Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Acta Physiol (Oxf) ; 229(2): e13438, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31900993

RESUMO

AIMS: Pathogenic variants of the SCN5A gene can cause Brugada syndrome (BrS) and long QT syndrome (LQTS), which predispose individuals to potentially fatal ventricular arrhythmias and sudden cardiac death. SCN5A encodes the NaV 1.5 protein, the pore forming α-subunit of the voltage-dependent cardiac Na+ channel. Using a WW domain, the E3 ubiquitin ligase Nedd4-2 binds to the PY-motif ([L/P]PxY) within the C-terminus of NaV 1.5, which results in decreased protein expression and current through NaV 1.5 ubiquitination. Here, we investigate the role of E3 ubiquitin ligase Nedd4-2-mediated NaV 1.5 degradation in the pathological mechanisms of the BrS-associated variant SCN5A-p.L1239P and LQTS-associated variant SCN5A-p.Y1977N. METHODS AND RESULTS: Using a combination of molecular biology, biochemical and electrophysiological approaches, we examined the expression, function and Nedd4-2 interactions of SCN5A-p.L1239P and SCN5A-p.Y1977N. SCN5A-p.L1239P is characterized as a loss-of-function, whereas SCN5A-p.Y1977N is a gain-of-function variant of the NaV 1.5 channel. Sequence alignment shows that BrS-associated SCN5A-p.L1239P has a new Nedd4-2-binding site (from LLxY to LPxY). This new Nedd4-2-binding site increases the interaction between NaV 1.5 and Nedd4-2, enhancing ubiquitination and degradation of the NaV 1.5 channel. Disruption of the new Nedd4-2-binding site of SCN5A-p.L1239P restores NaV 1.5 expression and function. However, the LQTS-associated SCN5A-p.Y1977N disrupts the usual Nedd4-2-binding site (from PPxY to PPxN). This decreases NaV 1.5-Nedd4-2 interaction, preventing ubiquitination and degradation of NaV 1.5 channels. CONCLUSIONS: Our data suggest that the PY-motif plays an essential role in modifying the expression/function of NaV 1.5 channels through Nedd4-2-mediated ubiquitination. Alterations of NaV 1.5-Nedd4-2 interaction represent a novel pathological mechanism for NaV 1.5 channel diseases caused by SCN5A variants.


Assuntos
Síndrome de Brugada/metabolismo , Síndrome do QT Longo/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Motivos de Aminoácidos , Síndrome de Brugada/enzimologia , Células HEK293 , Humanos , Síndrome do QT Longo/enzimologia , Ligação Proteica
2.
Arq. bras. cardiol ; 112(2): 173-178, Feb. 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-983835

RESUMO

Abstract Background: Trimetazidine (TMZ) is an anti-ischemic drug. In spite of its protective effects on cardiovascular system, there is no scientific study on the usefulness of TMZ treatment for prolonged QT interval and cardiac hypertrophy induced by diabetes. Objectives: To evaluate the effects of TMZ on QT interval prolongation and cardiac hypertrophy in the diabetic rats. Methods: Twenty-four male Sprague-Dawley rats (200-250 g) were randomly assigned into three groups (n = 8) by simple random sampling method. Control (C), diabetic (D), and diabetic administrated with TMZ at 10 mg/kg (T10). TMZ was administrated for 8 weeks. The echocardiogram was recorded before isolating the hearts and transfer to a Langendorff apparatus. Hemodynamic parameters, QT and corrected QT interval (QTc) intervals, heart rate and antioxidant enzymes were measured. The hypertrophy index was calculated. The results were evaluated by one-way ANOVA and paired t-test using SPSS (version 16) and p < 0.05 was regarded as significant. Results: The diabetic rats significantly indicated increased hypertrophy, QT and QTc intervals and decreased Left ventricular systolic pressure (LVSP), Left ventricular developed pressure (LVDP), rate pressure product (RPP), Max dp/dt, and min dp/dt (±dp/dt max), heart rate, superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase in the heart. Treatment with TMZ in the diabetic animals was significantly improved these parameters in comparison to the untreated diabetic group. Conclusions: TMZ improves QTc interval prolongation and cardiac hypertrophy in diabetes.


Resumo Fundamento: A trimetazidina (TMZ) é uma droga anti-isquêmica. Apesar de seus efeitos protetores sobre o sistema cardiovascular, não há estudos científicos sobre a utilidade do tratamento com TMZ para o intervalo QT prolongado e a hipertrofia cardíaca induzida pelo diabetes. Objetivo: Avaliar os efeitos da TMZ no prolongamento do intervalo QT e na hipertrofia cardíaca em ratos diabéticos. Métodos: Vinte e quatro ratos machos Sprague-Dawley (200-250 g) foram distribuídos aleatoriamente em três grupos (n = 8) pelo método de amostragem aleatória simples. Controle (C), diabético (D) e diabético administrado com TMZ a 10 mg/kg (T10). A TMZ foi administrada por 8 semanas. O ecocardiograma foi registrado antes de isolar os corações e transferir para um aparelho de Langendorff. Foram medidos os parâmetros hemodinâmicos, intervalo QT e intervalo QT corrigido (QTc), frequência cardíaca e enzimas antioxidantes. O índice de hipertrofia foi calculado. Os resultados foram avaliados pelo one-way ANOVA e pelo teste t pareado pelo SPSS (versão 16) e p < 0,05 foi considerado significativo. Resultados: Os ratos diabéticos indicaram hipertrofia aumentada, intervalos QT e QTc e diminuição da pressão sistólica no ventrículo esquerdo (PSVE), pressão desenvolvida no ventrículo esquerdo (PDVE), duplo produto (DP), Max dp/dt e min dp/dt (± dp/dt max), frequência cardíaca, superóxido dismutase (SOD), glutationa peroxidase (GPx) e catalase no coração. O tratamento com TMZ nos animais diabéticos melhorou significativamente esses parâmetros em comparação com o grupo diabético não tratado. Conclusões: A TMZ melhora o prolongamento do intervalo QTc e a hipertrofia cardíaca no diabetes.


Assuntos
Animais , Masculino , Trimetazidina/farmacologia , Síndrome do QT Longo/tratamento farmacológico , Cardiomegalia/tratamento farmacológico , Substâncias Protetoras/farmacologia , Complicações do Diabetes/tratamento farmacológico , Superóxido Dismutase/análise , Fatores de Tempo , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Ecocardiografia , Catalase/análise , Distribuição Aleatória , Reprodutibilidade dos Testes , Ratos Sprague-Dawley , Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Complicações do Diabetes/enzimologia , Complicações do Diabetes/fisiopatologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/fisiopatologia , Glutationa Peroxidase/análise , Hemodinâmica/efeitos dos fármacos
3.
Heart Rhythm ; 16(7): 1091-1097, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30677491

RESUMO

BACKGROUND: We recently reported a quantitative relationship between the degree of functional perturbation reported in the literature for 356 variants in the cardiac sodium channel gene SCN5A and the penetrance of resulting arrhythmia phenotypes. In the course of that work, we identified multiple SCN5A variants, including R1193Q, that are common in populations but are reported in human embryonic kidney (HEK) cells to generate large late sodium current (INa-L). OBJECTIVE: The purpose of this study was to compare the functional properties of R1193Q with those of the well-studied type 3 long QT syndrome mutation ΔKPQ. METHODS: We compared functional properties of SCN5A R1193Q with those of ΔKPQ in Chinese hamster ovary (CHO) cells at baseline and after exposure to intracellular phosphatidylinositol (3,4,5)-trisphosphate (PIP3), which inhibits INa-L generated by decreased Phosphoinositide 3-kinase (PI3K) activity. We also used CRISPR/Cas9 editing to generate R1193Q in human-induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs). RESULTS: Both R1193Q and ΔKPQ generated robust INa-L in CHO cells. PIP3 abrogated the late current phenotype in R1193Q cells but had no effect on ΔKPQ. Homozygous R1193Q hiPSC-CMs displayed increased INa-L and long action potentials with frequent triggered beats, which were reversed with the addition of PIP3. CONCLUSION: The consistency between the late current produced in HEK cells, CHO cells, and hiPSC-CMs suggests that the late current is a feature of the SCN5A R1193Q variant in human cardiomyocytes but that the mechanism by which the late current is produced is distinct and indirect, as compared with the more highly penetrant ΔKPQ. These data suggest that observing a late current in an in vitro setting does not necessarily translate to highly pathogenic type 3 long QT syndrome phenotype but depends on the underlying mechanism.


Assuntos
Doença do Sistema de Condução Cardíaco/enzimologia , Doença do Sistema de Condução Cardíaco/genética , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Potenciais de Ação/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Rim/citologia , Miócitos Cardíacos/citologia , Fenótipo
4.
Arq Bras Cardiol ; 112(2): 173-178, 2019 02.
Artigo em Inglês, Português | MEDLINE | ID: mdl-30570065

RESUMO

BACKGROUND: Trimetazidine (TMZ) is an anti-ischemic drug. In spite of its protective effects on cardiovascular system, there is no scientific study on the usefulness of TMZ treatment for prolonged QT interval and cardiac hypertrophy induced by diabetes. OBJECTIVES: To evaluate the effects of TMZ on QT interval prolongation and cardiac hypertrophy in the diabetic rats. METHODS: Twenty-four male Sprague-Dawley rats (200-250 g) were randomly assigned into three groups (n = 8) by simple random sampling method. Control (C), diabetic (D), and diabetic administrated with TMZ at 10 mg/kg (T10). TMZ was administrated for 8 weeks. The echocardiogram was recorded before isolating the hearts and transfer to a Langendorff apparatus. Hemodynamic parameters, QT and corrected QT interval (QTc) intervals, heart rate and antioxidant enzymes were measured. The hypertrophy index was calculated. The results were evaluated by one-way ANOVA and paired t-test using SPSS (version 16) and p < 0.05 was regarded as significant. RESULTS: The diabetic rats significantly indicated increased hypertrophy, QT and QTc intervals and decreased Left ventricular systolic pressure (LVSP), Left ventricular developed pressure (LVDP), rate pressure product (RPP), Max dp/dt, and min dp/dt (±dp/dt max), heart rate, superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase in the heart. Treatment with TMZ in the diabetic animals was significantly improved these parameters in comparison to the untreated diabetic group. CONCLUSIONS: TMZ improves QTc interval prolongation and cardiac hypertrophy in diabetes.


Assuntos
Cardiomegalia/tratamento farmacológico , Complicações do Diabetes/tratamento farmacológico , Síndrome do QT Longo/tratamento farmacológico , Substâncias Protetoras/farmacologia , Trimetazidina/farmacologia , Animais , Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Catalase/análise , Complicações do Diabetes/enzimologia , Complicações do Diabetes/fisiopatologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/fisiopatologia , Ecocardiografia , Glutationa Peroxidase/análise , Hemodinâmica/efeitos dos fármacos , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Masculino , Distribuição Aleatória , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Superóxido Dismutase/análise , Fatores de Tempo
5.
Trends Cardiovasc Med ; 27(7): 451-459, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28687226

RESUMO

While it is well known that mutation of several different ion channels can cause congenital long QT syndrome, block of IKr is widely thought to be responsible for most cases of drug-induced acquired long QT syndrome (aLQTS). In this article, we review evidence supporting another cause of aLQTS due to inhibition of phosphoinositide 3-kinase (PI3K) signaling. Inhibition of PI3K affects multiple plateau currents, reducing IKr, IKs, and ICaL while increasing the persistent sodium current (INaP). The effects of PI3K inhibitors develop slowly, requiring hours to days to reach steady state. Dofetilide and terfenadine, an antihistamine on which much of the original IKr hypothesis was based, are among the many drugs that inhibit the PI3K pathway. Reduced PI3K signaling may also play a role in aLQTS associated with diabetes. Drug safety testing to identify aLQTS risk may be improved by examining PI3K-dependent effects that develop over time.


Assuntos
Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Síndrome do QT Longo/induzido quimicamente , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/efeitos adversos , Potenciais de Ação , Animais , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Fosfatidilinositol 3-Quinase/metabolismo , Bloqueadores dos Canais de Potássio/efeitos adversos , Fatores de Risco , Bloqueadores dos Canais de Sódio/efeitos adversos
6.
J Cell Sci ; 127(Pt 18): 3943-55, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25037568

RESUMO

KCNQ1 and KCNE1 co-assembly generates the I(KS) K(+) current, which is crucial to the cardiac action potential repolarization. Mutations in their corresponding genes cause long QT syndrome (LQT) and atrial fibrillation. The A-kinase anchor protein, yotiao (also known as AKAP9), brings the I(KS) channel complex together with signaling proteins to achieve regulation upon ß1-adrenergic stimulation. Recently, we have shown that KCNQ1 helix C interacts with the KCNE1 distal C-terminus. We postulated that this interface is crucial for I(KS) channel modulation. Here, we examined the yet unknown molecular mechanisms of LQT mutations located at this intracellular intersubunit interface. All LQT mutations disrupted the internal KCNQ1-KCNE1 intersubunit interaction. LQT mutants in KCNQ1 helix C led to a decreased current density and a depolarizing shift of channel activation, mainly arising from impaired phosphatidylinositol-4,5-bisphosphate (PIP2) modulation. In the KCNE1 distal C-terminus, the LQT mutation P127T suppressed yotiao-dependent cAMP-mediated upregulation of the I(KS) current, which was caused by reduced KCNQ1 phosphorylation at S27. Thus, KCNQ1 helix C is important for channel modulation by PIP2, whereas the KCNE1 distal C-terminus appears essential for the regulation of IKS by yotiao-mediated PKA phosphorylation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/genética , Mutação de Sentido Incorreto , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Humanos , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/metabolismo , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , Estrutura Secundária de Proteína
8.
Cardiovasc Res ; 100(3): 520-8, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23963841

RESUMO

AIMS: We examined the role of Hsp90 in expression and maturation of wild-type (WT) and mutant ether-a-go-go related gene (HERG) proteins by using Hsp90 inhibitors, geldanamycin (GA) and radicicol, and Hsp90 overexpression. METHODS AND RESULTS: The proteins were expressed in HEK293 cells or collected from HL-1 mouse cardiomyocytes, and analysed by western blotting, immunoprecipitation, immunofluorescence, and whole-cell patch-clamp techniques. GA and radicicol suppressed maturation of HERG-FLAG proteins and increased their immature forms. Co-expression of Hsp90 counteracted the effects of Hsp90 inhibitors and suppressed ubiquitination of HERG proteins. Overexpressed Hsp90 also inhibited the binding of endogenous C-terminus of Hsp70-interacting protein (CHIP) to HERG-FLAG proteins. Hsp90-induced increase of functional HERG proteins was verified by their increased expression on the cell surface and enhanced HERG channel currents. CHIP overexpression decreased both mature and immature forms of HERG-FLAG proteins in cells treated with GA. Hsp90 facilitated maturation of endogenous ERG proteins, whereas CHIP decreased both forms of ERG proteins in HL-1 cells. Mutant HERG proteins harbouring disease-causing missense mutations were mainly in the immature form and had a higher binding capacity to CHIP than the WT; Hsp90 overexpression suppressed this association. Overexpressed Hsp90 increased the mature form of HERG(1122fs/147) proteins, reduced its ubiquitinated form, increased its immunoreactivity in the endoplasmic reticulum and on the plasma membrane, and increased the mutant-mediated membrane current. CHIP overexpression decreased the immature form of HERG(1122fs/147) proteins. CONCLUSION: Enhancement of HERG protein expression through Hsp90 inhibition of CHIP binding might be a novel therapeutic strategy for long QT syndrome 2 caused by trafficking abnormalities of HERG proteins.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Miócitos Cardíacos/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Benzoquinonas/farmacologia , Membrana Celular/enzimologia , Canal de Potássio ERG1 , Retículo Endoplasmático/enzimologia , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Humanos , Lactamas Macrocíclicas/farmacologia , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Macrolídeos/farmacologia , Potenciais da Membrana , Camundongos , Mutação de Sentido Incorreto , Miócitos Cardíacos/efeitos dos fármacos , Transporte Proteico , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
9.
Drug Saf ; 36(5): 295-316, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23620167

RESUMO

The development of tyrosine kinase inhibitors (TKI) represents a major milestone in oncology. However, their use has been found to be associated with serious toxicities that impinge on various vital organs including the heart. Sixteen TKIs have been approved for use in oncology as of 30 September 2012, and a large number of others are in development or under regulatory review. Cardiovascular safety of medicinal products is a major public health issue that has concerned all the stakeholders. This review focuses on three specific cardiovascular safety aspects of TKIs, namely their propensity to induce QT interval prolongation, left ventricular (LV) dysfunction and hypertension (both systemic and pulmonary). Analyses of information in drug labels, the data submitted to the regulatory authorities and the published literature show that a number of TKIs are associated with these undesirable effects. Whereas LV dysfunction and systemic hypertension are on-target effects related to the inhibition of ligand-related signalling pathways, QT interval prolongation appears to be an off-target class III electrophysiologic effect, possibly related to the presence of a fluorine-based pharmacophore. If not adequately managed, these cardiovascular effects significantly increase the morbidity and mortality in a population already at high risk. Hitherto, the QT effect of most QT-prolonging TKIs (except lapatinib, nilotinib, sunitinib and vandetanib) is relatively mild at clinical doses and has not led to appreciable morbidity clinically. In contrast, LV dysfunction and untreated hypertension have resulted in significant morbidity. Inevitably, dilemmas arise in determining the risk/benefit of a TKI therapy in an individual patient who develops any of these effects following the treatment of the TKI-sensitive cancer. QT interval prolongation, hypertension and LV dysfunction can be managed effectively by using reliable methods of measurement and careful monitoring of patients whose clinical management requires optimisation by a close collaboration between an oncologist and a cardiologist, an evolving subspecialty referred to as cardio-oncology. Despite their potential adverse clinical impact, the effects of TKIs on hypertension and LV function are generally inadequately characterised during their development. As has been the case with QT liability of drugs, there is now a persuasive case for a regulatory requirement to study TKIs systematically for these effects. Furthermore, since most of these novel drugs are studied in trials with relatively small sample sizes and approved on an expedited basis, there is also a compelling case for their effective pharmacovigilance and on-going reassessment of their risk/benefit after approval.


Assuntos
Cardiotoxinas/efeitos adversos , Doenças Cardiovasculares/induzido quimicamente , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Cardiotoxinas/farmacocinética , Doenças Cardiovasculares/enzimologia , Sistema de Condução Cardíaco/enzimologia , Frequência Cardíaca/fisiologia , Humanos , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/enzimologia , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo
10.
Sci Transl Med ; 4(131): 131ra50, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539774

RESUMO

Many drugs, including some commonly used medications, can cause abnormal heart rhythms and sudden death, as manifest by a prolonged QT interval in the electrocardiogram. Cardiac arrhythmias caused by drug-induced long QT syndrome are thought to result mainly from reductions in the delayed rectifier potassium ion (K(+)) current I(Kr). Here, we report a mechanism for drug-induced QT prolongation that involves changes in multiple ion currents caused by a decrease in phosphoinositide 3-kinase (PI3K) signaling. Treatment of canine cardiac myocytes with inhibitors of tyrosine kinases or PI3Ks caused an increase in action potential duration that was reversed by intracellular infusion of phosphatidylinositol 3,4,5-trisphosphate. The inhibitors decreased the delayed rectifier K(+) currents I(Kr) and I(Ks), the L-type calcium ion (Ca(2+)) current I(Ca,L), and the peak sodium ion (Na(+)) current I(Na) and increased the persistent Na(+) current I(NaP). Computer modeling of the canine ventricular action potential showed that the drug-induced change in any one current accounted for less than 50% of the increase in action potential duration. Mouse hearts lacking the PI3K p110α catalytic subunit exhibited a prolonged action potential and QT interval that were at least partly a result of an increase in I(NaP). These results indicate that down-regulation of PI3K signaling directly or indirectly via tyrosine kinase inhibition prolongs the QT interval by affecting multiple ion channels. This mechanism may explain why some tyrosine kinase inhibitors in clinical use are associated with increased risk of life-threatening arrhythmias.


Assuntos
Síndrome do QT Longo/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/toxicidade , Transdução de Sinais/efeitos dos fármacos , Potenciais de Ação , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Classe I de Fosfatidilinositol 3-Quinases , Simulação por Computador , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/metabolismo , Cães , Eletrocardiografia , Feminino , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Modelos Cardiovasculares , Miócitos Cardíacos/enzimologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Medição de Risco , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Fatores de Tempo
11.
Nat Med ; 17(12): 1657-62, 2011 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-22120178

RESUMO

Monogenic neurodevelopmental disorders provide key insights into the pathogenesis of disease and help us understand how specific genes control the development of the human brain. Timothy syndrome is caused by a missense mutation in the L-type calcium channel Ca(v)1.2 that is associated with developmental delay and autism. We generated cortical neuronal precursor cells and neurons from induced pluripotent stem cells derived from individuals with Timothy syndrome. Cells from these individuals have defects in calcium (Ca(2+)) signaling and activity-dependent gene expression. They also show abnormalities in differentiation, including decreased expression of genes that are expressed in lower cortical layers and in callosal projection neurons. In addition, neurons derived from individuals with Timothy syndrome show abnormal expression of tyrosine hydroxylase and increased production of norepinephrine and dopamine. This phenotype can be reversed by treatment with roscovitine, a cyclin-dependent kinase inhibitor and atypical L-type-channel blocker. These findings provide strong evidence that Ca(v)1.2 regulates the differentiation of cortical neurons in humans and offer new insights into the causes of autism in individuals with Timothy syndrome.


Assuntos
Sinalização do Cálcio , Células-Tronco Pluripotentes Induzidas/citologia , Síndrome do QT Longo/fisiopatologia , Neurônios/citologia , Sindactilia/fisiopatologia , Tirosina 3-Mono-Oxigenase/genética , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular , Dopamina/metabolismo , Regulação da Expressão Gênica , Humanos , Síndrome do QT Longo/enzimologia , Análise em Microsséries , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Norepinefrina/metabolismo , Fenótipo , Purinas/farmacologia , Roscovitina , Sindactilia/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Sci China Life Sci ; 54(8): 691-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21786192

RESUMO

The plasma membrane calcium ATPases (PMCA) are a family of genes which extrude Ca(2+) from the cell and are involved in the maintenance of intracellular free calcium levels and/or with Ca(2+) signalling, depending on the cell type. In the cardiovascular system, Ca(2+) is not only essential for contraction and relaxation but also has a vital role as a second messenger in signal transduction pathways. A complex array of mechanisms regulate intracellular free calcium levels in the heart and vasculature and a failure in these systems to maintain normal Ca(2+) homeostasis has been linked to both heart failure and hypertension. This article focuses on the functions of PMCA, in particular isoform 4 (PMCA4), in the heart and vasculature and the reported links between PMCAs and contractile function, cardiac hypertrophy, cardiac rhythm and sudden cardiac death, and blood pressure control and hypertension. It is becoming clear that this family of calcium extrusion pumps have essential roles in both cardiovascular health and disease.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Isoenzimas/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Doenças Cardiovasculares/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Homeostase , Humanos , Isoenzimas/genética , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Contração Muscular/fisiologia , Músculo Liso Vascular/enzimologia , Miocárdio/metabolismo , Miocárdio/patologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética
13.
Am J Physiol Heart Circ Physiol ; 301(3): H813-23, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21685264

RESUMO

Patients with very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency frequently present cardiomyopathy and heartbeat disorders. However, the underlying factors, which may be of cardiac or extra cardiac origins, remain to be elucidated. In this study, we tested for metabolic and functional alterations in the heart from 3- and 7-mo-old VLCAD null mice and their littermate counterparts, using validated experimental paradigms, namely, 1) ex vivo perfusion in working mode, with concomitant evaluation of myocardial contractility and metabolic fluxes using (13)C-labeled substrates under various conditions; as well as 2) in vivo targeted lipidomics, gene expression analysis as well as electrocardiogram monitoring by telemetry in mice fed various diets. Unexpectedly, when perfused ex vivo, working VLCAD null mouse hearts maintained values similar to those of the controls for functional parameters and for the contribution of exogenous palmitate to ß-oxidation (energy production), even at high palmitate concentration (1 mM) and increased energy demand (with 1 µM epinephrine) or after fasting. However, in vivo, these hearts displayed a prolonged rate-corrected QT (QTc) interval under all conditions examined, as well as the following lipid alterations: 1) age- and condition-dependent accumulation of triglycerides, and 2) 20% lower docosahexaenoic acid (an omega-3 polyunsaturated fatty acid) in membrane phospholipids. The latter was independent of liver but affected by feeding a diet enriched in saturated fat (exacerbated) or fish oil (attenuated). Our finding of a longer QTc interval in VLCAD null mice appears to be most relevant given that such condition increases the risk of sudden cardiac death.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Sistema de Condução Cardíaco/fisiopatologia , Metabolismo dos Lipídeos/genética , Síndrome do QT Longo/enzimologia , Erros Inatos do Metabolismo/enzimologia , Doenças Mitocondriais/enzimologia , Doenças Musculares/enzimologia , Miocárdio/enzimologia , Acil-CoA Desidrogenase de Cadeia Longa/genética , Fatores Etários , Envelhecimento , Análise de Variância , Animais , Síndrome Congênita de Insuficiência da Medula Óssea , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/metabolismo , Eletrocardiografia Ambulatorial , Óleos de Peixe/administração & dosagem , Óleos de Peixe/metabolismo , Erros Inatos do Metabolismo Lipídico , Fígado/metabolismo , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Síndrome do QT Longo/prevenção & controle , Masculino , Erros Inatos do Metabolismo/complicações , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Mitocondriais/complicações , Doenças Mitocondriais/genética , Doenças Mitocondriais/fisiopatologia , Doenças Musculares/complicações , Doenças Musculares/genética , Doenças Musculares/fisiopatologia , Contração Miocárdica , Oxirredução , Ácido Palmítico/metabolismo , Perfusão , Telemetria , Triglicerídeos/metabolismo
14.
J Clin Pharmacol ; 51(9): 1256-63, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21148043

RESUMO

Tasocitinib (CP-690,550), a selective inhibitor of the Janus kinase (JAK) family, is being developed for the treatment of several autoimmune diseases and prevention of allograft rejection. The aim of this study was to characterize the effect of tasocitinib on QT interval. Sixty male and female healthy adults were enrolled in a single-dose, randomized, 3-period, crossover study of a supratherapeutic dose of tasocitinib (100 mg), placebo, and moxifloxacin 400 mg. Triplicate electrocardiograms were performed at predose baseline and serially over 24 hours postdose in each treatment period. The upper limits of the 2-sided 90% confidence intervals (CIs) for the difference in QTc interval, corrected using Fridericia correction (QTcF), between tasocitinib and placebo were less than 5 ms at all time points. Concentration-QTcF analysis showed that the predicted mean change (90% CI) in QTcF at the observed mean C(max) was -0.12 (-1.18, 0.94) ms. For moxifloxacin, mean (90% CI) estimates of the change in QTcF from placebo were 11.3 (9.4, 13.1) and 12.5 (10.7, 14.4) ms at 2 and 4 hours, respectively, thereby establishing study sensitivity. A single supratherapeutic dose of tasocitinib 100 mg was well tolerated and not associated with QTc prolongation.


Assuntos
Frequência Cardíaca/efeitos dos fármacos , Janus Quinase 3/antagonistas & inibidores , Síndrome do QT Longo , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Sístole/efeitos dos fármacos , Adulto , Estudos Cross-Over , Método Duplo-Cego , Feminino , Seguimentos , Frequência Cardíaca/fisiologia , Humanos , Janus Quinase 3/metabolismo , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Masculino , Pessoa de Meia-Idade , Piperidinas , Pirimidinas/efeitos adversos , Pirróis/efeitos adversos , Sístole/fisiologia , Adulto Jovem
15.
J Biol Chem ; 286(4): 2843-52, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21097842

RESUMO

The most common cause of cardiac side effects of pharmaco-therapy is acquired long QT syndrome, which is characterized by abnormal cardiac repolarization and most often caused by direct blockade of the cardiac potassium channel human ether a-go-go-related gene (hERG). However, little is known about therapeutic compounds that target ion channels other than hERG. We have discovered that arsenic trioxide (As(2)O(3)), a very potent antineoplastic compound for the treatment of acute promyelocytic leukemia, is proarrhythmic via two separate mechanisms: a well characterized inhibition of hERG/I(Kr) trafficking and a poorly understood increase of cardiac calcium currents. We have analyzed the latter mechanism in the present study using biochemical and electrophysiological methods. We find that oxidative inactivation of the lipid phosphatase PTEN by As(2)O(3) enhances cardiac calcium currents in the therapeutic concentration range via a PI3Kα-dependent increase in phosphatidylinositol 3,4,5-triphosphate (PIP(3)) production. In guinea pig ventricular myocytes, even a modest reduction in PTEN activity is sufficient to increase cellular PIP(3) levels. Under control conditions, PIP(3) levels are kept low by PTEN and do not affect calcium current amplitudes. Based on pharmacological experiments and intracellular infusion of PIP(3), we propose that in guinea pig ventricular myocytes, PIP(3) regulates calcium currents independently of the protein kinase Akt along a pathway that includes a secondary oxidation-sensitive target. Overall, our report describes a novel form of acquired long QT syndrome where the target modified by As(2)O(3) is an intracellular signaling cascade.


Assuntos
Antineoplásicos/efeitos adversos , Arsenicais/efeitos adversos , Cálcio/metabolismo , Ventrículos do Coração/enzimologia , Síndrome do QT Longo/enzimologia , Miócitos Cardíacos/enzimologia , Óxidos/efeitos adversos , PTEN Fosfo-Hidrolase/metabolismo , Animais , Antineoplásicos/farmacologia , Trióxido de Arsênio , Arsenicais/farmacologia , Células Cultivadas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Cobaias , Humanos , Síndrome do QT Longo/induzido quimicamente , Oxirredução/efeitos dos fármacos , Óxidos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Life Sci ; 84(11-12): 380-7, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19167409

RESUMO

AIMS: In the type 3 long QT syndrome (LQT3), shortening of the QT interval by overdrive pacing is used to prevent life-threatening arrhythmias. However, it is unclear whether accelerated heart rate induced by beta-adrenergic agents produces similar effects on the late sodium current (I(Na)) to those by overdrive pacing therapy. We analyzed the beta-adrenergic-like effects of protein kinase A and fluoride on I(Na) in R1623Q mutant channels. MAIN METHODS: cDNA encoding either wild-type (WT) or R1623Q mutant of hNa(v)1.5 was stably transfected into HEK293 cells. I(Na) was recorded using a whole-cell patch-clamp technique at 23 degrees C. KEY FINDINGS: In R1623Q channels, 2 mM pCPT-AMP and 120 mM fluoride significantly delayed macroscopic current decay and increased relative amplitude of the late I(Na) in a time-dependent manner. Modulations of peak I(Na) gating kinetics (activation, inactivation, recovery from inactivation) by fluoride were similar in WT and R1623Q channels. The effects of fluoride were almost completely abolished by concomitant dialysis with a protein kinase inhibitor. We also compared the effect of pacing with that of beta-adrenergic stimulation by analyzing the frequency-dependence of the late I(Na). Fluoride augmented frequency-dependent reduction of the late I(Na), which was due to preferential delay of recovery of late I(Na). However, the increase in late I(Na) by fluoride at steady-state was more potent than the frequency-dependent reduction of late I(Na). SIGNIFICANCE: Different basic mechanisms participate in the QT interval shortening by pacing and beta-adrenergic stimulation in the LQT3.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação do Canal Iônico , Síndrome do QT Longo/metabolismo , Proteínas Musculares/genética , Canais de Sódio/genética , Potenciais de Ação/efeitos dos fármacos , Linhagem Celular , Clonagem Molecular , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Fluoretos/farmacologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Inibidores de Proteínas Quinases/farmacologia , Tionucleotídeos/farmacologia , Transfecção
17.
Proc Natl Acad Sci U S A ; 105(27): 9355-60, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18591664

RESUMO

Mutations in 11 genes that encode ion channels or their associated proteins cause inherited long QT syndrome (LQTS) and account for approximately 75-80% of cases (LQT1-11). Direct sequencing of SNTA1, the gene encoding alpha1-syntrophin, was performed in a cohort of LQTS patients that were negative for mutations in the 11 known LQTS-susceptibility genes. A missense mutation (A390V-SNTA1) was found in a patient with recurrent syncope and markedly prolonged QT interval (QTc, 530 ms). SNTA1 links neuronal nitric oxide synthase (nNOS) to the nNOS inhibitor plasma membrane Ca-ATPase subtype 4b (PMCA4b); SNTA1 also is known to associate with the cardiac sodium channel SCN5A. By using a GST-fusion protein of the C terminus of SCN5A, we showed that WT-SNTA1 interacted with SCN5A, nNOS, and PMCA4b. In contrast, A390V-SNTA1 selectively disrupted association of PMCA4b with this complex and increased direct nitrosylation of SCN5A. A390V-SNTA1 expressed with SCN5A, nNOS, and PMCA4b in heterologous cells increased peak and late sodium current compared with WT-SNTA1, and the increase was partially inhibited by NOS blockers. Expression of A390V-SNTA1 in cardiac myocytes also increased late sodium current. We conclude that the A390V mutation disrupted binding with PMCA4b, released inhibition of nNOS, caused S-nitrosylation of SCN5A, and was associated with increased late sodium current, which is the characteristic biophysical dysfunction for sodium-channel-mediated LQTS (LQT3). These results establish an SNTA1-based nNOS complex attached to SCN5A as a key regulator of sodium current and suggest that SNTA1 be considered a rare LQTS-susceptibility gene.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Predisposição Genética para Doença , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Proteínas de Membrana/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Canais de Sódio/metabolismo , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/química , Linhagem Celular , Ativação Enzimática , Feminino , Genótipo , Humanos , Ativação do Canal Iônico , Substâncias Macromoleculares/metabolismo , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Mutantes/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5 , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Canais de Sódio/química
18.
J Am Geriatr Soc ; 56(5): 792-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18363676

RESUMO

OBJECTIVES: To determine whether high levels of lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) are associated with prevalent cardiovascular disease (CVD) and to evaluate factors most influencing Lp-PLA(2) levels in a community-based cohort of older adults. DESIGN: Cross-sectional. SETTING: The Cardiovascular Health Study (CHS), a population-based cohort study of men and women aged 65 and older. PARTICIPANTS: Five thousand five hundred thirty-one CHS participants. MEASUREMENTS: Levels of Lp-PLA(2) activity were determined using stored blood samples from the baseline examination. RESULTS: Mean Lp-PLA(2) was higher in participants with electrocardiographically determined ventricular conduction defect and major Q-wave abnormality and was positively correlated with left ventricular (LV) mass. It was high in those with echocardiographically determined abnormal LV ejection fraction, which persisted after adjustment. Mean Lp-PLA(2) was also higher in participants with mild renal insufficiency and kidney disease. After multivariable adjustment, there was a modest but significant 27% greater risk of prevalent CHF per standard deviation increment of Lp-PLA(2) and a modest but significant 12% greater risk of prevalent myocardial infarction. Lp-PLA(2) was weakly but mainly most strongly correlated with cholesterol and lipoproteins, but those correlations were not especially strong. Lp-PLA(2) was weakly positively correlated with soluble intercellular adhesion molecule-1 but not interleukin-6. In total, all factors considered could explain only 29% of Lp-PLA(2) activity. CONCLUSION: Novel findings in the study are the associations, in those aged 65 and older, between Lp-PLA(2) activity and LV dysfunction, CHF, and renal disease. CVD risk factors only minimally explain levels of Lp-PLA(2).


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/sangue , Doenças Cardiovasculares/enzimologia , Idoso , Aterosclerose/diagnóstico , Aterosclerose/enzimologia , Aterosclerose/epidemiologia , Índice de Massa Corporal , Baixo Débito Cardíaco/diagnóstico , Baixo Débito Cardíaco/enzimologia , Baixo Débito Cardíaco/epidemiologia , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/epidemiologia , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Estudos de Coortes , Estudos Transversais , Eletrocardiografia , Feminino , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/epidemiologia , Humanos , Hipertrofia Ventricular Esquerda/diagnóstico , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/epidemiologia , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/epidemiologia , Masculino , Valores de Referência , Insuficiência Renal/diagnóstico , Insuficiência Renal/enzimologia , Insuficiência Renal/epidemiologia , Fatores de Risco , Estatística como Assunto , Triglicerídeos/sangue
19.
Biochemistry ; 46(41): 11459-72, 2007 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17892302

RESUMO

KCNE1, also known as minK, is a member of the KCNE family of membrane proteins that modulate the function of KCNQ1 and certain other voltage-gated potassium channels (KV). Mutations in human KCNE1 cause congenital deafness and congenital long QT syndrome, an inherited predisposition to potentially life-threatening cardiac arrhythmias. Although its modulation of KCNQ1 function has been extensively characterized, many questions remain regarding KCNE1's structure and location within the channel complex. In this study, KCNE1 was overexpressed in Escherichia coli and purified. Micellar solutions of the protein were then microinjected into Xenopus oocytes expressing KCNQ1 channels, followed by electrophysiological recordings aimed at testing whether recombinant KCNE1 can co-assemble with the channel. Nativelike modulation of channel properties was observed following injection of KCNE1 in lyso-myristoylphosphatidylglycerol (LMPG) micelles, indicating that KCNE1 is not irreversibly misfolded and that LMPG is able to act as a vehicle for delivering membrane proteins into the membranes of viable cells. 1H-15N TROSY NMR experiments indicated that LMPG micelles are well-suited for structural studies of KCNE1, leading to assignment of its backbone resonances and to relaxation studies. The chemical shift data confirmed that KCNE1's secondary structure includes several alpha-helices and demonstrated that its distal C-terminus is disordered. Surprisingly, for KCNE1 in LMPG micelles, there appears to be a break in alpha-helicity at sites 59-61, near the middle of the transmembrane segment, a feature that is accompanied by increased local backbone mobility. Given that this segment overlaps with sites 57-59, which are known to play a critical role in modulating KCNQ1 channel activation kinetics, this unusual structural feature likely has considerable functional relevance.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Sequência de Aminoácidos , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/genética , Clonagem Molecular , Surdez/enzimologia , Surdez/genética , Escherichia coli , Feminino , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Oócitos/fisiologia , Reação em Cadeia da Polimerase , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Soluções , Xenopus/fisiologia
20.
Expert Opin Pharmacother ; 7(12): 1583-90, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16872261

RESUMO

Individuals vary widely in their responses to therapy with most drugs. Indeed, responses to antiarrhythmic drugs are so highly variable that study of the underlying mechanisms has elucidated important lessons for understanding variable responses to drug therapy in general. Variability in drug response may reflect variability in the relationship between a drug dose and the concentrations of the drug and metabolite(s) at relevant target sites; this is termed pharmacokinetic variability. Another mechanism is that individuals vary in their response to identical exposures to a drug (pharmacodynamic variability). In this case, there may be variability in the target molecule(s) with which a drug interacts or, more generally, in the broad biological context in which the drug-target interaction occurs. Variants (polymorphisms and mutations) in the genes that encode proteins that are important for pharmacokinetics or for pharmacodynamics have now been described as important contributors to variable drug actions, including proarrhythmia, and these are described in this review. However, the translation of pharmacogenetics into clinical practice has been slow. To this end, the creation of large, well-characterised DNA databases and appropriate control groups, as well as large prospective trials to evaluate the impact of genetic variation on drug therapy, may hasten the impact of pharmacogenetics and pharmacogenomics in terms of delivering personalised drug therapy and to avoid therapeutic failure and serious side effects.


Assuntos
Antiarrítmicos/efeitos adversos , Arritmias Cardíacas/etiologia , Síndrome do QT Longo/etiologia , Assistência Individualizada de Saúde/tendências , Farmacogenética/tendências , Torsades de Pointes/etiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antiarrítmicos/farmacocinética , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/genética , Arilamina N-Acetiltransferase/genética , Arilamina N-Acetiltransferase/metabolismo , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Digoxina/efeitos adversos , Digoxina/farmacocinética , Humanos , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/genética , Polimorfismo Genético , Canais de Potássio/genética , Canais de Potássio/metabolismo , Propafenona/efeitos adversos , Propafenona/farmacocinética , Fatores de Risco , Bloqueadores dos Canais de Sódio/efeitos adversos , Bloqueadores dos Canais de Sódio/farmacocinética , Canais de Sódio/genética , Canais de Sódio/metabolismo , Torsades de Pointes/enzimologia , Torsades de Pointes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA