Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Mol Cell Cardiol ; 130: 107-121, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30935998

RESUMEN

Ca2+-sensing receptors (CaSRs) belong to the class C of G protein-coupled receptors and are activated by extracellular Ca2+. CaSRs display biased G protein signaling by coupling to different classes of heterotrimeric G proteins depending on agonist and cell type. In this study we used fluorescent biosensors to directly analyze G protein coupling to CaSRs and downstream signaling in living cells. In HEK 293 cells, CaSRs displayed biased signaling: elevation of extracellular Ca2+ or application of the alternative agonist spermine caused activation of Gi- and Gq-proteins. Adult cardiac myocytes express endogenous CaSRs, which have been implicated in regulating Ca2+ signaling and contractility. Biased signaling of CaSRs has not been investigated in these cells. To evaluate efficiencies of Gi- and Gq-signaling via CaSRs in rat atrial myocytes, we measured G protein-activated K+ (GIRK) channels. Activation of GIRK requires binding of Gßγ subunits released from Gi proteins, whereas Gq-signaling results in inhibition of GIRK channel activity. Stimulation of CaSRs by Ca2+ or spermine failed to directly activate Gi and GIRK channels. When GIRK channels were pre-activated via endogenous M2 receptors, stimulation of CaSRs caused pronounced inhibition of GIRK currents. This effect was specific to CaSR activation: GIRK current inhibition was sensitive to NPS-2143, a negative allosteric modulator of CaSRs, and abrogated by FR900359, a direct inhibitor of Gq. GIRK current inhibition was also sensitive to the PKC inhibitor chelerythrine, suggesting that following activation of CaSR and Gq, GIRK currents are modulated by PKC phosphorylation. We conclude from this data that cardiac CaSRs do not activate Gi and affect GIRK currents preferentially via the Gq/PKC pathway.


Asunto(s)
Señalización del Calcio , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Humanos , Masculino , Miocitos Cardíacos/patología , Naftalenos/farmacología , Proteína Quinasa C/metabolismo , Ratas , Ratas Endogámicas WKY
2.
J Biol Chem ; 291(51): 26410-26426, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27834678

RESUMEN

Activation of Gq protein-coupled receptors (GqPCRs) might induce divergent cellular responses, related to receptor-specific activation of different branches of the Gq signaling pathway. Receptor-specific desensitization provides a mechanism of effector modulation by restricting the spatiotemporal activation of signaling components downstream of Gq We quantified signaling events downstream of GqPCR activation with FRET-based biosensors in CHO and HEK 293 cells. KCNQ1/KCNE1 channels (IKs) were measured as a functional readout of receptor-specific activation. Activation of muscarinic M1 receptors (M1-Rs) caused robust and reversible inhibition of IKs. In contrast, activation of α1B-adrenergic receptors (α1B-ARs) induced transient inhibition of IKs, which turned into delayed facilitation after agonist withdrawal. As a novel finding, we demonstrate that GqPCR-specific kinetics of IKs modulation are determined by receptor-specific desensitization, evident at the level of Gαq activation, phosphatidylinositol 4,5-bisphosphate (PIP2) depletion, and diacylglycerol production. Sustained IKs inhibition during M1-R stimulation is attributed to robust membrane PIP2 depletion, whereas the rapid desensitization of α1B-AR delimits PIP2 reduction and augments current activation by protein kinase C (PKC). Overexpression of Ca2+-independent PKCδ did not affect the time course of α1B-AR-induced diacylglycerol formation, excluding a contribution of PKCδ to α1B-AR desensitization. Pharmacological inhibition of Ca2+-dependent PKC isoforms abolished fast α1B receptor desensitization and augmented IKs reduction, but did not affect IKs facilitation. These data indicate a contribution of Ca2+-dependent PKCs to α1B-AR desensitization, whereas IKs facilitation is induced by Ca2+-independent PKC isoforms. In contrast, neither inhibition of Ca2+-dependent/Ca2+-independent isoforms nor overexpression of PKCδ induced M1 receptor desensitization, excluding a contribution of PKC to M1-R-induced IKs modulation.


Asunto(s)
Señalización del Calcio/fisiología , Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Células CHO , Calcio/metabolismo , Cricetinae , Cricetulus , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Canal de Potasio KCNQ1/genética , Fosfatidilinositol 4,5-Difosfato/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Proteína Quinasa C-delta/metabolismo , Receptor Muscarínico M1/genética , Receptores Adrenérgicos alfa 1/genética
3.
J Biol Chem ; 289(33): 22749-22758, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24947509

RESUMEN

Kv7.1 to Kv7.5 α-subunits belong to the family of voltage-gated potassium channels (Kv). Assembled with the ß-subunit KCNE1, Kv7.1 conducts the slowly activating potassium current IKs, which is one of the major currents underlying repolarization of the cardiac action potential. A known regulator of Kv7 channels is the lipid phosphatidylinositol 4,5-bisphosphate (PIP2). PIP2 increases the macroscopic current amplitude by stabilizing the open conformation of 7.1/KCNE1 channels. However, knowledge about the exact nature of the interaction is incomplete. The aim of this study was the identification of the amino acids responsible for the interaction between Kv7.1 and PIP2. We generated 13 charge neutralizing point mutations at the intracellular membrane border and characterized them electrophysiologically in complex with KCNE1 under the influence of diC8-PIP2. Electrophysiological analysis of corresponding long QT syndrome mutants suggested impaired PIP2 regulation as the cause for channel dysfunction. To clarify the underlying structural mechanism of PIP2 binding, molecular dynamics simulations of Kv7.1/KCNE1 complexes containing two PIP2 molecules in each subunit at specific sites were performed. Here, we identified a subset of nine residues participating in the interaction of PIP2 and Kv7.1/KCNE1. These residues may form at least two binding pockets per subunit, leading to the stabilization of channel conformations upon PIP2 binding.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Potenciales de Acción/fisiología , Sustitución de Aminoácidos , Animales , Sitios de Unión , Humanos , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/genética , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/genética , Mutación Puntual , Unión Proteica , Xenopus laevis
4.
Pflugers Arch ; 466(10): 1885-97, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24389605

RESUMEN

Ionotropic glutamate receptors are the most important excitatory receptors in the central nervous system, and their impairment can lead to multiple neuronal diseases. Here, we show that glutamate-induced currents in oocytes expressing GluA1 are increased by coexpression of the schizophrenia-associated phosphoinositide kinase PIP5K2A. This effect was due to enhanced membrane abundance and was blunted by a point mutation (N251S) in PIP5K2A. An increase in GluA1 currents was also observed upon acute injection of PI(4,5)P2, the main product of PIP5K2A. By expression of wild-type and mutant PIP5K2A in human embryonic kidney cells, we were able to provide evidence of impaired kinase activity of the mutant PIP5K2A. We defined the region K813-K823 of GluA1 as critical for the PI(4,5)P2 effect by performing an alanine scan that suggested PI(4,5)P2 binding to this area. A PIP strip assay revealed PI(4,5)P2 binding to the C-terminal GluA1 peptide. The present observations disclose a novel mechanism in the regulation of GluA1.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Receptores AMPA/química , Alanina/química , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células HEK293 , Humanos , Datos de Secuencia Molecular , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Receptores AMPA/genética , Receptores AMPA/metabolismo , Xenopus
5.
FASEB J ; 27(10): 4108-21, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23825229

RESUMEN

Infections with coxsackieviruses of type B (CVBs), which are known to induce severe forms of acute and chronic myocarditis, are often accompanied by ventricular arrhythmias and sudden cardiac death. The mechanisms underlying the development of virus-induced, life-threatening arrhythmias, which are phenotypically similar to those observed in patients having functionally impaired cardiac ion channels, remain, however, enigmatic. In the present study, we show, for the first time, modulating time-dependent effects of CVB3 on the cardiac ion channels KCNQ1, hERG1, and Cav1.2 in heterologous expression. Channel protein abundance in cellular plasma membrane and patterns of their subcellular distribution were altered in infected murine hearts. The antiviral compound AG7088 did not prevent these effects on channels. In silico analyses of infected human myocytes suggest pronounced alterations of electrical and calcium signaling and increased risk of arrhythmogenesis. These modifications are attenuated by the common Asian polymorphism KCNQ1 P448R, a genetic determinant preventing coxsackievirus-induced effects in vitro. This study provides a previously unknown explanation for the development of arrhythmias in enteroviral myocarditis, which will help to develop therapeutic strategies for arrhythmia treatment.


Asunto(s)
Enterovirus Humano B/clasificación , Enterovirus Humano B/fisiología , Regulación de la Expresión Génica/fisiología , Canales Iónicos/metabolismo , Miocitos Cardíacos/metabolismo , Transporte de Proteínas/fisiología , Animales , Simulación por Computador , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Miocitos Cardíacos/virología , Oocitos , Polimorfismo Genético , Xenopus
6.
J Mol Cell Cardiol ; 61: 142-52, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23531443

RESUMEN

Shortened action-potential duration (APD) and blunted APD rate adaptation are hallmarks of chronic atrial fibrillation (cAF). Basal and muscarinic (M)-receptor-activated inward-rectifier K(+) currents (IK1 and IK,ACh, respectively) contribute to regulation of human atrial APD and are subject to cAF-dependent remodeling. Intracellular Na(+) ([Na(+)]i) enhances IK,ACh in experimental models but the effect of [Na(+)]i-dependent regulation of inward-rectifier K(+) currents on APD in human atrial myocytes is currently unknown. Here, we report a [Na(+)]i-dependent inhibition of outward IK1 in atrial myocytes from sinus rhythm (SR) or cAF patients. In contrast, IK,ACh activated by carbachol, a non-selective M-receptor agonist, increased with elevation of [Na(+)]i in SR. This [Na(+)]i-dependent IK,ACh regulation was absent in cAF. Including [Na(+)]i dependence of IK1 and IK,ACh in a recent computational model of the human atrial myocyte revealed that [Na(+)]i accumulation at fast rates inhibits IK1 and blunts physiological APD rate dependence in both groups. [Na(+)]i-dependent IK,ACh augmentation at fast rates increased APD rate dependence in SR, but not in cAF. These results identify impaired Na(+)-sensitivity of IK,ACh as one potential mechanism contributing to the blunted APD rate dependence in patients with cAF. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".


Asunto(s)
Acetilcolina/farmacología , Potenciales de Acción , Fibrilación Atrial/fisiopatología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Agonistas Muscarínicos/farmacología , Anciano , Arritmia Sinusal/metabolismo , Arritmia Sinusal/fisiopatología , Fibrilación Atrial/metabolismo , Carbacol/farmacología , Femenino , Humanos , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Subunidades de Proteína/metabolismo , Sodio/metabolismo
7.
FASEB J ; 26(2): 513-22, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22002906

RESUMEN

Inward rectifier potassium channels of the Kir2 subfamily are important determinants of the electrical activity of brain and muscle cells. Genetic mutations in Kir2.1 associate with Andersen-Tawil syndrome (ATS), a familial disorder leading to stress-triggered periodic paralysis and ventricular arrhythmia. To identify the molecular mechanisms of this stress trigger, we analyze Kir channel function and localization electrophysiologically and by time-resolved confocal microscopy. Furthermore, we employ a mathematical model of muscular membrane potential. We identify a novel corticoid signaling pathway that, when activated by glucocorticoids, leads to enrichment of Kir2 channels in the plasma membranes of mammalian cell lines and isolated cardiac and skeletal muscle cells. We further demonstrate that activation of this pathway can either partly restore (40% of cases) or further impair (20% of cases) the function of mutant ATS channels, depending on the particular Kir2.1 mutation. This means that glucocorticoid treatment might either alleviate or deteriorate symptoms of ATS depending on the patient's individual Kir2.1 genotype. Thus, our findings provide a possible explanation for the contradictory effects of glucocorticoid treatment on symptoms in patients with ATS and may open new pathways for the design of personalized medicines in ATS therapy.


Asunto(s)
Síndrome de Andersen/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Síndrome de Andersen/tratamiento farmacológico , Síndrome de Andersen/genética , Animales , Femenino , Glucocorticoides/uso terapéutico , Cobayas , Células HEK293 , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Técnicas In Vitro , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miocitos Cardíacos/metabolismo , Oocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Estrés Fisiológico , Xenopus laevis
8.
J Biol Chem ; 286(1): 290-8, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21041301

RESUMEN

A large conductance (∼300 picosiemens) channel (LCC) of unknown molecular identity, activated by Ca(2+) release from the sarcoplasmic reticulum, particularly when augmented by caffeine, has been described previously in isolated cardiac myocytes. A potential candidate for this channel is pannexin 1 (Panx1), which has been shown to form large ion channels when expressed in Xenopus oocytes and mammalian cells. Panx1 function is implicated in ATP-mediated auto-/paracrine signaling, and a crucial role in several cell death pathways has been suggested. Here, we demonstrate that after culturing for 4 days LCC activity is no longer detected in myocytes but can be rescued by adenoviral gene transfer of Panx1. Endogenous LCCs and those related to expression of Panx1 share key pharmacological properties previously used for identifying and characterizing Panx1 channels. These data demonstrate that Panx1 constitutes the LCC of cardiac myocytes. Sporadic openings of single Panx1 channels in the absence of Ca(2+) release can trigger action potentials, suggesting that Panx1 channels potentially promote arrhythmogenic activities.


Asunto(s)
Conexinas/metabolismo , Canales Iónicos/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Potenciales de Acción , Adenosina Trifosfato/metabolismo , Adenoviridae/genética , Animales , Fenómenos Biomecánicos , Conexinas/genética , Femenino , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Masculino , Miocitos Cardíacos/citología , Proteínas del Tejido Nervioso/genética , Ratas , Ratas Wistar , Retículo Sarcoplasmático/metabolismo , Factores de Tiempo
9.
Pflugers Arch ; 461(1): 165-76, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21061016

RESUMEN

G protein-activated K(+) channels composed of Kir3 (GIRK) subunits contribute to regulation of heart rate and excitability. Opening of these channels in myocytes is increased by binding of G(ßγ) upon activation of muscarinic M(2) receptors (M(2)-R) or A(1) adenosine receptors (A(1)-R). It has been shown that saturating activation of A(1)-R resulted in a smaller GIRK current than activation of M(2)-R. Adenovirus-driven overexpression of the A(1)-R caused an increase in current induced by adenosine (I(K(Ado))), whereas the M(2)-R-activated current (I(K(ACh))) was reduced. Here, we sought to get deeper insight into the mechanism causing this negative crosstalk. GIRK current in cultured rat atrial myocytes was recorded in whole cell mode. Adenovirus-driven RNA interference targeting the M(2)-R resulted in a reduction in I(K(ACh)) without affecting I(K(Ado)), arguing against a competition of the two receptors for common signaling complexes. The negative effect of A(1)-R overexpression on I(K(ACh)) was reduced by the A(1)-R antagonist DPCPX and augmented by the agonist chloro-N6-cyclopentyladenosin (CCPA). In native myocytes incubation with either CCPA or the muscarinic agonist carbachol resulted in reduction in I(K(ACh)) and I(K(Ado)), suggesting common pathways of A(1)-R and M(2)-R downregulation. In the absence of agonist, inhibition of adenosine deaminase by EHNA or exposure to AMP, less to ADP, but not ATP resulted in reduction of I(K(ACh)) and I(K(Ado)). Our data indicate that atrial myocytes generate adenosine from extracellular AMP, which activates A(1)-R in an autocrine fashion. Chronic activation of A(1)-R causes parallel downregulation of both A(1)-R and M(2)-R.


Asunto(s)
Comunicación Autocrina , Miocitos Cardíacos/metabolismo , Receptor de Adenosina A1/metabolismo , Receptor Muscarínico M2/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Agonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A1/farmacología , Adenosina Desaminasa/metabolismo , Inhibidores de la Adenosina Desaminasa/farmacología , Adenosina Monofosfato/metabolismo , Animales , Carbacol/farmacología , Células Cultivadas , Regulación hacia Abajo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Atrios Cardíacos , Agonistas Muscarínicos/farmacología , Técnicas de Placa-Clamp , Interferencia de ARN , Ratas , Receptor Cross-Talk , Receptor de Adenosina A1/genética , Receptor Muscarínico M2/genética , Xantinas/farmacología
10.
Cell Physiol Biochem ; 26(4-5): 503-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21063088

RESUMEN

RATIONALE: The plateau phase of the ventricular action potential is the result of balanced Ca(2+) influx and K(+) efflux. The action potential is terminated by repolarizing K(+) currents. Under ß-adrenergic stimulation, both the Ca(2+)-influx and the delayed rectifier K(+) currents I(K) are stimulated to adjust the cardiac action potential duration to the enhanced heart rate and to ascertain adequate increase in net Ca(2+) influx. Intracellularly, a Calsequestrin2 (CASQ2)-ryanodine receptor complex serves as the most effective Ca(2+) reservoir/release system to aid the control of intracellular Ca(2+) levels. Currently, it is unclear if disease-associated CASQ2 gene variants alter intracellular free Ca(2+) concentrations and if cardiac ion channels are affected by it. OBJECTIVE: The goal of this study is to test if CASQ2 determines intracellular free Ca(2+) concentrations and to identify cardiac ion channels that are affected by it. Further, we aim to study disease-associated CASQ2 gene variants in this context. METHODS AND RESULTS: Here, we study the effects of the CASQ2 mutations R33Q, F189L, and D307H, located in highly conserved regions, on the functions of cardiac potassium channels in Xenopus oocytes using two electrode voltage clamp. As a result, CASQ2 wild type and CASQ2-mutants modulated hERG functions differently. Free Ca(2+) measurements and molecular dynamics simulations imply alterations in Ca(2+) buffer capacity paralled by changes in the dynamic behavior of the CASQ2-mutants compared to CASQ2 wild type. CONCLUSIONS: These in vitro and in silico data suggest a regulatory role of CASQ2 on cytosolic Ca(2+) and hERG channels which may contribute to the etiology of CPVT.


Asunto(s)
Calsecuestrina/fisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Potenciales de Acción/fisiología , Sustitución de Aminoácidos , Animales , Calcio/metabolismo , Calsecuestrina/genética , Calsecuestrina/metabolismo , Humanos , Simulación de Dinámica Molecular , Mutación , Oocitos/metabolismo , Potasio/metabolismo , Estructura Terciaria de Proteína , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Taquicardia Ventricular/etiología , Xenopus/crecimiento & desarrollo
11.
ACS Chem Biol ; 15(1): 33-38, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31855412

RESUMEN

Phosphoinositides constitute a critical family of lipids that regulate numerous cellular processes. Phosphatidylinositol 4,5-bisphosphate (PIP2) is arguably the most important plasma membrane phosphoinositide and is involved in regulating diverse processes. It is also the precursor of phosphatidylinositol 3,4,5-trisphosphate (PIP3), which is critical for growth factor signaling, as well as membrane polarization and dynamics. Studying these lipids remains challenging, because of their compartmentalized activities and location-dependent signaling profiles. Here, we introduce several new genetically encoded fluorescent biosensors, including FRET-based and dimerization-dependent fluorescent protein (ddFP)-based biosensors, that enable real-time monitoring of PIP2 levels in live cells. In addition, we developed a red fluorescent biosensor for 3-phosphoinositides that can be co-imaged with the green PIP2 indicator. Simultaneous visualization of the dynamics of PIP2 and 3-phosphoinositides in the same cell shows that plasma membrane PIP3 formation upon EGF stimulation is coupled to a decrease in the local pool of PIP2.


Asunto(s)
Colorantes Fluorescentes/química , Imagen Óptica/métodos , Fosfatidilinositoles/análisis , Técnicas Biosensibles , Línea Celular , Membrana Celular/metabolismo , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Humanos , Cinética , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Dominios Proteicos , Transducción de Señal
12.
Methods Mol Biol ; 515: 107-23, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378115

RESUMEN

RNA interference (RNAi) represents the most frequently utilized technique to analyze proteins by loss of function assays. Protein synthesis is impaired by sequence-specific degradation of mRNA, which is triggered by short (19-28 nt) silencing RNAs (siRNA). Efficient gene silencing using RNAi has been demonstrated in numerous cell lines and primary cultured cells. Incorporation of siRNA into terminally differentiated mammalian cells, such as adult cardiac myocytes is limited by their resistance to standard transfection protocols. Viral delivery of short-hairpin RNA (shRNA) overcomes these limitations and allows efficient gene silencing in these cells. This chapter describes the generation and characterization of recombinant siRNA-encoding adenoviruses and their application to adult cardiac myocytes, which represent a standard experimental model in research related to cardiac physiology and pathophysiology. Feasibility of this approach is demonstrated by effective ablation (>80%) of both, a transgene encoding for eGFP and the endogenous muscarinic M(2) acetylcholine receptor.


Asunto(s)
Adenoviridae/genética , Diferenciación Celular , Técnicas de Transferencia de Gen , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Interferencia de ARN , Adenoviridae/aislamiento & purificación , Animales , Células Cultivadas , Clonación Molecular , Vectores Genéticos/genética , Humanos , Ratas , Virión/genética , Virión/aislamiento & purificación
13.
Circ Genom Precis Med ; 12(1): e002238, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30645171

RESUMEN

BACKGROUND: Inherited forms of sinus node dysfunction (SND) clinically include bradycardia, sinus arrest, and chronotropic incompetence and may serve as disease models to understand sinus node physiology and impulse generation. Recently, a gain-of-function mutation in the G-protein gene GNB2 led to enhanced activation of the GIRK (G-protein activated inwardly rectifying K+ channel). Thus, human cardiac GIRK channels are important for heart rate regulation and subsequently, genes encoding their subunits Kir3.1 and Kir3.4 ( KCNJ3 and KCNJ5) are potential candidates for inherited SND in human. METHODS: We performed a combined approach of targeted sequencing of KCNJ3 and KCNJ5 in 52 patients with idiopathic SND and subsequent whole exome sequencing of additional family members in a genetically affected patient. A putative novel disease-associated gene variant was functionally analyzed by voltage-clamp experiments using various heterologous cell expression systems (Xenopus oocytes, CHO cells, and rat atrial cardiomyocytes). RESULTS: In a 3-generation family with SND we identified a novel variant in KCNJ5 which leads to an amino acid substitution (p.Trp101Cys) in the first transmembrane domain of the Kir3.4 subunit of the cardiac GIRK channel. The identified variant cosegregated with the disease in the family and was absent in the Exome Variant Server and Exome Aggregation Consortium databases. Expression of mutant Kir3.4 (±native Kir3.1) in different heterologous cell expression systems resulted in increased GIRK currents ( IK,ACh) and a reduced inward rectification which was not compensated by intracellular spermidine. Moreover, in silico modeling of heterotetrameric mutant GIRK channels indicates a structurally altered binding site for spermine. CONCLUSIONS: For the first time, an inherited gain-of-function mutation in the human GIRK3.4 causes familial human SND. The increased activity of GIRK channels is likely to lead to a sustained hyperpolarization of pacemaker cells and thereby reduces heart rate. Modulation of human GIRK channels may pave a way for further treatment of cardiac pacemaking.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Mutación con Ganancia de Función , Predisposición Genética a la Enfermedad , Activación del Canal Iónico , Síndrome del Seno Enfermo/genética , Síndrome del Seno Enfermo/patología , Adolescente , Adulto , Anciano , Niño , Femenino , Humanos , Masculino , Potenciales de la Membrana , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Adulto Joven
14.
J Physiol ; 586(8): 2049-60, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18276732

RESUMEN

The effect of beta-adrenergic stimulation on endogenous G-protein-activated K(+) (GIRK) current has been investigated in atrial myocytes from hearts of adult rats. Beta-adrenergic stimulation (10 microm isoprenaline, Iso) had no effect on activation kinetics, peak current or steady-state current but resulted in slowing of deactivation upon washout of acetylcholine (ACh), the time constant (tau(d)) being increased by a factor of about 2.5. The effect of Iso could be mimicked by inclusion of cAMP (500 microm) in the filling solution of the patch clamp pipette. The Iso-induced increase in tau(d) was blocked by the selective beta(1) receptor antagonist CGP-20112A (2 microm) and by the PKA inhibitor H9 (100 microm included in the pipette solution). A candidate for mediating these effects is RGS10, one of the regulators of G-protein signalling (RGS) species expressed in cardiac myocytes. Overexpression of RGS10 by adenoviral gene transfer resulted in a reduction in tau(d) of 60%. Sensitivity of tau(d) to Iso remained in cells overexpressing RGS10. Overexpression of RGS4 caused a comparable reduction in tau(d), which became insensitive to Iso. Expression of an RGS10 carrying a mutation (RGS10-S168A), which deletes a PKA phosphorylation site, caused a decrease in tau(d) comparable to overexpression of wild-type RGS10. Sensitivity of tau(d) to Iso was lost in RGS10-S168A-expressing myocytes. Silencing of RGS10 by means of adenovirus-mediated transcription of a short hairpin RNA did not affect basal tau(d) but removed sensitivity to Iso. These data suggest that endogenous RGS10 has GTPase-activating protein (GAP) activity on the G-protein species that mediates activation of atrial GIRK channels. Moreover, RGS10, via PKA-dependent phosphorylation, enables a crosstalk between beta-adrenergic and muscarinic cholinergic signalling.


Asunto(s)
Función Atrial/fisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Potasio/metabolismo , Proteínas RGS/metabolismo , Animales , Células Cultivadas , Femenino , Atrios Cardíacos/citología , Masculino , Ratas , Ratas Endogámicas WKY
15.
Cell Physiol Biochem ; 21(4): 259-68, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18441514

RESUMEN

Inwardly-rectifying K+ channel subunits are not homogenously expressed in different cardiac tissues. In ventricular myocytes (VM) the background current-voltage relation is dominated by I(K1), carried by channels composed of Kir2.x subunits, which is less important in atrial myocytes (AM). On the other hand in AM a large G protein gated current carried by Kir3.1/3.4 complexes can be activated by stimulation of muscarinic M(2) receptors (I(K(ACh))), which is assumed to be marginal in VM. Recent evidence suggests that total current carried by cardiac inward-rectifiers (I(K(ATP)), I(K(ACh)), I(K1)) in both, AM and VM is limited, due to K+ accumulation/depletion. This lead us to hypothesize that in conventional whole celI recordings I(K(ACh)) in VM is underestimated as a consequence of constitutive I(K1). In that case a reduction in density of I(K1) should be paralleled by an increase in density of I(K(ACh)). Three different experimental strategies have been used to test for this hypothesis: (i) Adenovirus-driven expression of a dominant-negative mutant of Kir2.1, one of the subunits supposed to form I(K1) channels, in VM caused a reduction in I(K1)-density by about 80 %. In those cells I(K(ACh)) was increased about 4 fold. (ii) A comparable increase in I(K(ACh)) was observed upon reduction of I(K1) caused by adenovirus-mediated RNA interference.(iii) Ba2+ in a concentration of 2 microM blocks I(K1) in VM by about 60 % without affecting atrial I(K(ACh)). The reduction in I(K1) by 2 microM Ba2+ is paralleled by a reversible increase in I(K(ACh)) by about 100%. These data demonstrate that the increase in K+ conductance underlying ventricular I(K(ACh)) is largely underestimated, suggesting that it might be of greater physiological relevance than previously thought.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Arterias/metabolismo , Supervivencia Celular , Células Cultivadas , Electrofisiología , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , ARN Interferente Pequeño/genética , Ratas
16.
J Clin Invest ; 114(7): 994-1001, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467839

RESUMEN

Parasympathetic slowing of the heart rate is predominantly mediated by acetylcholine-dependent activation of the G protein-gated potassium (K+) channel (IK,ACh). This channel is composed of 2 inward-rectifier K+ (Kir) channel subunits, Kir3.1 and Kir3.4, that display distinct functional properties. Here we show that subunit composition of IK,ACh changes during embryonic development. At early stages, IK,ACh is primarily formed by Kir3.1, while in late embryonic and adult cells, Kir3.4 is the predominant subunit. This change in subunit composition results in reduced rectification of IK,ACh, allowing for marked K+ currents over the whole physiological voltage range. As a consequence, IK,ACh is able to generate the membrane hyperpolarization that underlies the strong negative chronotropy occurring in late- but not early-stage atrial cardiomyocytes upon application of muscarinic agonists. Both strong negative chronotropy and membrane hyperpolarization can be induced in early-stage cardiomyocytes by viral overexpression of the mildly rectifying Kir3.4 subunit. Thus, a switch in subunit composition is used to adopt IK,ACh to its functional role in adult cardiomyocytes.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Corazón/embriología , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Subunidades de Proteína/metabolismo , Acetilcolina/metabolismo , Potenciales de Acción/fisiología , Animales , Venenos de Abeja/farmacología , Carbacol/farmacología , Células Cultivadas , Agonistas Colinérgicos/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Ratones , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Subunidades de Proteína/genética , Receptor Muscarínico M2/metabolismo , Proteínas Recombinantes/metabolismo , Vasodilatadores/metabolismo
17.
Nat Commun ; 7: 12795, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731317

RESUMEN

Most small-molecule inhibitors of voltage-gated ion channels display poor subtype specificity because they bind to highly conserved residues located in the channel's central cavity. Using a combined approach of scanning mutagenesis, electrophysiology, chemical ligand modification, chemical cross-linking, MS/MS-analyses and molecular modelling, we provide evidence for the binding site for adamantane derivatives and their putative access pathway in Kv7.1/KCNE1 channels. The adamantane compounds, exemplified by JNJ303, are highly potent gating modifiers that bind to fenestrations that become available when KCNE1 accessory subunits are bound to Kv7.1 channels. This mode of regulation by auxiliary subunits may facilitate the future development of potent and highly subtype-specific Kv channel inhibitors.


Asunto(s)
Adamantano/análogos & derivados , Adamantano/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio KCNQ1/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Adamantano/química , Regulación Alostérica/efectos de los fármacos , Animales , Sitios de Unión , Reactivos de Enlaces Cruzados/química , Humanos , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Modelos Moleculares , Mutagénesis , Mutación , Oocitos , Bloqueadores de los Canales de Potasio/química , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Espectrometría de Masas en Tándem , Xenopus laevis
18.
Biochim Biophys Acta ; 1642(1-2): 67-77, 2003 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-12972295

RESUMEN

The predominant histamine receptor subtype in the supraventricular and ventricular tissue of various mammalian species is the H2 receptor (H2-R) subtype, which is known to couple to stimulatory G proteins (Gs), i.e. the major effects of this autacoid are an increase in sinus rate and in force of contraction. To investigate histamine effects in H2-R-transfected rat atrial myocytes, endogenous GIRK currents and L-type Ca2+ currents were used as functional assays. In H2-R-transfected myocytes, exposure to His resulted in a reversible augmentation of L-type Ca2+ currents, consistent with the established coupling of this receptor to the Gs-cAMP-PKA signalling pathway. Mammalian K+ channels composed of GIRK (Kir3.x) subunits are directly controlled by interaction with betagamma subunits released from G proteins, which couple to seven-helix receptors. In mock-transfected atrial cardiomyocytes, activation of muscarinic K+ channels (IK(ACh)) was limited to Gi-coupled receptors (M2R, A1R). In H2-R-overexpressing cells, histamine activated IK(ACh) via Gs-derived betagamma subunits since the histamine-induced current was insensitive to pertussis toxin. These data indicate that overexpression of Gs-coupled H2-R results in a loss of target specificity due to an increased agonist-induced release of Gs-derived betagamma subunits. When IK(ACh) was maximally activated by GTP-gamma-S, histamine induced an irreversible inhibition of the inward current in a fraction of H2-R-transfected cells. This inhibition is supposed to be mediated via a G(q/11)-PLC-mediated depletion of PIP2, suggesting a partial coupling of overexpressed H2-R to G(q/11). Dual coupling of H2-Rs to Gs and Gq is demonstrated for the first time in cardiac myocytes. It represents a novel mechanism to augment positive inotropic effects by activating two different signalling pathways via one type of histamine receptor. Activation of the Gs-cAMP-PKA pathway promotes Ca2+ influx through phosphorylation of L-type Ca2+ channels. Simultaneous activation of Gq-signalling pathways might result in phosphoinositide turnover and Ca2+ release from intracellular stores, thereby augmenting H2-induced increases in [Ca2+]i.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Atrios Cardíacos/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna , Receptores Histamínicos H2/metabolismo , Animales , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Subunidades alfa de la Proteína de Unión al GTP Gs/efectos de los fármacos , Atrios Cardíacos/efectos de los fármacos , Proteínas de Unión al GTP Heterotriméricas/efectos de los fármacos , Histamina/metabolismo , Histamina/farmacología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Ratas , Receptor Muscarínico M2 , Receptores Histamínicos H2/efectos de los fármacos , Receptores Histamínicos H2/genética , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección
19.
Med Klin (Munich) ; 100(11): 697-703, 2005 Nov 15.
Artículo en Alemán | MEDLINE | ID: mdl-16328176

RESUMEN

BACKGROUND AND PURPOSE: Vagus-induced atrial fibrillation is of particular clinical interest. The muscarinic potassium current I(K(ACh)) mediates the induction of vagus-induced atrial fibrillation. Selective inhibition of I(K(ACh)) seems to be an option to treat atrial fibrillation. The application of amiodarone, presently one of the most important antiarrhythmic agents in the parmacological treatment of atrial fibrillation, is limited by its adverse effects. KB130015, a new amiodarone derivative, and ibutilide are new class III antiarrhythmic agents. METHODS: In guinea-pig atrial myocytes the muscarinic potassium current (I(K(ACh))) was activated by acetylcholine and adenosine. The effect of KB130015 on I(K(ACh)) was measured using the whole-cell voltage-clamp method. RESULTS: KB130015 and ibutilide in a concentration of 50 microM effectively inhibited the muscarinic potassium current. The effect was concentrationdependent and reversible. The half-maximum effective concentration was 0.8 microM (KB130015) and 2.8 microM (ibutilide). The inhibition of I(K(ACh)) was independent of the mode of its activation. The adenosine-induced ion current was as well inhibited by both drugs as the acetylcholine-induced ion current. Via GTP-gamma-S irreversibly activated I(K(ACh)) was also inhibited by KB130015 and ibutilide, whereas intracellular application showed no effect on I(K(ACh)). CONCLUSION: KB130015 and ibutilide are potent inhibitors of IK(ACh). Their effect is most likely mediated by direct interaction with the extracellular part of the ion channel. Acute effects of KB130015 on ventricular myocardium are not known so far. Ibutilide on the other hand is known to inhibit I(kr). KB130015 is a promising antiarrhythmic agent for the pharmacotherapy of vagus-induced atrial fibrillation.


Asunto(s)
Antiarrítmicos/farmacología , Fibrilación Atrial/tratamiento farmacológico , Benzofuranos/farmacología , Atrios Cardíacos/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Antiarrítmicos/uso terapéutico , Benzofuranos/uso terapéutico , Células Cultivadas , Cobayas , Atrios Cardíacos/citología , Canales de Potasio/efectos de los fármacos , Receptores Muscarínicos/efectos de los fármacos , Sulfonamidas/uso terapéutico
20.
Mol Cell Endocrinol ; 412: 272-80, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25998841

RESUMEN

As a major cause of aldosterone producing adenomas, numerous gain-of-function mutations in the KCNJ5 gene (encoding the K(+) channel subunit GIRK4) have been identified. The human adrenocortical carcinoma cell line NCI-H295R is the most frequently used cellular model for in vitro studies related to regulation of aldosterone-synthesis. Because of the undefined role of KCNJ5 (GIRK4) in regulating synthesis of aldosterone, we aimed at identifying basal and G protein-activated GIRK4 currents in this paradigmatic cell line. The GIRK-specific blocker Tertiapin-Q did not affect basal current. Neither loading of the cells with GTP-γ-S via the patch-clamp pipette nor agonist stimulation of an infected A1-adenosine receptor resulted in activation of GIRK current. In cells co-infected with KCNJ5, robust activation of basal and adenosine-activated inward-rectifying current was observed. Although GIRK4 protein can be detected in Western blots of H295R homogenates, we suggest that GIRK4 in aldosterone-producing cells does not form functional G(ßγ)-activated channels.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Hiperaldosteronismo/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Humanos , Hiperaldosteronismo/genética , Potenciales de la Membrana , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA