Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 17(12): 1245-1253, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33169015

RESUMO

Impaired protein stability or trafficking underlies diverse ion channelopathies and represents an unexploited unifying principle for developing common treatments for otherwise dissimilar diseases. Ubiquitination limits ion channel surface density, but targeting this pathway for the purposes of basic study or therapy is challenging because of its prevalent role in proteostasis. We developed engineered deubiquitinases (enDUBs) that enable selective ubiquitin chain removal from target proteins to rescue the functional expression of disparate mutant ion channels that underlie long QT syndrome (LQT) and cystic fibrosis (CF). In an LQT type 1 (LQT1) cardiomyocyte model, enDUB treatment restored delayed rectifier potassium currents and normalized action potential duration. CF-targeted enDUBs synergistically rescued common (ΔF508) and pharmacotherapy-resistant (N1303K) CF mutations when combined with the US Food and Drug Administation (FDA)-approved drugs Orkambi (lumacaftor/ivacaftor) and Trikafta (elexacaftor/tezacaftor/ivacaftor and ivacaftor). Altogether, targeted deubiquitination via enDUBs provides a powerful protein stabilization method that not only corrects diverse diseases caused by impaired ion channel trafficking, but also introduces a new tool for deconstructing the ubiquitin code in situ.


Assuntos
Canalopatias/patologia , Fibrose Cística/patologia , Enzimas Desubiquitinantes/metabolismo , Transporte de Íons/fisiologia , Síndrome do QT Longo/patologia , Canais de Potássio/fisiologia , Aminofenóis/farmacologia , Aminopiridinas/farmacologia , Benzodioxóis/farmacologia , Canalopatias/genética , Fibrose Cística/genética , Enzimas Desubiquitinantes/genética , Combinação de Medicamentos , Humanos , Indóis/farmacologia , Transporte de Íons/genética , Síndrome do QT Longo/genética , Miócitos Cardíacos/fisiologia , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Canais de Potássio/genética , Pirazóis/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Quinolonas/farmacologia
2.
Nature ; 545(7652): 54-59, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28445465

RESUMO

The development of the nervous system involves a coordinated succession of events including the migration of GABAergic (γ-aminobutyric-acid-releasing) neurons from ventral to dorsal forebrain and their integration into cortical circuits. However, these interregional interactions have not yet been modelled with human cells. Here we generate three-dimensional spheroids from human pluripotent stem cells that resemble either the dorsal or ventral forebrain and contain cortical glutamatergic or GABAergic neurons. These subdomain-specific forebrain spheroids can be assembled in vitro to recapitulate the saltatory migration of interneurons observed in the fetal forebrain. Using this system, we find that in Timothy syndrome-a neurodevelopmental disorder that is caused by mutations in the CaV1.2 calcium channel-interneurons display abnormal migratory saltations. We also show that after migration, interneurons functionally integrate with glutamatergic neurons to form a microphysiological system. We anticipate that this approach will be useful for studying neural development and disease, and for deriving spheroids that resemble other brain regions to assemble circuits in vitro.


Assuntos
Neurônios/citologia , Prosencéfalo/citologia , Prosencéfalo/crescimento & desenvolvimento , Esferoides Celulares/citologia , Transtorno Autístico/genética , Transtorno Autístico/patologia , Linhagem Celular , Movimento Celular , Células Cultivadas , Feminino , Neurônios GABAérgicos/citologia , Ácido Glutâmico/metabolismo , Humanos , Interneurônios/citologia , Interneurônios/patologia , Síndrome do QT Longo/genética , Síndrome do QT Longo/patologia , Masculino , Modelos Biológicos , Neurogênese , Neurônios/patologia , Células-Tronco Pluripotentes/citologia , Prosencéfalo/anatomia & histologia , Sinapses/fisiologia , Sindactilia/genética , Sindactilia/patologia
3.
J Cell Mol Med ; 25(11): 4938-4949, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33939251

RESUMO

Drug-mediated or medical condition-mediated disruption of hERG function accounts for the main cause of acquired long-QT syndrome (acLQTs), which predisposes affected individuals to ventricular arrhythmias (VA) and sudden death. Many Chinese herbal medicines, especially alkaloids, have risks of arrhythmia in clinical application. The characterized mechanisms behind this adverse effect are frequently associated with inhibition of cardiac hERG channels. The present study aimed to assess the potent effect of Rutaecarpine (Rut) on hERG channels. hERG-HEK293 cell was applied for evaluating the effect of Rut on hERG channels and the underlying mechanism. hERG current (IhERG ) was measured by patch-clamp technique. Protein levels were analysed by Western blot, and the phosphorylation of Sp1 was determined by immunoprecipitation. Optical mapping and programmed electrical stimulation were used to evaluate cardiac electrophysiological activities, such as APD, QT/QTc, occurrence of arrhythmia, phase singularities (PSs), and dominant frequency (DF). Our results demonstrated that Rut reduced the IhERG by binding to F656 and Y652 amino acid residues of hERG channel instantaneously, subsequently accelerating the channel inactivation, and being trapped in the channel. The level of hERG channels was reduced by incubating with Rut for 24 hours, and Sp1 in nucleus was inhibited simultaneously. Mechanismly, Rut reduced threonine (Thr)/ tyrosine (Tyr) phosphorylation of Sp1 through PI3K/Akt pathway to regulate hERG channels expression. Cell-based model unables to fully reveal the pathological process of arrhythmia. In vivo study, we found that Rut prolonged QT/QTc intervals and increased induction rate of ventricular fibrillation (VF) in guinea pig heart after being dosed Rut for 2 weeks. The critical reasons led to increased incidence of arrhythmias eventually were prolonged APD90 and APD50 and the increase of DF, numbers of PSs, incidence of early after-depolarizations (EADs). Collectively, the results of this study suggest that Rut could reduce the IhERG by binding to hERG channels through F656 and Y652 instantaneously. While, the PI3K/Akt/Sp1 axis may play an essential role in the regulation of hERG channels, from the perspective of the long-term effects of Rut (incubating for 24 hours). Importantly, the changes of electrophysiological properties by Rut were the main cause of VA.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/patologia , Canal de Potássio ERG1/antagonistas & inibidores , Alcaloides Indólicos/efeitos adversos , Síndrome do QT Longo/patologia , Quinazolinas/efeitos adversos , Vasodilatadores/efeitos adversos , Disfunção Ventricular/patologia , Animais , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/metabolismo , Células Cultivadas , Fenômenos Eletrofisiológicos , Cobaias , Células HEK293 , Humanos , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/metabolismo , Masculino , Disfunção Ventricular/induzido quimicamente , Disfunção Ventricular/metabolismo
4.
Bioorg Med Chem Lett ; 46: 128162, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34062251

RESUMO

In the United States, approximately one million individuals are hospitalized every year for arrhythmias, making arrhythmias one of the top causes of healthcare expenditures. Mexiletine is currently used as an antiarrhythmic drug but has limitations. The purpose of this work was to use normal and Long QT syndrome Type 3 (LQTS3) patient-derived human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to identify an analog of mexiletine with superior drug-like properties. Compared to racemic mexiletine, medicinal chemistry optimization of substituted racemic pyridyl phenyl mexiletine analogs resulted in a more potent sodium channel inhibitor with greater selectivity for the sodium over the potassium channel and for late over peak sodium current.


Assuntos
Doença do Sistema de Condução Cardíaco/patologia , Células-Tronco Pluripotentes Induzidas/química , Síndrome do QT Longo/patologia , Mexiletina/farmacologia , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Piridinas/farmacologia , Relação Dose-Resposta a Droga , Humanos , Mexiletina/química , Estrutura Molecular , Piridinas/química , Relação Estrutura-Atividade
5.
Int J Mol Sci ; 22(20)2021 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-34681909

RESUMO

In the heart, the delayed rectifier K current, IK, composed of the rapid (IKr) and slow (IKs) components contributes prominently to normal cardiac repolarization. In lipotoxicity, chronic elevation of pro-inflammatory cytokines may remodel IK, elevating the risk for ventricular arrythmias and sudden cardiac death. We investigated whether and how the pro-inflammatory interleukin-6 altered IK in the heart, using electrophysiology to evaluate changes in IK in adult guinea pig ventricular myocytes. We found that palmitic acid (a potent inducer of lipotoxicity), induced a rapid (~24 h) and significant increase in IL-6 in RAW264.7 cells. PA-diet fed guinea pigs displayed a severely prolonged QT interval when compared to low-fat diet fed controls. Exposure to isoproterenol induced torsade de pointes, and ventricular fibrillation in lipotoxic guinea pigs. Pre-exposure to IL-6 with the soluble IL-6 receptor produced a profound depression of IKr and IKs densities, prolonged action potential duration, and impaired mitochondrial ATP production. Only with the inhibition of IKr did a proarrhythmic phenotype of IKs depression emerge, manifested as a further prolongation of action potential duration and QT interval. Our data offer unique mechanistic insights with implications for pathological QT interval in patients and vulnerability to fatal arrhythmias.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/patologia , Interleucina-6/metabolismo , Síndrome do QT Longo/patologia , Macrófagos/metabolismo , Miócitos Cardíacos/patologia , Canais de Potássio/química , Animais , Arritmias Cardíacas/metabolismo , Cardiotoxicidade/fisiopatologia , Feminino , Cobaias , Ativação do Canal Iônico , Metabolismo dos Lipídeos , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo
6.
J Hum Genet ; 65(12): 1083-1091, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32681117

RESUMO

Mutations in KCNQ1, KCNH2, and SCN5A are the major cause of long QT syndrome (LQTS). More than 90% of the genotyped patients have been reported to carry mutations in any of these three genes. Thanks to increasing popularity of next generation sequencer (NGS), novel CACNA1C mutations have been identified among LQTS patients without extra-cardiac phenotypes. We aimed to clarify the frequency of genotypes in LQTS patients in the era of NGS. The study comprised 160 congenital LQTS patients (71 males) registered from November 2015 to September 2018. Inclusion criteria was QTc > 460 ms and Schwartz score ≥ 3. We performed genetic analysis using target gene method by NGS and confirmed the mutations by Sanger method. The median age for genetic screening was 13 (0-68) years. Sixteen patients suffered cardiac arrest, 47 syncope, and 97 were asymptomatic. We identified genetic mutations in 111 (69.4%) patients including 6 CACNA1C (5.4% of the genotyped patients) with 4 asymptomatic patients. Five (3.1%) patients carried double mutations; three out of them with RYR2 and KCNQ1 or KCNH2. In conclusion, CACNA1C screening would be recommended even if the patient is asymptomatic to elucidate the genetic background of the LQTS patients.


Assuntos
Canais de Cálcio Tipo L/genética , Predisposição Genética para Doença , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Canal de Potássio ERG1/genética , Feminino , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Recém-Nascido , Síndrome do QT Longo/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Linhagem , Adulto Jovem
7.
Circ Res ; 122(3): e5-e16, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29282212

RESUMO

RATIONALE: There are several methods to measure cardiomyocyte and muscle contraction, but these require customized hardware, expensive apparatus, and advanced informatics or can only be used in single experimental models. Consequently, data and techniques have been difficult to reproduce across models and laboratories, analysis is time consuming, and only specialist researchers can quantify data. OBJECTIVE: Here, we describe and validate an automated, open-source software tool (MUSCLEMOTION) adaptable for use with standard laboratory and clinical imaging equipment that enables quantitative analysis of normal cardiac contraction, disease phenotypes, and pharmacological responses. METHODS AND RESULTS: MUSCLEMOTION allowed rapid and easy measurement of movement from high-speed movies in (1) 1-dimensional in vitro models, such as isolated adult and human pluripotent stem cell-derived cardiomyocytes; (2) 2-dimensional in vitro models, such as beating cardiomyocyte monolayers or small clusters of human pluripotent stem cell-derived cardiomyocytes; (3) 3-dimensional multicellular in vitro or in vivo contractile tissues, such as cardiac "organoids," engineered heart tissues, and zebrafish and human hearts. MUSCLEMOTION was effective under different recording conditions (bright-field microscopy with simultaneous patch-clamp recording, phase contrast microscopy, and traction force microscopy). Outcomes were virtually identical to the current gold standards for contraction measurement, such as optical flow, post deflection, edge-detection systems, or manual analyses. Finally, we used the algorithm to quantify contraction in in vitro and in vivo arrhythmia models and to measure pharmacological responses. CONCLUSIONS: Using a single open-source method for processing video recordings, we obtained reliable pharmacological data and measures of cardiac disease phenotype in experimental cell, animal, and human models.


Assuntos
Contração Miocárdica , Miócitos Cardíacos/fisiologia , Software , Algoritmos , Animais , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Fármacos Cardiovasculares/farmacologia , Diferenciação Celular , Células Cultivadas , Subunidades beta da Proteína de Ligação ao GTP/deficiência , Subunidades beta da Proteína de Ligação ao GTP/genética , Humanos , Síndrome do QT Longo/patologia , Síndrome do QT Longo/fisiopatologia , Masculino , Microscopia/métodos , Modelos Cardiovasculares , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Fenótipo , Células-Tronco Pluripotentes/citologia , Coelhos , Gravação em Vídeo , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
8.
J Pediatr Hematol Oncol ; 42(2): e121-e124, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30830033

RESUMO

Studies have been conducted on adults prescribed with methadone to determine the necessary frequency of QTc monitoring but no consensus has been reached and no similar research has been conducted in the pediatric population. The objective of this retrospective study was to determine the occurrence rate of QTc interval prolongation associated with methadone use in a pediatric oncologic population. In total, 18% of patients developed QTc interval prolongation. These patients had longer baseline QTc intervals and were on more QTc interval-prolonging medications. Our data suggest that these variables may be able to risk stratify patients who require more frequent monitoring.


Assuntos
Analgésicos Opioides/efeitos adversos , Dor do Câncer/tratamento farmacológico , Síndrome do QT Longo/epidemiologia , Metadona/efeitos adversos , Neoplasias/complicações , Adolescente , Adulto , Dor do Câncer/etiologia , Dor do Câncer/patologia , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Incidência , Lactente , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/patologia , Masculino , New York/epidemiologia , Prognóstico , Estudos Retrospectivos , Adulto Jovem
9.
Proc Natl Acad Sci U S A ; 114(21): E4288-E4295, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28490495

RESUMO

Alternative splicing changes the CaV1.2 calcium channel electrophysiological property, but the in vivo significance of such altered channel function is lacking. Structure-function studies of heterologously expressed CaV1.2 channels could not recapitulate channel function in the native milieu of the cardiomyocyte. To address this gap in knowledge, we investigated the role of alternative exon 33 of the CaV1.2 calcium channel in heart function. Exclusion of exon 33 in CaV1.2 channels has been reported to shift the activation potential -10.4 mV to the hyperpolarized direction, and increased expression of CaV1.2Δ33 channels was observed in rat myocardial infarcted hearts. However, how a change in CaV1.2 channel electrophysiological property, due to alternative splicing, might affect cardiac function in vivo is unknown. To address these questions, we generated mCacna1c exon 33-/--null mice. These mice contained CaV1.2Δ33 channels with a gain-of-function that included conduction of larger currents that reflects a shift in voltage dependence and a modest increase in single-channel open probability. This altered channel property underscored the development of ventricular arrhythmia, which is reflected in significantly more deaths of exon 33-/- mice from ß-adrenergic stimulation. In vivo telemetric recordings also confirmed increased frequencies in premature ventricular contractions, tachycardia, and lengthened QT interval. Taken together, the significant decrease or absence of exon 33-containing CaV1.2 channels is potentially proarrhythmic in the heart. Of clinical relevance, human ischemic and dilated cardiomyopathy hearts showed increased inclusion of exon 33. However, the possible role that inclusion of exon 33 in CaV1.2 channels may play in the pathogenesis of human heart failure remains unclear.


Assuntos
Potenciais de Ação/genética , Canais de Cálcio Tipo L/genética , Síndrome do QT Longo/genética , Taquicardia/genética , Complexos Ventriculares Prematuros/genética , Potenciais de Ação/fisiologia , Processamento Alternativo/genética , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Colforsina/farmacologia , Fenômenos Eletrofisiológicos/genética , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Isoproterenol/farmacologia , Síndrome do QT Longo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Nifedipino/farmacologia , Ratos , Deleção de Sequência/genética , Taquicardia/patologia , Complexos Ventriculares Prematuros/patologia
10.
Heart Lung Circ ; 29(4): 529-537, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31959550

RESUMO

Sympathetically triggered inherited arrhythmia syndromes, including the long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT), can cause sudden cardiac death in young individuals with structurally normal hearts. With cardiac events typically triggered by physical or emotional stress, not surprisingly, two of the most common treatments are neuromodulators, including mainstay beta blocker pharmacotherapy, and surgical sympathetic cardiac denervation. This review updates the clinician on the relevant anatomy and physiology of the cardiac autonomic nervous system, outlines neurocardiac arrhythmia mechanisms, and discusses the latest rationale for a neurocardiac therapeutic approach to manage sympathetic-induced arrhythmia in patients with inherited cardiac disease.


Assuntos
Encéfalo , Coração/fisiopatologia , Síndrome do QT Longo , Angústia Psicológica , Sistema Nervoso Simpático , Taquicardia Ventricular , Antagonistas Adrenérgicos beta/uso terapêutico , Encéfalo/patologia , Encéfalo/fisiopatologia , Humanos , Síndrome do QT Longo/patologia , Síndrome do QT Longo/fisiopatologia , Síndrome do QT Longo/terapia , Sistema Nervoso Simpático/patologia , Sistema Nervoso Simpático/fisiopatologia , Taquicardia Ventricular/patologia , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/terapia
11.
J Cell Mol Med ; 23(9): 6331-6342, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31361068

RESUMO

The K+ voltage-gated channel subfamily H member 2 (KCNH2) transports the rapid component of the cardiac delayed rectifying K+ current. The aim of this study was to characterize the biophysical properties of a C-terminus-truncated KCNH2 channel, G1006fs/49 causing long QT syndrome type II in heterozygous members of an Italian family. Mutant carriers underwent clinical workup, including 12-lead electrocardiogram, transthoracic echocardiography and 24-hour ECG recording. Electrophysiological experiments compared the biophysical properties of G1006fs/49 with those of KCNH2 both expressed either as homotetramers or as heterotetramers in HEK293 cells. Major findings of this work are as follows: (a) G1006fs/49 is functional at the plasma membrane even when co-expressed with KCNH2, (b) G1006fs/49 exerts a dominant-negative effect on KCNH2 conferring specific biophysical properties to the heterotetrameric channel such as a significant delay in the voltage-sensitive transition to the open state, faster kinetics of both inactivation and recovery from the inactivation and (c) the activation kinetics of the G1006fs/49 heterotetrameric channels is partially restored by a specific KCNH2 activator. The functional characterization of G1006fs/49 homo/heterotetramers provided crucial findings about the pathogenesis of LQTS type II in the mutant carriers, thus providing a new and potential pharmacological strategy.


Assuntos
Canal de Potássio ERG1/genética , Síndrome do QT Longo/genética , Síndrome do QT Longo/patologia , Mutação/genética , Adolescente , Adulto , Linhagem Celular , Membrana Celular/genética , Criança , Eletrocardiografia/métodos , Feminino , Células HEK293 , Humanos , Masculino , Fenótipo , Transporte Proteico/genética , Adulto Jovem
12.
Circulation ; 138(21): 2345-2358, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30571576

RESUMO

BACKGROUND: Long QT syndrome (LQTS) is associated with potentially fatal arrhythmias. Treatment is very effective, but its diagnosis may be challenging. Importantly, different methods are used to assess the QT interval, which makes its recognition difficult. QT experts advocate manual measurements with the tangent or threshold method. However, differences between these methods and their performance in LQTS diagnosis have not been established. We aimed to assess similarities and differences between these 2 methods for QT interval analysis to aid in accurate QT assessment for LQTS. METHODS: Patients with a confirmed pathogenic variant in KCNQ1(LQT1), KCNH2(LQT2), or SCN5A(LQT3) genes and their family members were included. Genotype-positive patients were identified as LQTS cases and genotype-negative family members as controls. ECGs were analyzed with both methods, providing inter- and intrareader validity and diagnostic accuracy. Cutoff values based on control population's 95th and 99th percentiles, and LQTS-patients' 1st and 5th percentiles were established based on the method to correct for heart rate, age, and sex. RESULTS: We included 1484 individuals from 265 families, aged 33±21 years and 55% females. In the total cohort, QTTangent was 10.4 ms shorter compared with QTThreshold (95% limits of agreement±20.5 ms, P<0.0001). For all genotypes, QTTangent was shorter than QTThreshold ( P<0.0001), but this was less pronounced in LQT2. Both methods yielded a high inter- and intrareader validity (intraclass correlation coefficient >0.96), and a high diagnostic accuracy (area under the curve >0.84). Using the current guideline cutoff (QTc interval 480 ms), both methods had similar specificity but yielded a different sensitivity. QTc interval cutoff values of QTTangent were lower compared with QTThreshold and different depending on the correction for heart rate, age, and sex. CONCLUSION: The QT interval varies depending on the method used for its assessment, yet both methods have a high validity and can both be used in diagnosing LQTS. However, for diagnostic purposes current guideline cutoff values yield different results for these 2 methods and could result in inappropriate reassurance or treatment. Adjusted cutoff values are therefore specified for method, correction formula, age, and sex. In addition, a freely accessible online probability calculator for LQTS ( www.QTcalculator.org ) has been made available as an aid in the interpretation of the QT interval.


Assuntos
Eletrocardiografia , Síndrome do QT Longo/diagnóstico , Adolescente , Adulto , Fatores Etários , Estudos de Casos e Controles , Criança , Bases de Dados Factuais , Canal de Potássio ERG1/genética , Feminino , Genótipo , Humanos , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/patologia , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Estudos Retrospectivos , Fatores Sexuais , Adulto Jovem
13.
Clin Exp Allergy ; 49(12): 1615-1623, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31519068

RESUMO

The symptoms of chronic urticaria, be it chronic spontaneous urticaria (CSU) or chronic inducible urticaria (CindU), are mediated primarily by the actions of histamine on H1 receptors located on endothelial cells (the weal) and on sensory nerves (neurogenic flare and pruritus). Thus, second-generation H1 antihistamines (sgAHs) are the primary treatment of these conditions. However, many patients are poorly responsive to licensed doses of antihistamines. In these patients, the current EAACI/GA2 LEN/EDF/WAO guideline for urticaria suggests updosing of sgAHs up to fourfold. However, such updosing is off-label and the responsibility resides with the prescribing physician. Therefore, the safety of the drug when used above its licensed dose is of paramount importance. An important aspect of safety is potential cardiotoxicity. This problem was initially identified some 20 years ago with cardiotoxic deaths occurring with astemizole and terfenadine, two early sgAHs. In this review, we discuss the mechanisms and assessments of potential cardiotoxicity of H1 antihistamines when updosed to four times their licensed dose. In particular, we have focused on the potential of H1 antihistamines to block hERG (human Ether-a-go-go-Related Gene) voltage-gated K+ channels, also known as Kv11.1 channels according to the IUPHAR classification. Blockade of these channels causes QT prolongation leading to torsade de pointes that may possibly degenerate into ventricular fibrillation and sudden death. We considered in detail bilastine, cetirizine, levocetirizine, ebastine, fexofenadine, loratadine, desloratadine, mizolastine and rupatadine and concluded that all these drugs have an excellent safety profile with no evidence of cardiotoxicity even when updosed up to four times their standard licensed dose, provided that the prescribers carefully consider and rule out potential risk factors for cardiotoxicity, such as the presence of inherited long QT syndrome, older age, cardiovascular disorders, hypokalemia and hypomagnesemia, or the use of drugs that either have direct QT prolonging effects or inhibit sgAH metabolism.


Assuntos
Cardiotoxicidade , Urticária Crônica , Canal de Potássio ERG1 , Antagonistas dos Receptores Histamínicos H1 , Torsades de Pointes , Fatores Etários , Cardiotoxicidade/imunologia , Cardiotoxicidade/patologia , Cardiotoxicidade/prevenção & controle , Urticária Crônica/tratamento farmacológico , Urticária Crônica/imunologia , Urticária Crônica/patologia , Canal de Potássio ERG1/imunologia , Feminino , Antagonistas dos Receptores Histamínicos H1/efeitos adversos , Antagonistas dos Receptores Histamínicos H1/uso terapêutico , Humanos , Síndrome do QT Longo/imunologia , Síndrome do QT Longo/patologia , Masculino , Fatores de Risco , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/imunologia , Torsades de Pointes/patologia
14.
J Hum Genet ; 64(3): 233-238, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30542207

RESUMO

Long QT syndrome (LQTS) is a rare inherited arrhythmia disease characterized by a prolonged QT interval on 12-lead electrocardiograms. It is the crucial factor to induce syncope, ventricular fibrillation, and even sudden cardiac death. Previous studies have proved that mutations of ion channels-related genes play an important role in LQTS patients. In this study, we enrolled a Chinese family with LQTS and syncope. With the help of whole-exome sequencing, we identified a novel nonsense mutation (c.439C>T/p.Q147X) of Ring Finger Protein 207 (RNF207) in this family. The novel mutation, resulting in a premature stop codon in exon 4 of the RNF207 gene, co-segregated with the affected individuals. Bioinformatics analysis and real-time PCR further proved that the newly identified mutation might induce nonsense-mediated mRNA decay. In mutation carriers, the level of RNF207 mRNA expression was much lower than controls, which may affect potassium channel KCNH2 and lead to LQTS and syncope. In this research, we reported a rare novel mutation of RNF207 in LQTS and syncope patients which further supports the significant role of RNF207 in potassium channel activation and expanded the spectrum of RNF207 mutations. These data may contribute to the genetic diagnosis and counseling of families with LQTS and syncope.


Assuntos
Povo Asiático/genética , Códon sem Sentido , Sequenciamento do Exoma/métodos , Exoma , Síndrome do QT Longo/genética , Ubiquitina-Proteína Ligases/genética , Adolescente , Adulto , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Síndrome do QT Longo/patologia , Masculino , Pessoa de Meia-Idade , Linhagem
15.
FASEB J ; 32(3): 1440-1451, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29133340

RESUMO

Cardiac arrhythmias are major life-threatening conditions. The landmark discovery of induced pluripotent stem cells has provided a promising in vitro system for modeling hereditary cardiac arrhythmias as well as drug development and toxicity testing. Nowadays, nutraceuticals are frequently used as supplements for cardiovascular therapy. Here we studied the cardiac effects of hawthorn ( Crataegus pentagyna) leaf extract using cardiomyocytes (CMs) differentiated from healthy human embryonic stem cells, long QT syndrome type 2 (LQTS2), and catecholaminergic polymorphic ventricular tachycardia type 1 (CPVT1) patient-specific induced pluripotent stem cells. The hydroalcoholic extract resulted in a dose-dependent negative chronotropic effect in all CM preparations leading to a significant reduction at 1000 µg/ml. This was accompanied by prolongation of field potential durations, although with different magnitudes in CMs from different human embryonic stem cell and iPSC lines. Hawthorn further prolonged field potential durations in LQTS2 CMs but reduced the beating frequencies and occurrence of immature field potentials triggered by ß1-adrenergic stimulation in CPVT1 CMs at 300 and 1000 µg/ml. Furthermore, isoquercetin and vitexin flavonoids significantly slowed down isoproterenol (5 µM)-induced beating frequencies at 3 and 10 µg/ml. Therefore, C. pentagyna leaf extract and its isoquercetin and vitexin flavonoids may be introduced as a novel nutraceutical with antiarrhythmic potential for CPVT1 patients.-Pahlavan, S., Tousi, M. S., Ayyari, M., Alirezalu, A., Ansari, H., Saric, T., Baharvand, H. Effects of hawthorn ( Crataegus pentagyna) leaf extract on electrophysiologic properties of cardiomyocytes derived from human cardiac arrhythmia-specific induced pluripotent stem cells.


Assuntos
Crataegus/química , Células-Tronco Pluripotentes Induzidas/metabolismo , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo , Extratos Vegetais/farmacologia , Taquicardia Ventricular/metabolismo , Técnicas Eletrofisiológicas Cardíacas , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome do QT Longo/patologia , Miócitos Cardíacos/patologia , Extratos Vegetais/química , Taquicardia Ventricular/patologia
16.
Curr Treat Options Oncol ; 20(7): 55, 2019 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-31129799

RESUMO

OPINION STATEMENT: Cardiovascular toxicities are potentially serious treatment limiting complications of many different cancer therapeutics including traditional cytotoxic chemotherapies as well as targeted- and immunotherapies. As a result, there is increased monitoring for cancer treatment-related cardiotoxicities, ranging from heart failure to arrhythmias. Many anticancer treatments are known to prolong the QT interval through a variety of mechanisms including direct effects on ion channels and indirectly via intracellular signaling pathways. While QT prolongation increases the risk for the potentially life-threatening ventricular arrhythmia torsades de pointes, the incidence of this arrhythmia in the setting of most cancer treatments is quite rare, and the majority of patients can continue safely receiving these medications despite their QT prolonging potential. A multidisciplinary approach to the cardiovascular care of the cancer patient is essential to mitigate risk of cardiotoxicity while minimizing unnecessary treatment disruption of potentially life-saving cancer treatments.


Assuntos
Antineoplásicos/efeitos adversos , Cardiotoxicidade/patologia , Síndrome do QT Longo/induzido quimicamente , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Cardiotoxicidade/etiologia , Humanos , Síndrome do QT Longo/patologia , Terapia de Alvo Molecular/efeitos adversos , Terapia de Alvo Molecular/métodos , Neoplasias/patologia , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto
17.
Biophys J ; 115(6): 1019-1032, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30173888

RESUMO

Long QT syndrome type 2 (LQT2) is a congenital disease characterized by loss of function mutations in hERG potassium channels (IKr). LQT2 is associated with fatal ventricular arrhythmias promoted by triggered activity in the form of early afterdepolarizations (EADs). We previously demonstrated that intracellular Ca2+ handling is remodeled in LQT2 myocytes. Remodeling leads to aberrant late RyR-mediated Ca2+ releases that drive forward-mode Na+-Ca2+ exchanger (NCX) current and slow repolarization to promote reopening of L-type calcium channels and EADs. Forward-mode NCX was found to be enhanced despite the fact that these late releases do not significantly alter the whole-cell cytosolic calcium concentration during a vulnerable period of phase 2 of the action potential corresponding to the onset of EADs. Here, we use a multiscale ventricular myocyte model to explain this finding. We show that because the local NCX current is a saturating nonlinear function of the local submembrane calcium concentration, a larger number of smaller-amplitude discrete Ca2+ release events can produce a large increase in whole-cell forward-mode NCX current without increasing significantly the whole-cell cytosolic calcium concentration. Furthermore, we develop novel insights, to our knowledge, into how alterations of stochastic RyR activity at the single-channel level cause late aberrant Ca2+ release events. Experimental measurements in transgenic LTQ2 rabbits confirm the critical arrhythmogenic role of NCX and identify this current as a potential target for antiarrhythmic therapies in LQT2.


Assuntos
Cálcio/metabolismo , Espaço Intracelular/metabolismo , Síndrome do QT Longo/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Trocador de Sódio e Cálcio/metabolismo , Animais , Citosol/metabolismo , Ventrículos do Coração/patologia , Ativação do Canal Iônico , Modelos Biológicos , Fenótipo , Probabilidade , Coelhos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
18.
Glia ; 66(11): 2324-2339, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30151840

RESUMO

To study the role of L-type voltage-gated Ca++ channels in oligodendrocyte development, we used a mouse model of Timothy syndrome (TS) in which a gain-of-function mutation in the α1 subunit of the L-type Ca++ channel Cav1.2 gives rise to an autism spectrum disorder (ASD). Oligodendrocyte progenitor cells (OPCs) isolated from the cortex of TS mice showed greater L-type Ca++ influx and displayed characteristics suggestive of advanced maturation compared to control OPCs, including a more complex morphology and higher levels of myelin protein expression. Consistent with this, expression of Cav1.2 channels bearing the TS mutation in wild-type OPCs triggered process formation and promoted oligodendrocyte-neuron interaction via the activation of Ca++ /calmodulin-dependent protein kinase II. To ascertain whether accelerated OPC maturation correlated with functional enhancements, we examined myelination in the TS brain at different postnatal time points. The expression of myelin proteins was significantly higher in the corpus callosum, cortex and striatum of TS animals, and immunohistochemical analysis for oligodendrocyte stage-specific markers revealed an increase in the density of myelinating oligodendrocytes in several areas of the TS brain. Along the same line, electron microscopy studies in the corpus callosum of TS animals showed significant increases both in the percentage of myelinated axons and in the thickness of myelin sheaths. In summary, these data indicate that OPC development and oligodendrocyte myelination is enhanced in the brain of TS mice, and suggest that this mouse model of a syndromic ASD is a useful tool to explore the role of L-type Ca++ channels in myelination.


Assuntos
Transtorno Autístico/complicações , Transtorno Autístico/patologia , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Síndrome do QT Longo/complicações , Síndrome do QT Longo/patologia , Proteínas da Mielina/metabolismo , Oligodendroglia/fisiologia , Sindactilia/complicações , Sindactilia/patologia , Animais , Animais Recém-Nascidos , Transtorno Autístico/genética , Proteínas Relacionadas à Autofagia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Técnicas de Cocultura , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Síndrome do QT Longo/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Oligodendroglia/patologia , Oligodendroglia/ultraestrutura , Potássio/farmacologia , Sindactilia/genética
19.
Cell Physiol Biochem ; 45(1): 192-202, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29339628

RESUMO

BACKGROUND/AIMS: Arsenic trioxide (ATO) is a known anti-acute promyelocytic leukemia (APL) reagent, whose clinical applications are limited by its serious cardiac toxicity and fatal adverse effects, such as sudden cardiac death resulting from long QT syndrome (LQTS). The mechanisms of cardiac arrhythmia due to ATO exposure still need to be elucidated. Long non-coding RNAs (lncRNAs) are emerging as major regulators of various pathophysiological processes. This study aimed to explore the involvement of lncRNAs in ATO-induced LQTS in vivo and in vitro. METHODS: For in vivo experiments, mice were administered ATO through the tail vein. For in vitro experiments, ATO was added to the culture medium of primary cultured neonatal mouse cardiomyocytes. To evaluate the effect of lncRNA Kcnq1ot1, siRNA and lentivirus-shRNA were synthesized to knockdown lncRNA Kcnq1ot1. RESULTS: After ATO treatment, the Kcnq1ot1 and Kcnq1 expression levels were down regulated. lncRNA Kcnq1ot1 knockdown prolonged the action potential duration (APD) in vitro and exerted LQTS in vivo. Correspondingly, Kcnq1 expression was decreased after silencing lncRNA Kcnq1ot1. However, the knockdown of Kcnq1 exerted no effect on lncRNA Kcnq1ot1 expression. CONCLUSIONS: To our knowledge, this report is the first to demonstrate that lncRNA Kcnq1ot1 downregulation is responsible for QT interval prolongation induced by ATO at least partially by repressing Kcnq1 expression. lncRNA Kcnq1ot1 has important pathophysiological functions in the heart and could become a novel antiarrhythmic target.


Assuntos
Arsenicais , Síndrome do QT Longo/induzido quimicamente , Óxidos , RNA Longo não Codificante/metabolismo , Potenciais de Ação , Animais , Trióxido de Arsênio , Células Cultivadas , Regulação para Baixo , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Lentivirus/genética , Síndrome do QT Longo/patologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Óxidos/toxicidade , Técnicas de Patch-Clamp , Fenótipo , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo
20.
Am J Med Genet A ; 176(7): 1657-1661, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29736926

RESUMO

Timothy syndrome is characterized by a unique combination of a prolongation of the corrected QT interval of the electrocardiogram and bilateral cutaneous syndactyly of the fingers and the toes and is caused by heterozygous mutations in CACNA1C, a gene encoding a calcium channel. After the discovery of the CACNA1C gene as the causative gene for Timothy syndrome, patients with CACNA1C mutations with QT prolongation but without syndactyly were described. Here, we report a 5-year-old female patient with cutaneous syndactyly, developmental delay, and pulmonary hypertension. Exome analysis showed a previously undescribed de novo heterozygous mutation in the CACNA1C gene, p.Arg1024Gly. To our knowledge, this patient is the first to exhibit syndactyly and to carry a CACNA1C mutation but to not have QT prolongation, which has long been considered an obligatory feature of Timothy syndrome.


Assuntos
Transtorno Autístico/patologia , Canais de Cálcio Tipo L/genética , Deficiências do Desenvolvimento/patologia , Hipertensão Pulmonar/patologia , Síndrome do QT Longo/patologia , Mutação , Sindactilia/patologia , Transtorno Autístico/genética , Deficiências do Desenvolvimento/genética , Feminino , Humanos , Hipertensão Pulmonar/genética , Recém-Nascido , Síndrome do QT Longo/genética , Fenótipo , Sindactilia/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA