacologia , Levobupivacaína/farmacologia , Lidocaína/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Adulto , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Recém-Nascido , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia
4.
Handb Exp Pharmacol ; 246: 161-184, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29032483

RESUMO

In the heart, voltage-gated sodium (Nav) channel (Nav1.5) is defined by its pore-forming α-subunit and its auxiliary ß-subunits, both of which are important for its critical contribution to the initiation and maintenance of the cardiac action potential (AP) that underlie normal heart rhythm. The physiological relevance of Nav1.5 is further marked by the fact that inherited or congenital mutations in Nav1.5 channel gene SCN5A lead to altered functional expression (including expression, trafficking, and current density), and are generally manifested in the form of distinct cardiac arrhythmic events, epilepsy, neuropathic pain, migraine, and neuromuscular disorders. However, despite significant advances in defining the pathophysiology of Nav1.5, the molecular mechanisms that underlie its regulation and contribution to cardiac disorders are poorly understood. It is rapidly becoming evident that the functional expression (localization, trafficking and gating) of Nav1.5 may be under modulation by post-translational modifications that are associated with phosphorylation. We review here the molecular basis of cardiac Na channel regulation by kinases (PKA and PKC) and the resulting functional consequences. Specifically, we discuss: (1) recent literature on the structural, molecular, and functional properties of cardiac Nav1.5 channels; (2) how these properties may be altered by phosphorylation in disease states underlain by congenital mutations in Nav1.5 channel and/or subunits such as long QT and Brugada syndromes. Our expectation is that understanding the roles of these distinct and complex phosphorylation processes on the functional expression of Nav1.5 is likely to provide crucial mechanistic insights into Na channel associated arrhythmogenic events and will facilitate the development of novel therapeutic strategies.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Proteína Quinase C/fisiologia , Animais , Humanos , Canal de Sódio Disparado por Voltagem NAV1.5/química , Fosforilação
5.
Handb Exp Pharmacol ; 246: 331-354, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28965168

RESUMO

The voltage-gated cardiac sodium channel (Nav1.5) is a mega-complex comprised of a pore-forming α subunit and 4 ancillary ß-subunits together with numerous protein partners. Genetic defects in the form of rare variants in one or more sodium channel-related genes can cause a loss- or gain-of-function of sodium channel current (INa) leading to the manifestation of various disease phenotypes, including Brugada syndrome, long QT syndrome, progressive cardiac conduction disease, sick sinus syndrome, multifocal ectopic Purkinje-related premature contractions, and atrial fibrillation. Some sodium channelopathies have also been shown to be responsible for sudden infant death syndrome (SIDS). Although these genetic defects often present as pure electrical diseases, recent studies point to a contribution of structural abnormalities to the electrocardiographic and arrhythmic manifestation in some cases, such as dilated cardiomyopathy. The same rare variants in SCN5A or related genes may present with different clinical phenotypes in different individuals and sometimes in members of the same family. Genetic background and epigenetic and environmental factors contribute to the expression of these overlap syndromes. Our goal in this chapter is to review and discuss what is known about the clinical phenotype and genotype of each cardiac sodium channelopathy, and to briefly discuss the underlying mechanisms.


Assuntos
Arritmias Cardíacas/etiologia , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Animais , Síndrome de Brugada/etiologia , Humanos , Síndrome do QT Longo/etiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia
6.
Handb Exp Pharmacol ; 246: 73-99, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28965170

RESUMO

Activation of the electrical signal and its transmission as a depolarizing wave in the whole heart requires highly organized myocyte architecture and cell-cell contacts. In addition, complex trafficking and anchoring intracellular machineries regulate the proper surface expression of channels and their targeting to distinct membrane domains. An increasing list of proteins, lipids, and second messengers can contribute to the normal targeting of ion channels in cardiac myocytes. However, their precise roles in the electrophysiology of the heart are far from been extensively understood. Nowadays, much effort in the field focuses on understanding the mechanisms that regulate ion channel targeting to sarcolemma microdomains and their organization into macromolecular complexes. The purpose of the present section is to provide an overview of the characterized partners of the main cardiac sodium channel, NaV1.5, involved in regulating the functional expression of this channel both in terms of trafficking and targeting into microdomains.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Conexina 43/fisiologia , Proteína 1 Homóloga a Discs-Large , Guanilato Quinases/fisiologia , Humanos , Proteínas de Membrana/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/química , Placofilinas/fisiologia
7.
Invest New Drugs ; 35(3): 277-289, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28164251

RESUMO

Background and Purpose IC261 (3-[(2,4,6-trimethoxyphenyl)methylidenyl]-indolin-2-one) has previously been introduced as an isoform specific inhibitor of casein kinase 1 (CK1) causing cell cycle arrest or cell death of established tumor cell lines. However, it is reasonable to assume that not all antitumor activities of IC261 are mediated by the inhibition of CK1. Meanwhile there is growing evidence that functional voltage-gated sodium channels are also implicated in the progression of tumors as their blockage suppresses tumor migration and invasion of different tumor cell lines. Thus, we asked whether IC261 functionally inhibits voltage-gated sodium channels. Experimental Approach Electrophysiological experiments were performed using the patch-clamp technique at human heart muscle sodium channels heterologously expressed in human TsA cells. Key Results IC261 inhibits sodium channels in a state-dependent manner. IC261 does not interact with the open channel and has only a low affinity for the resting state of the hNav1.5 (human voltage-gated sodium channel; Kr: 120 µM). The efficacy of IC261 strongly increases with membrane depolarisation, indicating that the inactivated state is an important target. The results of different experimental approaches finally revealed an affinity of IC261 to the inactivated state between 1 and 2 µM. Conclusion and Implications IC261 inhibits sodium channels at a similar concentration necessary to reduce CK1δ/ε activity by 50% (IC50 value 1 µM). Thus, inhibition of sodium channels might contribute to the antitumor activity of IC261.


Assuntos
Caseína Quinase I/antagonistas & inibidores , Indóis/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Floroglucinol/análogos & derivados , Bloqueadores dos Canais de Sódio/farmacologia , Linhagem Celular , Humanos , Floroglucinol/farmacologia
8.
Circ Res ; 116(12): 1971-88, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26044251

RESUMO

The movement of ions across specific channels embedded on the membrane of individual cardiomyocytes is crucial for the generation and propagation of the cardiac electric impulse. Emerging evidence over the past 20 years strongly suggests that the normal electric function of the heart is the result of dynamic interactions of membrane ion channels working in an orchestrated fashion as part of complex molecular networks. Such networks work together with exquisite temporal precision to generate each action potential and contraction. Macromolecular complexes play crucial roles in transcription, translation, oligomerization, trafficking, membrane retention, glycosylation, post-translational modification, turnover, function, and degradation of all cardiac ion channels known to date. In addition, the accurate timing of each cardiac beat and contraction demands, a comparable precision on the assembly and organizations of sodium, calcium, and potassium channel complexes within specific subcellular microdomains, where physical proximity allows for prompt and efficient interaction. This review article, part of the Compendium on Sudden Cardiac Death, discusses the major issues related to the role of ion channel macromolecular assemblies in normal cardiac electric function and the mechanisms of arrhythmias leading to sudden cardiac death. It provides an idea of how these issues are being addressed in the laboratory and in the clinic, which important questions remain unanswered, and what future research will be needed to improve knowledge and advance therapy.


Assuntos
Canalopatias/complicações , Morte Súbita Cardíaca/etiologia , Canais Iônicos/fisiologia , Substâncias Macromoleculares/química , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/fisiologia , Compartimento Celular , Canalopatias/genética , Canalopatias/fisiopatologia , Modelos Animais de Doenças , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/química , Canais Iônicos/efeitos dos fármacos , Canais KATP/química , Canais KATP/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Mapeamento de Interação de Proteínas , Subunidades Proteicas
9.
Circulation ; 132(7): 567-77, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26187182

RESUMO

BACKGROUND: Voltage-gated Na(+) channels (Nav) are essential for myocyte membrane excitability and cardiac function. Nav current (INa) is a large-amplitude, short-duration spike generated by rapid channel activation followed immediately by inactivation. However, even under normal conditions, a small late component of INa (INa,L) persists because of incomplete/failed inactivation of a subpopulation of channels. Notably, INa,L is directly linked with both congenital and acquired disease states. The multifunctional Ca(2+)/calmodulin-dependent kinase II (CaMKII) has been identified as an important activator of INa,L in disease. Several potential CaMKII phosphorylation sites have been discovered, including Ser571 in the Nav1.5 DI-DII linker, but the molecular mechanism underlying CaMKII-dependent regulation of INa,L in vivo remains unknown. METHODS AND RESULTS: To determine the in vivo role of Ser571, 2 Scn5a knock-in mouse models were generated expressing either: (1) Nav1.5 with a phosphomimetic mutation at Ser571 (S571E), or (2) Nav1.5 with the phosphorylation site ablated (S571A). Electrophysiology studies revealed that Ser571 regulates INa,L but not other channel properties previously linked to CaMKII. Ser571-mediated increases in INa,L promote abnormal repolarization and intracellular Ca(2+) handling and increase susceptibility to arrhythmia at the cellular and animal level. Importantly, Ser571 is required for maladaptive remodeling and arrhythmias in response to pressure overload. CONCLUSIONS: Our data provide the first in vivo evidence for the molecular mechanism underlying CaMKII activation of the pathogenic INa,L. Relevant for improved rational design of potential therapies, our findings demonstrate that Ser571-dependent regulation of Nav1.5 specifically tunes INa,L without altering critical physiological components of the current.


Assuntos
Arritmias Cardíacas/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Fosfosserina/metabolismo , Remodelação Ventricular/fisiologia , Acetanilidas/farmacologia , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cardiomegalia/fisiopatologia , Constrição , Técnicas de Introdução de Genes , Ativação do Canal Iônico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.5/química , Fosforilação , Piperazinas/farmacologia , Processamento de Proteína Pós-Traducional , Ranolazina , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia
10.
Nano Lett ; 15(3): 1743-50, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25639960

RESUMO

From its invention in the 1970s, the patch clamp technique is the gold standard in electrophysiology research and drug screening because it is the only tool enabling accurate investigation of voltage-gated ion channels, which are responsible for action potentials. Because of its key role in drug screening, innovation efforts are being made to reduce its complexity toward more automated systems. While some of these new approaches are being adopted in pharmaceutical companies, conventional patch-clamp remains unmatched in fundamental research due to its versatility. Here, we merged the patch clamp and atomic force microscope (AFM) techniques, thus equipping the patch-clamp with the sensitive AFM force control. This was possible using the FluidFM, a force-controlled nanopipette based on microchanneled AFM cantilevers. First, the compatibility of the system with patch-clamp electronics and its ability to record the activity of voltage-gated ion channels in whole-cell configuration was demonstrated with sodium (NaV1.5) channels. Second, we showed the feasibility of simultaneous recording of membrane current and force development during contraction of isolated cardiomyocytes. Force feedback allowed for a gentle and stable contact between AFM tip and cell membrane enabling serial patch clamping and injection without apparent cell damage.


Assuntos
Potenciais de Ação/fisiologia , Potenciais da Membrana/fisiologia , Sistemas Microeletromecânicos/instrumentação , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Técnicas de Patch-Clamp/instrumentação , Desenho de Equipamento , Análise de Falha de Equipamento , Retroalimentação , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Micromanipulação/instrumentação , Microscopia de Força Atômica/instrumentação , Contração Miocárdica/fisiologia , Estresse Mecânico
11.
Circulation ; 130(2): 147-60, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24895455

RESUMO

BACKGROUND: Sodium channel NaV1.5 underlies cardiac excitability and conduction. The last 3 residues of NaV1.5 (Ser-Ile-Val) constitute a PDZ domain-binding motif that interacts with PDZ proteins such as syntrophins and SAP97 at different locations within the cardiomyocyte, thus defining distinct pools of NaV1.5 multiprotein complexes. Here, we explored the in vivo and clinical impact of this motif through characterization of mutant mice and genetic screening of patients. METHODS AND RESULTS: To investigate in vivo the regulatory role of this motif, we generated knock-in mice lacking the SIV domain (ΔSIV). ΔSIV mice displayed reduced NaV1.5 expression and sodium current (INa), specifically at the lateral myocyte membrane, whereas NaV1.5 expression and INa at the intercalated disks were unaffected. Optical mapping of ΔSIV hearts revealed that ventricular conduction velocity was preferentially decreased in the transversal direction to myocardial fiber orientation, leading to increased anisotropy of ventricular conduction. Internalization of wild-type and ΔSIV channels was unchanged in HEK293 cells. However, the proteasome inhibitor MG132 rescued ΔSIV INa, suggesting that the SIV motif is important for regulation of NaV1.5 degradation. A missense mutation within the SIV motif (p.V2016M) was identified in a patient with Brugada syndrome. The mutation decreased NaV1.5 cell surface expression and INa when expressed in HEK293 cells. CONCLUSIONS: Our results demonstrate the in vivo significance of the PDZ domain-binding motif in the correct expression of NaV1.5 at the lateral cardiomyocyte membrane and underline the functional role of lateral NaV1.5 in ventricular conduction. Furthermore, we reveal a clinical relevance of the SIV motif in cardiac disease.


Assuntos
Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/biossíntese , Domínios PDZ/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Animais , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia
12.
Circulation ; 130(3): 224-34, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24895457

RESUMO

BACKGROUND: New drugs are routinely screened for IKr blocking properties thought to predict QT prolonging and arrhythmogenic liability. However, recent data suggest that chronic (hours) drug exposure to phosphoinositide 3-kinase inhibitors used in cancer can prolong QT by inhibiting potassium currents and increasing late sodium current (INa-L) in cardiomyocytes. We tested the extent to which IKr blockers with known QT liability generate arrhythmias through this pathway. METHODS AND RESULTS: Acute exposure to dofetilide, an IKr blocker without other recognized electropharmacologic actions, produced no change in ion currents or action potentials in adult mouse cardiomyocytes, which lack IKr. By contrast, 2 to 48 hours of exposure to the drug generated arrhythmogenic afterdepolarizations and ≥15-fold increases in INa-L. Including phosphatidylinositol 3,4,5-trisphosphate, a downstream effector for the phosphoinositide 3-kinase pathway, in the pipette inhibited these effects. INa-L was also increased, and inhibitable by phosphatidylinositol 3,4,5-trisphosphate, with hours of dofetilide exposure in human-induced pluripotent stem cell-derived cardiomyocytes and in Chinese hamster ovary cells transfected with SCN5A, encoding sodium current. Cardiomyocytes from dofetilide-treated mice similarly demonstrated increased INa-L and afterdepolarizations. Other agents with variable IKr-blocking potencies and arrhythmia liability produced a range of effects on INa-L, from marked increases (E-4031, d-sotalol, thioridazine, and erythromycin) to little or no effect (haloperidol, moxifloxacin, and verapamil). CONCLUSIONS: Some but not all drugs designated as arrhythmogenic IKr blockers can generate arrhythmias by augmenting INa-L through the phosphoinositide 3-kinase pathway. These data identify a potential mechanism for individual susceptibility to proarrhythmia and highlight the need for a new paradigm to screen drugs for QT prolonging and arrhythmogenic liability.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Torsades de Pointes/epidemiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Risco , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia , Torsades de Pointes/fisiopatologia , Transfecção
13.
Circ Res ; 113(2): 137-41, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23661717

RESUMO

RATIONALE: In patients with Brugada syndrome, arrhythmias typically originate in the right ventricular outflow tract (RVOT). The RVOT develops from the slowly conducting embryonic outflow tract. OBJECTIVE: We hypothesize that this embryonic phenotype is maintained in the fetal and adult RVOT and leads to conduction slowing, especially after sodium current reduction. METHODS AND RESULTS: We determined expression patterns in the embryonic myocardium and performed activation mapping in fetal and adult hearts, including hearts from adult mice heterozygous for a mutation associated with Brugada syndrome (Scn5a1798insD/+). The embryonic RVOT was characterized by expression of Tbx2, a repressor of differentiation, and absence of expression of both Hey2, a ventricular transcription factor, and Gja1, encoding the principal gap-junction subunit for ventricular fast conduction. Also, conduction velocity was lower in the RVOT than in the right ventricular free wall. Later in the development, Gja1 and Scn5a expression remained lower in the subepicardial myocardium of the RVOT than in RV myocardium. Nevertheless, conduction velocity in the adult RVOT was similar to that of the right ventricular free wall. However, in hearts of Scn5a1798insD/+ mice and in normal hearts treated with ajmaline, conduction was slower in the RVOT than in the right ventricular wall. CONCLUSIONS: The slowly conducting embryonic phenotype is maintained in the fetal and adult RVOT and is unmasked when cardiac sodium channel function is reduced.


Assuntos
Regulação para Baixo/fisiologia , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Função Ventricular Direita/fisiologia , Fatores Etários , Animais , Síndrome de Brugada/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Técnicas de Cultura de Órgãos
14.
J Neurophysiol ; 112(5): 1091-104, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24872539

RESUMO

Olfactory sensory neurons (OSNs) fire spontaneously as well as in response to odor; both forms of firing are physiologically important. We studied voltage-gated Na(+) channels in OSNs to assess their role in spontaneous activity. Whole cell patch-clamp recordings from OSNs demonstrated both tetrodotoxin-sensitive and tetrodotoxin-resistant components of Na(+) current. RT-PCR showed mRNAs for five of the nine different Na(+) channel α-subunits in olfactory tissue; only one was tetrodotoxin resistant, the so-called cardiac subtype NaV1.5. Immunohistochemical analysis indicated that NaV1.5 is present in the apical knob of OSN dendrites but not in the axon. The NaV1.5 channels in OSNs exhibited two important features: 1) a half-inactivation potential near -100 mV, well below the resting potential, and 2) a window current centered near the resting potential. The negative half-inactivation potential renders most NaV1.5 channels in OSNs inactivated at the resting potential, while the window current indicates that the minor fraction of noninactivated NaV1.5 channels have a small probability of opening spontaneously at the resting potential. When the tetrodotoxin-sensitive Na(+) channels were blocked by nanomolar tetrodotoxin at the resting potential, spontaneous firing was suppressed as expected. Furthermore, selectively blocking NaV1.5 channels with Zn(2+) in the absence of tetrodotoxin also suppressed spontaneous firing, indicating that NaV1.5 channels are required for spontaneous activity despite resting inactivation. We propose that window currents produced by noninactivated NaV1.5 channels are one source of the generator potentials that trigger spontaneous firing, while the upstroke and propagation of action potentials in OSNs are borne by the tetrodotoxin-sensitive Na(+) channel subtypes.


Assuntos
Potenciais de Ação/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Mucosa Olfatória/inervação , Células Receptoras Sensoriais/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Mucosa Olfatória/citologia , Mucosa Olfatória/metabolismo
15.
Br J Anaesth ; 113 Suppl 1: i39-i48, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24852501

RESUMO

BACKGROUND: Metastatic breast and colon cancer cells express neonatal and adult splice variants of NaV1.5 voltage-activated Na(+) channels (VASCs). Block of VASCs inhibits cell invasion. Local anaesthetics used during surgical tumour excision inhibit VASC activity on nociceptive neurones providing regional anaesthesia. Inhibition of VASCs on circulating metastatic cancer cells may also be beneficial during the perioperative period. However, ropivacaine, frequently used to provide analgesia during tumour resection, has not been tested on colon cancer cell VASC function or invasion. METHODS: We used reverse transcription-polymerase chain reaction and sequencing to identify NaV1.5 variants in the SW620 metastatic colon cancer cell line. Recombinant adult and neonatal NaV1.5 variants were expressed in human embryonic kidney cells. Voltage-clamp recordings and invasion assays were used to examine the effects of ropivacaine on recombinant NaV1.5 channels and the metastatic potential of SW620 cells, respectively. RESULTS: SW620 cells expressed adult and neonatal NaV1.5 variants, which had similar steady-state inactivation profiles, but distinctive activation curves with the neonatal variant having a V1/2 of activation 7.8 mV more depolarized than the adult variant. Ropivacaine caused a concentration-dependent block of both NaV1.5 variants, with IC50 values of 2.5 and 3.9 µM, respectively. However, the reduction in available steady-state current was selective for neonatal NaV1.5 channels. Ropivacaine inhibited SW620 invasion, with a potency similar to that of inhibition of NaV1.5 channels (3.8 µM). CONCLUSIONS: Ropivacaine is a potent inhibitor of both NaV1.5 channel activity and metastatic colon cancer cell invasion, which may be beneficial during surgical colon cancer excision.


Assuntos
Amidas/farmacologia , Anestésicos Locais/farmacologia , Neoplasias do Colo/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Adulto , Fatores Etários , Amidas/administração & dosagem , Anestésicos Locais/administração & dosagem , Movimento Celular/efeitos dos fármacos , Colágeno , Neoplasias do Colo/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Humanos , Recém-Nascido , Laminina , Lidocaína/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Técnicas de Patch-Clamp , Proteoglicanas , Ropivacaina
16.
Pacing Clin Electrophysiol ; 37(10): 1349-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24861429

RESUMO

BACKGROUND: Activation of the renin-angiotensin system plays an important role in atrial electrical remodeling; angiotensin-(1-7) (Ang-(1-7)) counterbalances the actions of angiotensin II. The aim of this study was to determine the effects of Ang-(1-7) on cardiac sodium current (INa ) in a canine model of atrial tachycardia. METHODS: Eighteen dogs were randomly assigned to sham, pacing, or pacing + Ang-(1-7) groups (n = 6 in each group). Rapid atrial pacing (500 beats/min) was maintained for 2 weeks, while the dogs in the sham group were not paced. Ang-(1-7) (6 µg/kg/h) was administered intravenously during pacing. Whole-cell patch clamp techniques were utilized to record INa from canine atrial myocytes. Reverse transcription-polymerase chain reaction was used to assess possible underlying changes in cardiac Na(+) channels (Nav1.5). RESULTS: Our results showed that INa density and expression of the Nav1.5 mRNA significantly decreased following pacing (P < 0.05 vs sham); however, the half-activation voltage (V1/2act ) and half-inactivation voltage (V1/2inact ) of INa were not significantly altered (P > 0.05 vs sham). Ang-(1-7) treatment significantly increased INa densities and hyperpolarized V1/2act without concomitant changes in V1/2inact but have no effect on the expression of the Nav1.5 gene. CONCLUSIONS: Ang-(1-7) significantly increased INa densities, which contributed to improving intraatrial conduction and decreasing the likelihood of atrial fibrillation maintenance.


Assuntos
Angiotensina I/uso terapêutico , Remodelamento Atrial/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Fragmentos de Peptídeos/uso terapêutico , Taquicardia Supraventricular/fisiopatologia , Animais , Cães , Feminino , Expressão Gênica , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Taquicardia Supraventricular/genética
17.
Chin J Physiol ; 57(4): 188-97, 2014 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-25246060

RESUMO

Sodium channels play an important role in action potentials. Moreover, some evidences recently suggested that sodium channels were responsible for murine sinoatrial node pacemaking. The aim of this study was to investigate the role of sodium channels in pacemaking in embryonic cardiomyocytes in early development stage (EDS). Whole-cell patch-clamp technique was employed to record sodium current of murine early embryonic cardiomyocytes. Current clamp technique was used to record the effect of 0.1, 1 and 10 µM tetrodotoxin (TTX) on embryonic cardiomyocytes pacemaking. Electro- physiology properties of sodium channels in embryonic cardiomyocytes corresponded to Nav1.5, and the IC50 of TTX was 5.24 µM. TTX at 0.1 µM concentration had no effects on the pacemaking. TTX at 1 µM concentration, however, dramatically slowed the spontaneous beating rate from 73.975 ± 10.478 to 50.268 ± 10.476 cycle/min (P < 0.05), and the maximum upstroke velocity (dV/dtmax) of phase 4 from 0.074 ± 0.006 to 0.046 ± 0.007 V/s (P < 0.01). Furthermore, 1 µM TTX reduced the dV/dtmax of phase 0 from 16.405 ± 0.056 to 12.801 ± 0.084 V/s (P < 0.01), and increased the period of phase 4 from 710.342 ± 110.983 to 1320.618 ± 250.483 ms (P < 0.05). TTX at 1 µM also had some effects on the peak of phase 0 decreasing it from 40.621 ± 3.012 to 37.407 ± 2.749 mV (P < 0.05). But TTX at 1 µM had no effects on the period of phase 0. In some cells (9/13), TTX at 10 µM caused complete cessation of spontaneous action potentials. Our results suggested that the main expression subtype of sodium channels was Nav1.5 of early embryonic cardiomyocytes. And TTX-resistant sodium channels contributed to the initiation of action potentials of early embryonic cardiomyocytes, while TTX-sensitive sodium channels were not involved in initiation of action potentials.


Assuntos
Potenciais de Ação/fisiologia , Coração/embriologia , Coração/fisiologia , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Feminino , Camundongos Endogâmicos , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Gravidez , Tetrodotoxina/farmacologia
18.
Circulation ; 126(18): 2208-19, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23019294

RESUMO

BACKGROUND: Heart failure is a growing cause of morbidity and mortality. Cardiac phosphatidylinositol 3-kinase signaling promotes cardiomyocyte survival and function, but it is paradoxically activated in heart failure, suggesting that chronic activation of this pathway may become maladaptive. Here, we investigated the downstream phosphatidylinositol 3-kinase effector, serum- and glucocorticoid-regulated kinase-1 (SGK1), in heart failure and its complications. METHODS AND RESULTS: We found that cardiac SGK1 is activated in human and murine heart failure. We investigated the role of SGK1 in the heart by using cardiac-specific expression of constitutively active or dominant-negative SGK1. Cardiac-specific activation of SGK1 in mice increased mortality, cardiac dysfunction, and ventricular arrhythmias. The proarrhythmic effects of SGK1 were linked to biochemical and functional changes in the cardiac sodium channel and could be reversed by treatment with ranolazine, a blocker of the late sodium current. Conversely, cardiac-specific inhibition of SGK1 protected mice after hemodynamic stress from fibrosis, heart failure, and sodium channel alterations. CONCLUSIONS: SGK1 appears both necessary and sufficient for key features of adverse ventricular remodeling and may provide a novel therapeutic target in cardiac disease.


Assuntos
Cardiomiopatia Dilatada/enzimologia , Insuficiência Cardíaca/enzimologia , Proteínas Imediatamente Precoces/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Remodelação Ventricular/fisiologia , Acetanilidas/uso terapêutico , Animais , Cardiomegalia Induzida por Exercícios , Sequência Consenso , Modelos Animais de Doenças , Eletrocardiografia , Indução Enzimática , Humanos , Hipertensão/complicações , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Piperazinas/uso terapêutico , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ranolazina , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Taquicardia Ventricular/enzimologia , Taquicardia Ventricular/etiologia
19.
Am J Physiol Heart Circ Physiol ; 305(8): H1213-21, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23893162

RESUMO

To determine the roles of the individual S4 segments in domains I and II to activation and inactivation kinetics of sodium current (INa) in NaV1.5, we used a tethered biotin and avidin approach after a site-directed cysteine substitution was made in the second outermost Arg in each S4 (DI-R2C and DII-R2C). We first determined the fraction of gating charge contributed by the individual S4's to maximal gating current (Qmax), and found that the outermost Arg residue in each S4 contributed ∼19% to Qmax with minimal contributions by other arginines. Stabilization of the S4's in DI-R2C and DII-R2C was confirmed by measuring the expected reduction in Qmax. In DI-R2C, stabilization resulted in a decrease in peak INa of ∼45%, while its peak current-voltage (I-V) and voltage-dependent Na channel availability (SSI) curves were nearly unchanged from wild type (WT). In contrast, stabilization of the DII-R2C enhanced activation with a negative shift in the peak I-V relationship by -7 mV and a larger -17 mV shift in the voltage-dependent SSI curve. Furthermore, its INa decay time constants and time-to-peak INa became more rapid than WT. An explanation for these results is that the depolarized conformation of DII-S4, but not DI-S4, affects the receptor for the inactivation particle formed by the interdomain linker between DIII and IV. In addition, the leftward shifts of both activation and inactivation and the decrease in Gmax after stabilization of the DII-S4 support previous studies that showed ß-scorpion toxins trap the voltage sensor of DII in an activated conformation.


Assuntos
Potenciais da Membrana/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Estrutura Terciária de Proteína/fisiologia , Arginina , Humanos , Técnicas de Patch-Clamp
20.
Zhongguo Dang Dai Er Ke Za Zhi ; 15(3): 223-6, 2013 Mar.
Artigo em Zh | MEDLINE | ID: mdl-23498767

RESUMO

OBJECTIVE: To construct the sodium channel gene SCN5A-delQKP1507-1509 mutation associated with congenital long QT syndrome, and its eukaryotic expression vector, and to examine the expression of mutation protein in human embryonic kidney (HEK) 293 cells. METHODS: Eukaryotic expression vector PEGFP-delQKP-hH1 for SCN5A-delQKP1507-1509 mutation was constructed by rapid site-directed mutagenesis. HEK293 cells were transfected with the wild or mutant vector using lipofectamine, and then subjected to confocal microscopy. The transfected cells were immunostained to visualize intracellular expression of the mutant molecules. RESULTS: Direct sequence and electrophoresis analysis revealed 9 basic group absences at position 1507-1509. The delQKP1507-1509 mutation eukaryotic expression vector was expressed in HEK293 cells. Immunostaining of transfected cells showed the expression of both wild type and mutant molecules on the plasma membrane and there was no difference in the amount of protein, which suggested that the mutant delQKP1507-1509 did not impair normal protein expression in HEK293 cells. CONCLUSIONS: Successful construction of mutant SCN5AdelQKP1507-1509 eukaryotic expression vector and expression of SCN5A protein in HEK293 cells provides a basis for further study on the functional effects of congenital long QT syndrome as a cause of SCN5A mutation.


Assuntos
Síndrome do QT Longo/genética , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Western Blotting , Células HEK293 , Humanos , Síndrome do QT Longo/congênito , Canal de Sódio Disparado por Voltagem NAV1.5/análise , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA