Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Gen Physiol ; 156(5)2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38557788

RESUMO

DSC1, a Drosophila channel with sequence similarity to the voltage-gated sodium channel (NaV), was identified over 20 years ago. This channel was suspected to function as a non-specific cation channel with the ability to facilitate the permeation of calcium ions (Ca2+). A honeybee channel homologous to DSC1 was recently cloned and shown to exhibit strict selectivity for Ca2+, while excluding sodium ions (Na+), thus defining a new family of Ca2+ channels, known as CaV4. In this study, we characterize CaV4, showing that it exhibits an unprecedented type of inactivation, which depends on both an IFM motif and on the permeating divalent cation, like NaV and CaV1 channels, respectively. CaV4 displays a specific pharmacology with an unusual response to the alkaloid veratrine. It also possesses an inactivation mechanism that uses the same structural domains as NaV but permeates Ca2+ ions instead. This distinctive feature may provide valuable insights into how voltage- and calcium-dependent modulation of voltage-gated Ca2+ and Na+ channels occur under conditions involving local changes in intracellular calcium concentrations. Our study underscores the unique profile of CaV4 and defines this channel as a novel class of voltage-gated Ca2+ channels.


Assuntos
Cálcio , Canais de Sódio Disparados por Voltagem , Abelhas , Animais , Canais de Sódio Disparados por Voltagem/química , Íons
2.
Sci Rep ; 8(1): 13804, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30218094

RESUMO

Dilated cardiomyopathy (DCM) is a structural heart disease that causes dilatation of cardiac chambers and impairs cardiac contractility. The SCN5A gene encodes Nav1.5, the predominant cardiac sodium channel alpha subunit. SCN5A mutations have been identified in patients with arrhythmic disorders associated with DCM. The characterization of Nav1.5 mutations located in the voltage sensor domain (VSD) and associated with DCM revealed divergent biophysical defects that do not fully explain the pathologies observed in these patients. The purpose of this study was to characterize the pathological consequences of a gating pore in the heart arising from the Nav1.5/R219H mutation in a patient with complex cardiac arrhythmias and DCM. We report its properties using cardiomyocytes derived from patient-specific human induced pluripotent stem cells. We showed that this mutation generates a proton leak (called gating pore current). We also described disrupted ionic homeostasis, altered cellular morphology, electrical properties, and contractile function, most probably linked to the proton leak. We thus propose a novel link between SCN5A mutation and the complex pathogenesis of cardiac arrhythmias and DCM. Furthermore, we suggest that leaky channels would constitute a common pathological mechanism underlying several neuronal, neuromuscular, and cardiac pathologies.


Assuntos
Arritmias Cardíacas/etiologia , Cardiomiopatia Dilatada/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Arritmias Cardíacas/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Mutação , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Linhagem , Canais de Sódio Disparados por Voltagem/genética , Canais de Sódio Disparados por Voltagem/fisiologia
3.
Sci Rep ; 8(1): 2041, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391559

RESUMO

Mutations in NaV1.4, the skeletal muscle voltage-gated Na+ channel, underlie several skeletal muscle channelopathies. We report here the functional characterization of two substitutions targeting the R1451 residue and resulting in 3 distinct clinical phenotypes. The R1451L is a novel pathogenic substitution found in two unrelated individuals. The first individual was diagnosed with non-dystrophic myotonia, whereas the second suffered from an unusual phenotype combining hyperkalemic and hypokalemic episodes of periodic paralysis (PP). The R1451C substitution was found in one individual with a single attack of hypoPP induced by glucocorticoids. To elucidate the biophysical mechanism underlying the phenotypes, we used the patch-clamp technique to study tsA201 cells expressing WT or R1451C/L channels. Our results showed that both substitutions shifted the inactivation to hyperpolarized potentials, slowed the kinetics of inactivation, slowed the recovery from slow inactivation and reduced the current density. Cooling further enhanced these abnormalities. Homology modeling revealed a disruption of hydrogen bonds in the voltage sensor domain caused by R1451C/L. We concluded that the altered biophysical properties of R1451C/L well account for the PMC-hyperPP cluster and that additional factors likely play a critical role in the inter-individual differences of clinical expression resulting from R1451C/L.


Assuntos
Mutação de Sentido Incorreto , Transtornos Miotônicos/genética , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.4/química , Canal de Sódio Disparado por Voltagem NAV1.4/genética
4.
FEBS Lett ; 591(23): 3842-3849, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29105054

RESUMO

Metaflumizone is the latest addition to the armamentarium of the Na+ channel inhibitor insecticide family. We used the Xenopus oocyte expression system and a Markovian model to assess the effect of metaflumizone on Apis mellifera Na+ channels (AmNaV 1). Our results reveal that metaflumizone inhibits AmNaV 1 channels by targeting the kinetics of recovery from slow inactivation. Multistate modeling of fast and slow inactivation of the AmNaV 1 channel made it possible to study the effects of metaflumizone on a set of rate constants underlying the transition between the open and inactivated conformations and provided insights into their specificity. We conclude that the methods we used could be extended to assessing the toxicity of other Na+ channel inhibitor insecticides.


Assuntos
Abelhas/metabolismo , Proteínas de Insetos/antagonistas & inibidores , Inseticidas/farmacologia , Semicarbazonas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Abelhas/genética , Feminino , Técnicas In Vitro , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Resistência a Inseticidas , Cinética , Modelos Biológicos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Sódio Disparados por Voltagem/genética , Xenopus
5.
FASEB J ; 31(7): 3066-3071, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28356346

RESUMO

The decline of the western honeybee (Apis mellifera) has been reported to be due to parasitism by Varroa destructor mites and to colony collapse disorder in which these mites may be involved. In-hive chemicals such as τ-fluvalinate are being used to control Vdestructor populations. This approach may lead to the chronic exposure of bees to this liposoluble chemical, which tends to accumulate in hives. We cloned a variant of the V. destructor voltage-dependent sodium (VdNaV1) channel and studied its biophysical characteristics and sensitivity to τ-fluvalinate using the Xenopus oocyte expression system and the 2-microelectrode voltage-clamp technique. We compared the affinity of VdNaV1 for τ-fluvalinate with the honeybee voltage-dependent sodium ortholog. Our results showed that the honeybee sodium channel is more sensitive to τ-fluvalinate than the V. destructor channel, suggesting that care must be taken when treating hives with this chemical.-Gosselin-Badaroudine, P., Chahine, M. Biophysical characterization of the Varroa destructor NaV1 sodium channel and its affinity for τ-fluvalinate insecticide.


Assuntos
Proteínas de Artrópodes/metabolismo , Inseticidas/farmacologia , Nitrilas/farmacologia , Piretrinas/farmacologia , Canais de Sódio/metabolismo , Varroidae/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/genética , Abelhas/parasitologia , Clonagem Molecular , Regulação da Expressão Gênica/fisiologia , Inseticidas/química , Nitrilas/química , Oócitos , Variantes Farmacogenômicos , Piretrinas/química , Xenopus
6.
J Gen Physiol ; 148(2): 133-45, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27432995

RESUMO

Bilaterian voltage-gated Na(+) channels (NaV) evolved from voltage-gated Ca(2+) channels (CaV). The Drosophila melanogaster Na(+) channel 1 (DSC1), which features a D-E-E-A selectivity filter sequence that is intermediate between CaV and NaV channels, is evidence of this evolution. Phylogenetic analysis has classified DSC1 as a Ca(2+)-permeable Na(+) channel belonging to the NaV2 family because of its sequence similarity with NaV channels. This is despite insect NaV2 channels (DSC1 and its orthologue in Blatella germanica, BSC1) being more permeable to Ca(2+) than Na(+) In this study, we report the cloning and molecular characterization of the honeybee (Apis mellifera) DSC1 orthologue. We reveal several sequence variations caused by alternative splicing, RNA editing, and genomic variations. Using the Xenopus oocyte heterologous expression system and the two-microelectrode voltage-clamp technique, we find that the channel exhibits slow activation and inactivation kinetics, insensitivity to tetrodotoxin, and block by Cd(2+) and Zn(2+) These characteristics are reminiscent of CaV channels. We also show a strong selectivity for Ca(2+) and Ba(2+) ions, marginal permeability to Li(+), and impermeability to Mg(2+) and Na(+) ions. Based on current ion channel nomenclature, the D-E-E-A selectivity filter, and the properties we have uncovered, we propose that DSC1 homologues should be classified as CaV4 rather than NaV2. Indeed, channels that contain the D-E-E-A selectivity sequence are likely to feature the same properties as the honeybee's channel, namely slow activation and inactivation kinetics and strong selectivity for Ca(2+) ions.


Assuntos
Abelhas/metabolismo , Canais de Cálcio/metabolismo , Processamento Alternativo , Animais , Canais de Cálcio/genética , Clonagem Molecular , Técnicas de Patch-Clamp , Filogenia
7.
Sci Rep ; 5: 12475, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26202396

RESUMO

Pollination is important for both agriculture and biodiversity. For a significant number of plants, this process is highly, and sometimes exclusively, dependent on the pollination activity of honeybees. The large numbers of honeybee colony losses reported in recent years have been attributed to colony collapse disorder. Various hypotheses, including pesticide overuse, have been suggested to explain the disorder. Using the Xenopus oocytes expression system and two microelectrode voltage-clamp, we report the functional expression and the molecular, biophysical, and pharmacological characterization of the western honeybee's sodium channel (Apis Mellifera NaV1). The NaV1 channel is the primary target for pyrethroid insecticides in insect pests. We further report that the honeybee's channel is also sensitive to permethrin and fenvalerate, respectively type I and type II pyrethroid insecticides. Molecular docking of these insecticides revealed a binding site that is similar to sites previously identified in other insects. We describe in vitro and in silico tools that can be used to test chemical compounds. Our findings could be used to assess the risks that current and next generation pesticides pose to honeybee populations.


Assuntos
Abelhas/metabolismo , Inseticidas/química , Inseticidas/toxicidade , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/química , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/toxicidade , Testes de Toxicidade , Canais de Sódio Disparados por Voltagem/ultraestrutura
8.
Channels (Austin) ; 9(3): 139-44, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26046592

RESUMO

Voltage-gated ion channels (VGIC) are transmembrane proteins responsible for the generation of electrical signals in excitable cells. VGIC were first described in 1952 by Hodgkin and Huxley, (1) and have since been associated with various physiological functions such as propagating nerve impulses, locomotion, and cardiac excitability. VGIC include channels specialized in the selective passage of K(+), Ca(2+) Na(+), or H(+). They are composed of 2 main structures: the pore domain (PD) and the voltage sensor domain (VSD). The PD ensures the physiological flow of ions and is typically composed of 8 transmembrane segments (TM). The VSD detects voltage variations and is composed of 4 TM (S1-S4). Given their crucial physiological role, VGIC dysfunctions are associated with diverse pathologies known as ion channelopathies. These dysfunctions usually affect the membrane expression of ion channels or voltage-dependent conformational changes of the pore. However, an increasing number of ion channelopathies, including periodic paralysis, dilated cardiomyopathy (DCM) associated with cardiac arrhythmias, and peripheral nerve hyperexcitability (PNH), have been linked to the appearance of a new pathological mechanism involving the creation of an alternative permeation pathway through the normally non-conductive VSD of VGIC. This permeation pathway is called the gating pore or omega pore.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Dilatada/fisiopatologia , Ativação do Canal Iônico/fisiologia , Humanos , Canais Iônicos/fisiologia , Estrutura Terciária de Proteína
9.
J Gen Physiol ; 145(2): 93-106, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25624448

RESUMO

The gating pore current, also called omega current, consists of a cation leak through the typically nonconductive voltage-sensor domain (VSD) of voltage-gated ion channels. Although the study of gating pore currents has refined our knowledge of the structure and the function of voltage-gated ion channels, their implication in cardiac disorders has not been established. Two Na(v)1.5 mutations (R222Q and R225W) located in the VSD are associated with atypical clinical phenotypes involving complex arrhythmias and dilated cardiomyopathy. Using the patch-clamp technique, in silico mutagenesis, and molecular dynamic simulations, we tested the hypothesis that these two mutations may generate gating pore currents, potentially accounting for their clinical phenotypes. Our findings suggest that the gating pore current generated by the R222Q and R225W mutations could constitute the underlying pathological mechanism that links Na(v)1.5 VSD mutations with human cardiac arrhythmias and dilatation of cardiac chambers.


Assuntos
Arritmias Cardíacas/genética , Cardiomiopatia Dilatada/genética , Ativação do Canal Iônico , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Sequência de Aminoácidos , Arritmias Cardíacas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Células HEK293 , Humanos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/genética
10.
Front Pharmacol ; 6: 301, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26733869

RESUMO

Voltage gated sodium channels (Nav) are transmembrane proteins responsible for action potential initiation. Mutations mainly located in the voltage sensor domain (VSD) of Nav1.5, the cardiac sodium channel, have been associated with the development of arrhythmias combined with dilated cardiomyopathy. Gating pore currents have been observed with three unrelated mutations associated with similar clinical phenotypes. However, gating pores have never been associated with mutations outside the first domain of Nav1.5. The aim of this study was to explore the possibility that gating pore currents might be caused by the Nav1.5 R225P and R814W mutations (R3, S4 in DI and DII, respectively), which are associated with rhythm disturbances and dilated cardiomyopathy. Nav1.5 WT and mutant channels were transiently expressed in tsA201 cells. The biophysical properties of the alpha pore currents and the presence of gating pore currents were investigated using the patch-clamp technique. We confirmed the previously reported gain of function of the alpha pores of the mutant channels, which mainly consisted of increased window currents mostly caused by shifts in the voltage dependence of activation. We also observed gating pore currents associated with the R225P and R814W mutations. This novel permeation pathway was open under depolarized conditions and remained temporarily open at hyperpolarized potentials after depolarization periods. Gating pore currents could represent a molecular basis for the development of uncommon electrical abnormalities and changes in cardiac morphology. We propose that this biophysical defect be routinely evaluated in the case of Nav1.5 mutations on the VSD.

11.
Q Rev Biophys ; 47(4): 364-88, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25382261

RESUMO

The voltage sensitive domain (VSD) is a pivotal structure of voltage-gated ion channels (VGICs) and plays an essential role in the generation of electrochemical signals by neurons, striated muscle cells, and endocrine cells. The VSD is not unique to VGICs. Recent studies have shown that a VSD regulates a phosphatase. Similarly, Hv1, a voltage-sensitive protein that lacks an apparent pore domain, is a self-contained voltage sensor that operates as an H⁺ channel. VSDs are formed by four transmembrane helices (S1-S4). The S4 helix is positively charged due to the presence of arginine and lysine residues. It is surrounded by two water crevices that extend into the membrane from both the extracellular and intracellular milieus. A hydrophobic septum disrupts communication between these water crevices thus preventing the permeation of ions. The septum is maintained by interactions between the charged residues of the S4 segment and the gating charge transfer center. Mutating the charged residue of the S4 segment allows the water crevices to communicate and generate gating pore or omega pore. Gating pore currents have been reported to underlie several neuronal and striated muscle channelopathies. Depending on which charged residue on the S4 segment is mutated, gating pores are permeant either at depolarized or hyperpolarized voltages. Gating pores are cation selective and seem to converge toward Eisenmann's first or second selectivity sequences. Most gating pores are blocked by guanidine derivatives as well as trivalent and quadrivalent cations. Gating pores can be used to study the movement of the voltage sensor and could serve as targets for novel small therapeutic molecules.


Assuntos
Fenômenos Biofísicos , Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/metabolismo , Biologia Molecular/métodos , Sequência de Aminoácidos , Animais , Humanos , Canais Iônicos/antagonistas & inibidores , Dados de Sequência Molecular , Porosidade
12.
Front Pharmacol ; 5: 53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24772081

RESUMO

Voltage sensor domains (VSDs) are a feature of voltage gated ion channels (VGICs) and voltage sensitive proteins. They are composed of four transmembrane (TM) segments (S1-S4). Currents leaking through VSDs are called omega or gating pore currents. Gating pores are caused by mutations of the highly conserved positively charged amino acids in the S4 segment that disrupt interactions between the S4 segment and the gating charge transfer center (GCTC). The GCTC separates the intracellular and extracellular water crevices. The disruption of S4-GCTC interactions allows these crevices to communicate and create a fast activating and non-inactivating alternative cation-selective permeation pathway of low conductance, or a gating pore. Gating pore currents have recently been shown to cause periodic paralysis phenotypes. There is also increasing evidence that gating pores are linked to several other familial diseases. For example, gating pores in Nav1.5 and Kv7.2 channels may underlie mixed arrhythmias associated with dilated cardiomyopathy (DCM) phenotypes and peripheral nerve hyperexcitability (PNH), respectively. There is little evidence for the existence of gating pore blockers. Moreover, it is known that a number of toxins bind to the VSD of a specific domain of Na(+) channels. These toxins may thus modulate gating pore currents. This focus on the VSD motif opens up a new area of research centered on developing molecules to treat a number of cell excitability disorders such as epilepsy, cardiac arrhythmias, and pain. The purpose of the present review is to summarize existing knowledge of the pathophysiology, biophysics, and pharmacology of gating pore currents and to serve as a guide for future studies aimed at improving our understanding of gating pores and their pathophysiological roles.

13.
Channels (Austin) ; 8(1): 90-4, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24300601

RESUMO

Nav 1.5 dysfunctions are commonly linked to rhythms disturbances that include type 3 long QT syndrome (LQT3), Brugada syndrome (BrS), sick sinus syndrome (SSS) and conduction defects. Recently, this channel protein has been also linked to structural heart diseases such as dilated cardiomyopathy (DCM).


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Humanos , Ativação do Canal Iônico , Mutação , Fenótipo
14.
Front Pharmacol ; 4: 126, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098284

RESUMO

Long QT syndrome (LQTS) is a congenital abnormality of cardiac repolarization that manifests as a prolonged QT interval on 12-lead electrocardiograms (ECGs). The syndrome may lead to syncope and sudden death from ventricular tachyarrhythmias known as torsades de pointes. An increased persistent Na(+) current is known to cause a Ca(2+) overload in case of ischemia for example. Such increased Na(+) persistent current is also usually associated to the LQT3 syndrome. The purpose of this study was to investigate the pathological consequences of a novel mutation in a family affected by LQTS. The impact of biophysical defects on cellular homeostasis are also investigated. Genomic DNA was extracted from blood samples, and a combination of PCR and DNA sequencing of several LQTS-linked genes was used to identify mutations. The mutation was reproduced in vitro and was characterized using the patch clamp technique and in silico quantitative analysis. A novel mutation (Q1476R) was identified on the SCN5A gene encoding the cardiac Na(+) channel. Cells expressing the Q1476R mutation exhibited biophysical alterations, including a shift of SS inactivation and a significant increase in the persistent Na(+) current. The in silico analysis confirmed the arrhythmogenic character of the Q1476R mutation. It further revealed that the increase in persistent Na(+) current causes a frequency-dependent Na(+) overload in cardiomyocytes co-expressing WT and mutant Nav1.5 channels that, in turn, exerts a moderating effect on the lengthening of the action potential (AP) duration caused by the mutation. The Q1476R mutation in SCN5A results in a three-fold increase in the window current and a persistent inward Na(+) current. These biophysical defects may expose the carrier of the mutation to arrhythmias that occur preferentially in the patient at rest or during tachycardia. However, the Na(+) overload counterbalances the gain-of-function of the mutation and is beneficial in that it prevents severe arrhythmias at intermediate heart rates.

15.
Proc Natl Acad Sci U S A ; 109(47): 19250-5, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23134726

RESUMO

Mammalian voltage-gated sodium channels are composed of four homologous voltage sensor domains (VSDs; DI, DII, DIII, and DIV) in which their S4 segments contain a variable number of positively charged residues. We used single histidine (H) substitutions of these charged residues in the Na(v)1.4 channel to probe the positions of the S4 segments at hyperpolarized potentials. The substitutions led to the formation of gating pores that were detected as proton leak currents through the VSDs. The leak currents indicated that the mutated residues are accessible from both sides of the membrane. Leak currents of different magnitudes appeared in the DI/R1H, DII/R1H, and DIII/R2H mutants, suggesting that the resting state position of S4 varies depending on the domain. Here, DI/R1H indicates the first arginine R1, in domain DI, has been mutated to histidine. The single R1H, R2H, and R3H mutations in DIV did not produce appreciable proton currents, indicating that the VSDs had different topologies. A structural model of the resting states of the four VSDs of Na(v)1.4 relaxed in their membrane/solution environment using molecular dynamics simulations is proposed based on the recent Na(v)Ab sodium channel X-ray structure. The model shows that the hydrophobic septa that isolate the intracellular and the extracellular media within the DI, DII, and DIII VSDs are ∼2 Šlong, similar to those of K(v) channels. However, the septum of DIV is longer, which prevents water molecules from hydrating the center of the VSD, thus breaking the proton conduction pathway. This structural model rationalizes the activation sequence of the different VSDs of the Na(v)1.4 channel.


Assuntos
Condutividade Elétrica , Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.4/química , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Oócitos , Porosidade , Estrutura Terciária de Proteína , Prótons , Alinhamento de Sequência , Xenopus
16.
PLoS One ; 7(5): e38331, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22675453

RESUMO

Cardiac Na(+) channels encoded by the SCN5A gene are essential for initiating heart beats and maintaining a regular heart rhythm. Mutations in these channels have recently been associated with atrial fibrillation, ventricular arrhythmias, conduction disorders, and dilated cardiomyopathy (DCM).We investigated a young male patient with a mixed phenotype composed of documented conduction disorder, atrial flutter, and ventricular tachycardia associated with DCM. Further family screening revealed DCM in the patient's mother and sister and in three of the mother's sisters. Because of the complex clinical phenotypes, we screened SCN5A and identified a novel mutation, R219H, which is located on a highly conserved region on the fourth helix of the voltage sensor domain of Na(v)1.5. Three family members with DCM carried the R219H mutation.The wild-type (WT) and mutant Na(+) channels were expressed in a heterologous expression system, and intracellular pH (pHi) was measured using a pH-sensitive electrode. The biophysical characterization of the mutant channel revealed an unexpected selective proton leak with no effect on its biophysical properties. The H(+) leak through the mutated Na(v)1.5 channel was not related to the Na(+) permeation pathway but occurred through an alternative pore, most probably a proton wire on the voltage sensor domain.We propose that acidification of cardiac myocytes and/or downstream events may cause the DCM phenotype and other electrical problems in affected family members. The identification of this clinically significant H(+) leak may lead to the development of more targeted treatments.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Dilatada/fisiopatologia , Prótons , Canais de Sódio/metabolismo , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arritmias Cardíacas/genética , Sequência de Bases , Cardiomiopatia Dilatada/genética , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Masculino , Dados de Sequência Molecular , Mutação , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Oócitos/metabolismo , Linhagem , Fenótipo , Canais de Sódio/genética , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...