Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 714: 149947, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38657442

RESUMEN

Here, we characterized the p.Arg583His (R583H) Kv7.1 mutation, identified in two unrelated families suffered from LQT syndrome. This mutation is located in the HС-HD linker of the cytoplasmic portion of the Kv7.1 channel. This linker, together with HD helix are responsible for binding the A-kinase anchoring protein 9 (AKAP9), Yotiao. We studied the electrophysiological characteristics of the mutated channel expressed in CHO-K1 along with KCNE1 subunit and Yotiao protein, using the whole-cell patch-clamp technique. We found that R583H mutation, even at the heterozygous state, impedes IKs activation. Molecular modeling showed that HС and HD helixes of the C-terminal part of Kv7.1 channel are swapped along the C-terminus length of the channel and that R583 position is exposed to the outer surface of HC-HD tandem coiled-coil. Interestingly, the adenylate cyclase activator, forskolin had a smaller effect on the mutant channel comparing with the WT protein, suggesting that R583H mutation may disrupt the interaction of the channel with the adaptor protein Yotiao and, therefore, may impair phosphorylation of the KCNQ1 channel.


Asunto(s)
Proteínas de Anclaje a la Quinasa A , Proteínas del Citoesqueleto , Canal de Potasio KCNQ1 , Síndrome de QT Prolongado , Animales , Femenino , Humanos , Masculino , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/química , Células CHO , Cricetulus , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Canal de Potasio KCNQ1/química , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Modelos Moleculares , Mutación , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Unión Proteica
2.
Europace ; 26(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38788213

RESUMEN

AIMS: Human induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCM) could be a helpful tool to study the physiology and diseases of the human atrium. To fulfil this expectation, the electrophysiology of hiPSC-aCM should closely resemble the situation in the human atrium. Data on the contribution of the slowly activating delayed rectifier currents (IKs) to repolarization are lacking for both human atrium and hiPSC-aCM. METHODS AND RESULTS: Human atrial tissues were obtained from patients with sinus rhythm (SR) or atrial fibrillation (AF). Currents were measured in human atrial cardiomyocytes (aCM) and compared with hiPSC-aCM and used to model IKs contribution to action potential (AP) shape. Action potential was recorded by sharp microelectrodes. HMR-1556 (1 µM) was used to identify IKs and to estimate IKs contribution to repolarization. Less than 50% of hiPSC-aCM and aCM possessed IKs. Frequency of occurrence, current densities, activation/deactivation kinetics, and voltage dependency of IKs did not differ significantly between hiPSC-aCM and aCM, neither in SR nor AF. ß-Adrenoceptor stimulation with isoprenaline did not increase IKs neither in aCM nor in hiPSC-aCM. In tissue from SR, block of IKs with HMR-1556 did not lengthen the action potential duration, even when repolarization reserve was reduced by block of the ultra-rapid repolarizing current with 4-aminopyridine or the rapidly activating delayed rectifier potassium outward current with E-4031. CONCLUSION: I Ks exists in hiPSC-aCM with biophysics not different from aCM. As in adult human atrium (SR and AF), IKs does not appear to relevantly contribute to repolarization in hiPSC-aCM.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial , Canales de Potasio de Tipo Rectificador Tardío , Atrios Cardíacos , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Atrios Cardíacos/fisiopatología , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/metabolismo , Femenino , Células Cultivadas , Masculino , Persona de Mediana Edad , Cinética , Anciano , Diferenciación Celular , Modelos Cardiovasculares , Bloqueadores de los Canales de Potasio/farmacología
3.
J Anesth ; 37(5): 806-810, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37524993

RESUMEN

This simulation study was designed to predict the torsadogenicity of sevoflurane and propofol in healthy control, as well as type 1 and type 2 long QT syndrome (LQT1 and LQT2, respectively), using the O'Hara-Rudy dynamic model. LQT1 and LQT2 models were simulated by decreasing the conductances of slowly and rapidly activating delayed rectifier K+ currents (IKs and IKr, respectively) by 50%, respectively. Action potential duration at 50% repolarization level (APD50) and diastolic intracellular Ca2+ concentration were measured in epicardial cell during administration of sevoflurane (1 ~ 5%) and propofol (1 ~ 10 µM). Torsadogenicity can be predicted from the relationship between APD50 and diastolic intracellular Ca2+ concentration, which is classified by the decision boundary. Whereas the relationships in control and LQT1 models were distributed on nontorsadogenic side in the presence of sevoflurane at all tested concentrations, those in LQT2 models were shifted to torsadogenic side by concentrations of ≥ 2%. In all three models, propofol shifted the relationships in a direction away from the decision boundary on nontorsadogenic side. Our findings suggest that sevoflurane, but not propofol, exerts torsadogenicity in patients with reduced IKr, such as LQT2 patients. Caution should be paid to the occurrence of arrhythmia during sevoflurane anesthesia in patients with reduced IKr.


Asunto(s)
Anestésicos , Síndrome de QT Prolongado , Propofol , Humanos , Sevoflurano/efectos adversos , Propofol/efectos adversos , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/metabolismo , Anestésicos/farmacología , Potenciales de Acción
4.
J Mol Cell Cardiol ; 161: 86-97, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34375616

RESUMEN

Delayed rectifier K+ current (IKs) is a key contributor to repolarization of action potentials. This study investigated the mechanisms underlying the adrenoceptor-induced potentiation of IKs in pulmonary vein cardiomyocytes (PVC). PVC were isolated from guinea pig pulmonary vein. The action potentials and IKs current were recorded using perforated and conventional whole-cell patch-clamp techniques. The expression of IKs was examined using immunocytochemistry and Western blotting. KCNQ1, a IKs pore-forming protein was detected as a signal band approximately 100 kDa in size, and its immunofluorescence signal was found to be mainly localized on the cell membrane. The IKs current in PVC was markedly enhanced by both ß1- and ß2-adrenoceptor stimulation with a negative voltage shift in the current activation, although the potentiation was more effectively induced by ß2-adrenoceptor stimulation than ß1-adrenoceptor stimulation. Both ß-adrenoceptor-mediated increases in IKs were attenuated by treatment with the adenylyl cyclase (AC) inhibitor or protein kinase A (PKA) inhibitor. Furthermore, the IKs current was increased by α1-adrenoceptor agonist but attenuated by the protein kinase C (PKC) inhibitor. PVC exhibited action potentials in normal Tyrode solution which was slightly reduced by HMR-1556 a selective IKs blocker. However, HMR-1556 markedly reduced the ß-adrenoceptor-potentiated firing rate. The stimulatory effects of ß- and α1-adrenoceptor on IKs in PVC are mediated via the PKA and PKC signal pathways. HMR-1556 effectively reduced the firing rate under ß-adrenoceptor activation, suggesting that the functional role of IKs might increase during sympathetic excitation under in vivo conditions.


Asunto(s)
Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Miocitos Cardíacos/metabolismo , Venas Pulmonares/metabolismo , Receptores Adrenérgicos/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Cobayas , Atrios Cardíacos/metabolismo , Isoproterenol/farmacología , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Proteína Quinasa C/metabolismo , Venas Pulmonares/citología , Transducción de Señal
5.
Pflugers Arch ; 473(12): 1885-1898, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34704178

RESUMEN

The atrioventricular (AV) node is the only conduction pathway where electrical impulse can pass from atria to ventricles and exhibits spontaneous automaticity. This study examined the function of the rapid- and slow-activating delayed rectifier K+ currents (IKr and IKs) in the regulation of AV node automaticity. Isolated AV node cells from guinea pigs were current- and voltage-clamped to record the action potentials and the IKr and IKs current. The expression of IKr or IKs was confirmed in the AV node cells by immunocytochemistry, and the positive signals of both channels were localized mainly on the cell membrane. The basal spontaneous automaticity was equally reduced by E4031 and HMR-1556, selective blockers of IKr and IKs, respectively. The nonselective ß-adrenoceptor agonist isoproterenol markedly increased the firing rate of action potentials. In the presence of isoproterenol, the firing rate of action potentials was more effectively reduced by the IKs inhibitor HMR-1556 than by the IKr inhibitor E4031. Both E4031 and HMR-1556 prolonged the action potential duration and depolarized the maximum diastolic potential under basal and ß-adrenoceptor-stimulated conditions. IKr was not significantly influenced by ß-adrenoceptor stimulation, but IKs was concentration-dependently enhanced by isoproterenol (EC50: 15 nM), with a significant negative voltage shift in the channel activation. These findings suggest that both the IKr and IKs channels might exert similar effects on regulating the repolarization process of AV node action potentials under basal conditions; however, when the ß-adrenoceptor is activated, IKs modulation may become more important.


Asunto(s)
Potenciales de Acción/fisiología , Nodo Atrioventricular/metabolismo , Ventrículos Cardíacos/metabolismo , Canales de Potasio/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas Adrenérgicos beta/farmacología , Animales , Nodo Atrioventricular/efectos de los fármacos , Femenino , Cobayas , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Isoproterenol/farmacología , Miocardio/metabolismo , Técnicas de Placa-Clamp/métodos
6.
J Pharmacol Sci ; 143(1): 56-59, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32144028

RESUMEN

Since microminipig is becoming attractive model for various cardiac electropharmacological applications, which may meet consideration of 3Rs. We characterized microminipigs by analyzing how multi-ionic channel inhibitor bepridil may affect their in situ hearts in comparison with dogs. Bepridil in doses of 0.3 and 3.0 mg/kg were intravenously administered over 10 min under halothane anesthesia (n = 4). Microminipigs may be less sensitive for ICaT inhibition of bepridil, whereas they are more responsive to INa, IKr and IKs suppression than dogs. This information would help predict cardiovascular effects of a drug in patients with the remodeled hearts having similar electrophysiological profile to microminipigs.


Asunto(s)
Animales de Laboratorio , Bepridil/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Sistema Cardiovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Porcinos Enanos , Animales , Bepridil/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Perros , Relación Dosis-Respuesta a Droga , Infusiones Intravenosas , Porcinos
7.
Exp Cell Res ; 363(2): 315-320, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29395134

RESUMEN

BACKGROUND: Recent studies have revealed that mutation in KCNE1, ß-subunits of cardiac potassium channel, involved in ventricular fibrillation. Whereas its role in early repolarization syndrome (ERS) is less well understood. OBJECTIVE: To study whether mutant in KCNE1 is associated with ERS and explore the possible underlying molecular mechanisms. METHODS: Whole genome from four unrelated families with ERS was amplified and sequenced. Wild-type (WT) KCNE1 and/or KCNE1-S38G (S38G) were expressed in HEK293 cells with KCNQ1. Functional studies included whole-cell patch-clamp, western blot and immunofluorescence were performed to reveal the possible underlying mechanisms. RESULTS: The co-expression of KCNE1-S38G and KCNQ1 decreased tail current density of IKs but had little effect in modulation channel kinetics of IKs. Compared with KCNE1-WT, the expression and membrane location of KCNE1-S38G decreased. Co-expression of KCNE1-WT and KCNE1-S38G partially rescued the function of IKs channel. CONCLUSIONS: The S38G mutation induced a loss-of-function of IKs due to decreasing of KCNE1 protein expression and defecting in KCNE1 protein membrane trafficking. Our findings suggested that KCNE1 may be one of the possible modulatory genes associated to ERS.


Asunto(s)
Mutación/genética , Canales de Potasio con Entrada de Voltaje/genética , Adulto , Anciano , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Moduladores del Transporte de Membrana/metabolismo , Persona de Mediana Edad , Linaje , Potasio/metabolismo
8.
J Mol Cell Cardiol ; 116: 165-174, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29452158

RESUMEN

The slowly activating delayed rectifier K+ current (IKs) is one of the main repolarizing currents in the human heart. Evidence has shown that angiotensin II (Ang II) regulates IKs through the protein kinase C (PKC) pathway, but the related results are controversial. This study was designed to identify PKC isoenzymes involved in the regulation of IKs by Ang II and the underlying molecular mechanism. The whole-cell patch-clamp technique was used to record IKs in isolated guinea pig ventricular cardiomyocytes and in human embryonic kidney (HEK) 293 cells co-transfected with human KCNQ1/KCNE1 genes and Ang II type 1 receptor genes. Ang II inhibited IKs in a concentration-dependent manner in native cardiomyocytes. A broad PKC inhibitor Gö6983 (not inhibiting PKCε) and a selective cPKC inhibitor Gö6976 did not affect the inhibitory action of Ang II. In contrast, the inhibition was significantly attenuated by PKCε-selective peptide inhibitor εV1-2. However, direct activation of PKC by phorbol 12-myristate 13-acetate (PMA) increased the cloned human IKs in HEK293 cells. Similarly, the cPKC peptide activator significantly enhanced the current. In contrast, the PKCε peptide activator inhibited the current. Further evidence showed that PKCε knockdown by siRNA antagonized the Ang II-induced inhibition on KCNQ1/KCNE1 current, whereas knockdown of cPKCs (PKCα and PKCß) attenuated the potentiation of the current by PMA. Moreover, deletion of four putative phosphorylation sites in the C-terminus of KCNQ1 abolished the action of PMA. Mutation of two putative phosphorylation sites in the N-terminus of KCNQ1 and one site in KCNE1 (S102) blocked the inhibition of Ang II. Our results demonstrate that PKCε isoenzyme mediates the inhibitory action of Ang II on IKs and by phosphorylating distinct sites in KCNQ1/KCNE1, cPKC and PKCε isoenzymes produce the contrary regulatory effects on the channel. These findings have provided new insight into the molecular mechanism underlying the modulation of the KCNQ1/KCNE1 channel.


Asunto(s)
Angiotensina II/farmacología , Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Cobayas , Células HEK293 , Ventrículos Cardíacos/metabolismo , Humanos , Isoenzimas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología
9.
J Mol Cell Cardiol ; 99: 1-13, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27590098

RESUMEN

Patients with HIV present with a higher prevalence of QT prolongation, of which molecular bases are still not clear. Among HIV proteins, Tat serves as a transactivator that stimulates viral genes expression and is required for efficient HIV replication. Tat is actively secreted into the blood by infected T-cells and affects organs such as the heart. Tat has been shown to alter cardiac repolarization in animal models but how this is mediated and whether this is also the case in human cells is unknown. In the present study, we show that Tat transfection in heterologous expression systems led to a decrease in hERG (underlying cardiac IKr) and human KCNE1-KCNQ1 (underlying cardiac IKs) currents and to an acceleration of their deactivation. This is consistent with a decrease in available phosphatidylinositol-(4,5)-bisphosphate (PIP2). A mutant Tat, unable to bind PIP2, did not reproduce the observed effects. In addition, WT-Tat had no effect on a mutant KCNQ1 which is PIP2-insensitive, further confirming the hypothesis. Twenty-four-hour incubation of human induced pluripotent stem cells-derived cardiomyocytes with Wild-type Tat reduced IKr and accelerated its deactivation. Concordantly, this Tat incubation led to a prolongation of the action potential (AP) duration. Events of AP alternans were also recorded in the presence of Tat, and were exacerbated at a low pacing cycle length. Altogether, these data obtained on human K+ channels both in heterologous expression systems and in human cardiomyocytes suggest that Tat sequesters PIP2, leading to a reduction of IKr and IKs, and provide a molecular mechanism for QT prolongation in HIV-infected patients.


Asunto(s)
Potenciales de Acción , Fosfatidilinositol 4,5-Difosfato/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Células COS , Diferenciación Celular , Línea Celular , Canal de Potasio ERG1/metabolismo , Fenómenos Electrofisiológicos , Expresión Génica , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
10.
J Mol Cell Cardiol ; 89(Pt B): 335-48, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26454161

RESUMEN

KCNQ1 (Kv7.1) proteins form a homotetrameric channel, which produces a voltage-dependent K(+) current. Co-assembly of KCNQ1 with the auxiliary ß-subunit KCNE1 strongly up-regulates this current. In cardiac myocytes, KCNQ1/E1 complexes are thought to give rise to the delayed rectifier current IKs, which contributes to cardiac action potential repolarization. We report here that the type I membrane protein BACE1 (ß-site APP-cleaving enzyme 1), which is best known for its detrimental role in Alzheimer's disease, but is also, as reported here, present in cardiac myocytes, serves as a novel interaction partner of KCNQ1. Using HEK293T cells as heterologous expression system to study the electrophysiological effects of BACE1 and KCNE1 on KCNQ1 in different combinations, our main findings were the following: (1) BACE1 slowed the inactivation of KCNQ1 current producing an increased initial response to depolarizing voltage steps. (2) Activation kinetics of KCNQ1/E1 currents were significantly slowed in the presence of co-expressed BACE1. (3) BACE1 impaired reconstituted cardiac IKs when cardiac action potentials were used as voltage commands, but interestingly augmented the IKs of ATP-deprived cells, suggesting that the effect of BACE1 depends on the metabolic state of the cell. (4) The electrophysiological effects of BACE1 on KCNQ1 reported here were independent of its enzymatic activity, as they were preserved when the proteolytically inactive variant BACE1 D289N was co-transfected in lieu of BACE1 or when BACE1-expressing cells were treated with the BACE1-inhibiting compound C3. (5) Co-immunoprecipitation and fluorescence recovery after photobleaching (FRAP) supported our hypothesis that BACE1 modifies the biophysical properties of IKs by physically interacting with KCNQ1 in a ß-subunit-like fashion. Strongly underscoring the functional significance of this interaction, we detected BACE1 in human iPSC-derived cardiomyocytes and murine cardiac tissue and observed decreased IKs in atrial cardiomyocytes of BACE1-deficient mice.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/deficiencia , Ácido Aspártico Endopeptidasas/deficiencia , Activación del Canal Iónico , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Potenciales de Acción , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Cinética , Masculino , Ratones , Complejos Multiproteicos/metabolismo , Fenotipo , Unión Proteica , Proteolisis
11.
J Mol Cell Cardiol ; 72: 273-80, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24713462

RESUMEN

KCNQ1 encodes the α subunit of the voltage-gated channel that mediates the cardiac slow delayed rectifier K(+) current (IKs). Here, we report a KCNQ1 allele encoding an A590T mutation [KCNQ1(A590T)] found in a 39-year-old female with a mild QT prolongation. A590 is located in the C-terminal α helical region of KCNQ1 that mediates subunit tetramerization, membrane trafficking, and interaction with Yotiao. This interaction is known to be required for the proper modulation of IKs by cAMP. Since previous studies reported that mutations in the vicinity of A590 impair IKs channel surface expression and function, we examined whether and how the A590T mutation affects the IKs channel. Electrophysiological measurements in HEK-293T cells showed that the A590T mutation caused a reduction in IKs density and a right-shift of the current-voltage relation of channel activation. Immunocytochemical and immunoblot analyses showed the reduced cell surface expression of KCNQ1(A590T) subunit and its rescue by coexpression of the wild-type KCNQ1 [KCNQ1(WT)] subunit. Moreover, KCNQ1(A590T) subunit interacted with Yotiao and had a cAMP-responsiveness comparable to that of KCNQ1(WT) subunit. These findings indicate that the A590 of KCNQ1 subunit plays important roles in the maintenance of channel surface expression and function via a novel mechanism independent of interaction with Yotiao.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas del Citoesqueleto/metabolismo , Canal de Potasio KCNQ1/metabolismo , Síndrome de QT Prolongado/metabolismo , Mutación , Canales de Potasio con Entrada de Voltaje/metabolismo , Subunidades de Proteína/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Potenciales de Acción , Adulto , Secuencia de Aminoácidos , AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/genética , Femenino , Expresión Génica , Células HEK293 , Humanos , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Datos de Secuencia Molecular , Canales de Potasio con Entrada de Voltaje/genética , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Transporte de Proteínas , Alineación de Secuencia
12.
J Mol Cell Cardiol ; 67: 12-25, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24370890

RESUMEN

The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.


Asunto(s)
Diferenciación Celular , Fenómenos Electrofisiológicos , Miocitos Cardíacos/citología , Técnicas de Cultivo , Epigenómica , Corazón/embriología , Corazón/crecimiento & desarrollo , Humanos , Células Madre Pluripotentes/citología , Transducción de Señal
13.
J Electrocardiol ; 47(2): 158-65, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24411289

RESUMEN

Mutations in transmembrane domains of the KCNQ1 subunit of the I(Ks) potassium channel have been associated with familial atrial fibrillation. We have investigated mechanisms by which the S1 domain S140G KCNQ1 mutation influences atrial arrhythmia risk and, additionally, whether it can affect ventricular electrophysiology. In perforated-patch recordings, S140G-KCNQ1+KCNE1 exhibited leftward-shifted activation, slowed deactivation and marked residual current. In human atrial action potential (AP) simulations, AP duration and refractoriness were shortened and rate-dependence flattened. Simulated I(Ks) but not I(Kr) block offset AP shortening produced by the mutation. In atrial tissue simulations, temporal vulnerability to re-entry was little affected by the S140G mutation. Spatial vulnerability was markedly increased, leading to more stable and stationary spiral wave re-entry in 2D stimulations, which was offset by I(Ks) block, and to scroll waves in 3D simulations. These changes account for vulnerability to AF with this mutation. Ventricular AP clamp experiments indicate a propensity for increased ventricular I(Ks) with the S140G KCNQ1 mutation and ventricular AP simulations showed model-dependent ventricular AP abbreviation.


Asunto(s)
Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Canal de Potasio KCNQ1/genética , Mutación , Potenciales de Acción/fisiología , Animales , Células CHO , Simulación por Computador , Cricetulus , Humanos , Técnicas de Placa-Clamp , Disfunción Ventricular/genética , Disfunción Ventricular/fisiopatología
14.
J Mol Cell Cardiol ; 64: 90-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24051368

RESUMEN

Protein phosphorylation is a major control mechanism of a wide range of physiological processes and plays an important role in cardiac pathophysiology. Serine/threonine protein phosphatases control the dephosphorylation of a variety of cardiac proteins, thereby fine-tuning cardiac electrophysiology and function. Specificity of protein phosphatases type-1 and type-2A is achieved by multiprotein complexes that target the catalytic subunits to specific subcellular domains. Here, we describe the composition, regulation and target substrates of serine/threonine phosphatases in the heart. In addition, we provide an overview of pharmacological tools and genetic models to study the role of cardiac phosphatases. Finally, we review the role of protein phosphatases in the diseased heart, particularly in ventricular arrhythmias and atrial fibrillation and discuss their role as potential therapeutic targets.


Asunto(s)
Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Corazón/fisiología , Fosfoproteínas Fosfatasas/metabolismo , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Activación Enzimática , Regulación de la Expresión Génica , Cardiopatías/tratamiento farmacológico , Cardiopatías/genética , Humanos , Contracción Miocárdica/fisiología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosforilación
15.
Eur J Pharmacol ; 932: 175218, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36007604

RESUMEN

INTRODUCTION: Impaired IKs induced by drugs or due to a KCNQ1 mutation, diagnosed as long QT syndrome type 1 (LQT1) prolongs the QT interval and predisposes the heart to Torsade de Pointes (TdP) arrhythmias. The anesthetized chronic AV block (CAVB) dog is inducible for TdP after remodeling and IKr inhibitor dofetilide. We tested the proarrhythmic effect of IKs inhibition in the CAVB dog, and the proarrhythmic role of increased contractility herein. METHODS: Dofetilide-inducible animals were included to test the proarrhythmic effect of 1) IKs inhibition by JNJ303 (0.63 mg/kg/10min i.v.; n = 4), 2) IKs inhibition combined with enhanced inotropy (ouabain, 0.045 mg/kg/1min i.v.; n = 6), and 3) the washout period of the anesthetic regime (n = 10). RESULTS: JNJ303 prolonged the QTc interval (from 477 ± 53 ms to 565 ± 14 ms, P < 0.02) resembling standardized dofetilide-induced QTc prolongation. Single ectopic beats (n = 4) and ventricular tachycardia (VT) (n = 3) were present, increasing the arrhythmia score (AS) from 1.0 ± 0 to 7.1 ± 6.5. JNJ303 combined with ouabain increased contractile parameters (LVdP/dtmax from 1725 ± 273 to 4147 ± 611 mmHg/s, P < 0.01). Moreover, TdP arrhythmias were induced in 4/6 dogs and AS increased from 1.0 ± 0 to 20.2 ± 19.0 after JNJ303 and ouabain (P < 0.05). Finally, TdP arrhythmias were induced in 4/10 dogs during the anesthesia washout period and the AS increased from 1.1 ± 0.3 to 9.2 ± 11.2. CONCLUSION: Mimicking LQT1 using IKs inhibitor JNJ303 prolongs the QTc interval and triggers ectopic beats and non-sustained VT in the CAVB dog. Induction of the more severe arrhythmic events (TdP) demands a combination of IKs inhibition with enhanced inotropy or ending the anesthetic regime.


Asunto(s)
Bloqueo Atrioventricular , Torsades de Pointes , Animales , Antiarrítmicos/farmacología , Arritmias Cardíacas/inducido químicamente , Proteínas de Unión al ADN , Perros , Canal de Potasio KCNQ1 , Ouabaína , Fenetilaminas , Sulfonamidas , Torsades de Pointes/inducido químicamente
16.
Cell Signal ; 91: 110228, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34958868

RESUMEN

Activation of a specific protein kinase C (PKC) isoform during stimulation of Gq protein-coupled receptors (GqPCRs) is determined by homologous receptor desensitization that controls the spatiotemporal formation of downstream Gq signalling molecules. Furthermore, GqPCR-activated PKC isoforms specifically regulate receptor activity via a negative feedback mechanism. In the present study, we investigated the contribution of several phosphorylation sites in the α1B-adrenergic receptor (α1B-AR) for PKC and G protein coupled receptor kinase 2 (GRK2) to homologous receptor desensitization and effector modulation. We analyzed signalling events downstream to human wildtype α1B-ARs and α1B-ARs lacking PKC or GRK2 phosphorylation sites (Δ391-401, α1B-ΔPKC-AR and Δ402-520, α1B-ΔGRK-AR) by means of FRET-based biosensors in HEK293 that served as online-assays of receptor activity. K+ currents through KCNQ1/KCNE1 channels (IKs), which are regulated by both phosphatidylinositol 4,5-bisphosphate (PIP2)-depletion and/or phosphorylation by PKC, were measured as a functional readout of wildtype and mutant α1B-AR receptor activity. As a novel finding, we provide evidence that deletion of PKC and GRK2 phosphorylation sites in α1B-ARs abrogates the contribution of PKCα to homologous receptor desensitization. Instead, the time course of mutant receptor activity was specifically modulated by PKCß. Mutant α1B-ARs displayed pronounced homologous receptor desensitization that was abolished by PKCß-specific pharmacological inhibitors. IKs modulation during stimulation of wildtype and mutant α1B-ARs displayed transient inhibition and current facilitation after agonist withdrawal with reduced capability of mutant α1B-ARs to induce IKs inhibition. Pharmacological inhibition of the PKCß isoform did not augment IKs reduction by mutant α1B-ARs, but shifted IKs modulation towards current facilitation. Coexpression of an inactive (dominant-negative) PKCδ isoform (DN-PKCδ) abolished IKs facilitation in α1B-ΔGRK-AR-expressing cells, but not in α1B-ΔPKC-AR-expressing cells. The data indicate that the differential modulation of IKs activity by α1B-ΔGRK- and α1B-ΔPKC-receptors is attributed to the activation of entirely distinct novel PKC isoforms. To summarize, specific phosphorylation sites within the wildtype and mutant α1B-adrenergic receptors are targeted by different PKC isoforms, resulting in differential regulation of receptor desensitization and effector function.


Asunto(s)
Canal de Potasio KCNQ1 , Canales de Potasio con Entrada de Voltaje , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-alfa/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Células HEK293 , Humanos , Canal de Potasio KCNQ1/metabolismo , Fosforilación , Canales de Potasio con Entrada de Voltaje/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal
17.
Eur J Pharmacol ; 906: 174278, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34174270

RESUMEN

KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.


Asunto(s)
Analgésicos/farmacología , Antiarrítmicos/farmacología , Anticonvulsivantes/farmacología , Canales de Potasio KCNQ/antagonistas & inhibidores , Analgésicos/uso terapéutico , Animales , Antiarrítmicos/uso terapéutico , Anticonvulsivantes/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Epilepsia/tratamiento farmacológico , Corazón/efectos de los fármacos , Humanos , Canales de Potasio KCNQ/metabolismo , Ligandos , Miocardio/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/tratamiento farmacológico
18.
Br J Pharmacol ; 178(13): 2690-2708, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33763865

RESUMEN

BACKGROUND AND PURPOSE: The slowly activating delayed rectifier K+ channel (IKs ), composed of pore-forming KCNQ1 α-subunits and ancillary KCNE1 ß-subunits, regulates ventricular repolarization in human heart. Propofol, at clinically used concentrations, modestly inhibits the intact (wild-type) IKs channels and is therefore unlikely to appreciably prolong QT interval in ECG during anaesthesia. However, little information is available concerning the inhibitory effect of propofol on IKs channel associated with its gene variants implicated in QT prolongation. The KCNE1 single nucleotide polymorphism leading to D85N is associated with drug-induced QT prolongation and therefore regarded as a clinically important genetic variant. This study examined whether KCNE1-D85N affects the sensitivity of IKs to inhibition by propofol. EXPERIMENTAL APPROACH: Whole-cell patch-clamp and immunostaining experiments were conducted in HEK293 cells and/or mouse cardiomyocyte-derived HL-1 cells, transfected with wild-type KCNQ1, wild-type or variant KCNE1 cDNAs. KEY RESULTS: Propofol inhibited KCNQ1/KCNE1-D85N current more potently than KCNQ1/KCNE1 current in HEK293 cells and HL-1 cells. Immunostaining experiments in HEK293 cells revealed that pretreatment with propofol (10 µM) did not appreciably affect cell membrane expression of KCNQ1 and KCNE1 proteins in KCNQ1/KCNE1 and KCNQ1/KCNE1-D85N channels. CONCLUSION AND IMPLICATIONS: The KCNE1 polymorphism D85N significantly elevates the sensitivity of IKs to inhibition by propofol. This study detects a functionally important role of KCNE1-D85N polymorphism in conferring genetic susceptibility to propofol-induced QT prolongation and further suggests the possibility that the inhibitory action of anaesthetics on ionic currents becomes exaggerated in patients carrying variants in genes encoding ion channels.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Propofol , Animales , Células HEK293 , Humanos , Canal de Potasio KCNQ1/genética , Ratones , Canales de Potasio con Entrada de Voltaje/genética , Propofol/farmacología
19.
Channels (Austin) ; 15(1): 262-272, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33535882

RESUMEN

The slowly activating delayed rectifier K+ current (IKs) plays a key role in the repolarization of ventricular action potential in the human heart and is formed by the pore-forming α-subunit encoded by KCNQ1 (Kv7.1) and ß-subunit encoded by KCNE1. Evidence suggested that IKs was regulated through protein kinase C (PKC) pathway, but the mechanism is controversial. This study was designed to identify the specific PKC isoform involved in the long-term regulation of IKs current. The IKs current was recorded using whole-cell patch-clamp technique in human embryonic kidney (HEK) 293B cell co-transfected with human KCNQ1/KCNE1 genes. The results revealed that both chronic activation of Ang II and PMA reduced the IKs current in a long-term regulation (about 24 hours). Further evidence showed that PKCε knockdown by siRNA antagonized the AngII-induced chronic inhibition on the IKs current, whereas knockdown of cPKC (PKCα and PKCß) attenuated the inhibition effect of PMA on the current. Moreover, the forward transport inhibition of the channel with brefeldin A alleviated the Ang II-induced chronic inhibition on IKs current, while the channel endocytosis inhibition with dynasore alleviated both Ang II and PMA-induced chronic inhibition on IKs current. The above results showed that PKCε activation promoted the channel endocytosis and inhibited the channel forward transport to the plasma membrane, while cPKC activation only promoted the channel endocytosis, which both down regulated the channel current.


Asunto(s)
Canal de Potasio KCNQ1 , Células HEK293 , Humanos , Técnicas de Placa-Clamp , Potasio , Canales de Potasio con Entrada de Voltaje
20.
Eur J Pharmacol ; 883: 173378, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32710951

RESUMEN

The slowly and rapidly activating delayed rectifier K+ channels (IKs and IKr, respectively) contribute to the repolarization of ventricular action potential in human heart and thereby determine QT interval on an electrocardiogram. Loss-of-function mutations in genes encoding IKs and IKr cause type 1 and type 2 long QT syndrome (LQT1 and LQT2, respectively), accompanied by a high risk of malignant ventricular arrhythmias and sudden cardiac death. This study was designed to investigate which cardiac electrophysiological conditions exaggerate QT-prolonging and arrhythmogenic effects of sevoflurane. We used the O'Hara-Rudy dynamic model to reconstruct human ventricular action potential and a pseudo-electrocardiogram, and simulated LQT1 and LQT2 phenotypes by decreasing conductances of IKs and IKr, respectively. Sevoflurane, but not propofol, prolonged ventricular action potential duration and QT interval in wild-type, LQT1 and LQT2 models. The QT-prolonging effect of sevoflurane was more profound in LQT2 than in wild-type and LQT1 models. The potent inhibitory effect of sevoflurane on IKs was primarily responsible for its QT-prolonging effect. In LQT2 model, IKs was considerably enhanced during excessive prolongation of ventricular action potential duration by reduction of IKr and relative contribution of IKs to ventricular repolarization was markedly elevated, which appears to underlie more pronounced QT-prolonging effect of sevoflurane in LQT2 model, compared with wild-type and LQT1 models. This simulation study clearly elucidates the electrophysiological basis underlying the difference in QT-prolonging effect of sevoflurane among wild-type, LQT1 and LQT2 models, and may provide important information for developing anesthetic strategies for patients with long QT syndrome in clinical settings.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Síndrome de QT Prolongado/inducido químicamente , Modelos Cardiovasculares , Miocitos Cardíacos/efectos de los fármacos , Síndrome de Romano-Ward/inducido químicamente , Sevoflurano/toxicidad , Estudios de Casos y Controles , Simulación por Computador , Canales de Potasio de Tipo Rectificador Tardío/genética , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Miocitos Cardíacos/metabolismo , Propofol/toxicidad , Medición de Riesgo , Factores de Riesgo , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/metabolismo , Síndrome de Romano-Ward/fisiopatología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA