Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 326(2): H418-H425, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38099845

RESUMEN

Cardiac arrhythmias significantly contribute to mortality in Duchenne muscular dystrophy (DMD), a severe muscle illness caused by mutations in the gene encoding for the intracellular protein dystrophin. A major source for arrhythmia vulnerability in patients with DMD is impaired ventricular impulse conduction, which predisposes for ventricular asynchrony, decreased cardiac output, and the development of reentrant circuits. Using the dystrophin-deficient mdx mouse model for human DMD, we previously reported that the lack of dystrophin causes a significant loss of peak Na+ current (INa) in ventricular cardiomyocytes. This finding provided a mechanistic explanation for ventricular conduction defects and concomitant arrhythmias in the dystrophic heart. In the present study, we explored the hypothesis that empagliflozin (EMPA), an inhibitor of sodium/glucose cotransporter 2 in clinical use to treat type II diabetes and nondiabetic heart failure, rescues peak INa loss in dystrophin-deficient ventricular cardiomyocytes. We found that INa of cardiomyocytes derived from mdx mice, which had received clinically relevant doses of EMPA for 4 wk, was restored to wild-type level. Moreover, incubation of isolated mdx ventricular cardiomyocytes with 1 µM EMPA for 24 h significantly increased their peak INa. This effect was independent of Na+-H+ exchanger 1 inhibition by the drug. Our findings imply that EMPA treatment can rescue abnormally reduced peak INa of dystrophin-deficient ventricular cardiomyocytes. Long-term EMPA administration may diminish arrhythmia vulnerability in patients with DMD.NEW & NOTEWORTHY Dystrophin deficiency in cardiomyocytes leads to abnormally reduced Na+ currents. These can be rescued by long-term empagliflozin treatment.


Asunto(s)
Compuestos de Bencidrilo , Diabetes Mellitus Tipo 2 , Glucósidos , Distrofia Muscular de Duchenne , Animales , Ratones , Humanos , Distrofina/genética , Ratones Endogámicos mdx , Miocitos Cardíacos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Distrofia Muscular de Duchenne/genética , Arritmias Cardíacas/metabolismo , Sodio/metabolismo , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 24(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37298523

RESUMEN

Cortisol is a potent human steroid hormone that plays key roles in the central nervous system, influencing processes such as brain neuronal synaptic plasticity and regulating the expression of emotional and behavioral responses. The relevance of cortisol stands out in the disease, as its dysregulation is associated with debilitating conditions such as Alzheimer's Disease, chronic stress, anxiety and depression. Among other brain regions, cortisol importantly influences the function of the hippocampus, a structure central for memory and emotional information processing. The mechanisms fine-tuning the different synaptic responses of the hippocampus to steroid hormone signaling remain, however, poorly understood. Using ex vivo electrophysiology and wild type (WT) and miR-132/miR-212 microRNAs knockout (miRNA-132/212-/-) mice, we examined the effects of corticosterone (the rodent's equivalent to cortisol in humans) on the synaptic properties of the dorsal and ventral hippocampus. In WT mice, corticosterone predominantly inhibited metaplasticity in the dorsal WT hippocampi, whereas it significantly dysregulated both synaptic transmission and metaplasticity at dorsal and ventral regions of miR-132/212-/- hippocampi. Western blotting further revealed significantly augmented levels of endogenous CREB and a significant CREB reduction in response to corticosterone only in miR-132/212-/- hippocampi. Sirt1 levels were also endogenously enhanced in the miR-132/212-/- hippocampi but unaltered by corticosterone, whereas the levels of phospo-MSK1 were only reduced by corticosterone in WT, not in miR-132/212-/- hippocampi. In behavioral studies using the elevated plus maze, miRNA-132/212-/- mice further showed reduced anxiety-like behavior. These observations propose miRNA-132/212 as potential region-selective regulators of the effects of steroid hormones on hippocampal functions, thus likely fine-tuning hippocampus-dependent memory and emotional processing.


Asunto(s)
Corticosterona , MicroARNs , Ratones , Humanos , Animales , Corticosterona/farmacología , Corticosterona/metabolismo , Hidrocortisona/metabolismo , Hipocampo/metabolismo , MicroARNs/metabolismo , Plasticidad Neuronal
3.
Int J Neuropsychopharmacol ; 25(4): 280-282, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-34871422

RESUMEN

Psilocybin, a hallucinogen contained in "magic" mushrooms, holds great promise for the treatment of various psychiatric disorders, and early clinical trials are encouraging. Adverse cardiac events after intake of high doses of psilocybin and a trial reporting QT interval prolongation in the electrocardiogram attributed to the drug's main metabolite, psilocin, gave rise to safety concerns. Here we show that clinical concentrations of psilocin do not cause significant human ether-a-go-go-related gene (hERG) potassium channel inhibition, a major risk factor for adverse cardiac events. We conclude that hERG channel blockage by psilocin is not liable for psilocybin- associated cardiotoxic effects.


Asunto(s)
Alucinógenos , Trastornos Mentales , Cardiotoxicidad , Alucinógenos/efectos adversos , Humanos , Trastornos Mentales/inducido químicamente , Trastornos Mentales/tratamiento farmacológico , Canales de Potasio , Psilocibina/efectos adversos
4.
Pflugers Arch ; 472(1): 61-74, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31822999

RESUMEN

Neuronal nitric oxide synthase (nNOS) is considered a regulator of Cav1.2 L-type Ca2+ channels and downstream Ca2+ cycling in the heart. The commonest view is that nitric oxide (NO), generated by nNOS activity in cardiomyocytes, reduces the currents through Cav1.2 channels. This gives rise to a diminished Ca2+ release from the sarcoplasmic reticulum, and finally reduced contractility. Here, we report that nNOS inhibitor substances significantly increase intracellular Ca2+ transients in ventricular cardiomyocytes derived from adult mouse and rat hearts. This is consistent with an inhibitory effect of nNOS/NO activity on Ca2+ cycling and contractility. Whole cell currents through L-type Ca2+ channels in rodent myocytes, on the other hand, were not substantially affected by the application of various NOS inhibitors, or application of a NO donor substance. Moreover, the presence of NO donors had no effect on the single-channel open probability of purified human Cav1.2 channel protein reconstituted in artificial liposomes. These results indicate that nNOS/NO activity does not directly modify Cav1.2 channel function. We conclude that-against the currently prevailing view-basal Cav1.2 channel activity in ventricular cardiomyocytes is not substantially regulated by nNOS activity and NO. Hence, nNOS/NO inhibition of Ca2+ cycling and contractility occurs independently of direct regulation of Cav1.2 channels by NO.


Asunto(s)
Potenciales de Acción , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Femenino , Ventrículos Cardíacos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ornitina/análogos & derivados , Ornitina/farmacología , Ratas , Ratas Sprague-Dawley
5.
Am J Physiol Heart Circ Physiol ; 318(6): H1436-H1440, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32383994

RESUMEN

Cardiac arrhythmias significantly contribute to mortality in Duchenne muscular dystrophy (DMD), a degenerative muscle disease triggered by mutations in the gene encoding for the intracellular protein dystrophin. A major source for the arrhythmias in patients with DMD is impaired ventricular impulse conduction, which predisposes for ventricular asynchrony, decreased cardiac output, and the development of reentrant mechanisms. The reason for ventricular conduction impairments and the associated arrhythmias in the dystrophic heart has remained unidentified. In the present study, we explored the hypothesis that dystrophin-deficient cardiac Purkinje fibers have reduced Na+ currents (INa), which would represent a potential mechanism underlying slowed ventricular conduction in the dystrophic heart. Therefore, by using a Langendorff perfusion system, we isolated Purkinje fibers from the hearts of adult wild-type control and dystrophin-deficient mdx mice. Enhanced green fluorescent protein (eGFP) expression under control of the connexin 40 gene allowed us to discriminate Purkinje fibers from eGFP-negative ventricular working cardiomyocytes after cell isolation. Finally, we recorded INa from wild-type and dystrophic mdx Purkinje fibers for comparison by means of the whole cell patch clamp technique. We found substantially reduced INa densities in mdx compared with wild-type Purkinje fibers, suggesting that dystrophin deficiency diminishes INa. Because Na+ channels in the Purkinje fiber membrane represent key determinants of ventricular conduction velocity, we propose that reduced INa in Purkinje fibers at least partly explains impaired ventricular conduction and the associated arrhythmias in the dystrophic heart.NEW & NOTEWORTHY Dystrophic cardiac Purkinje fibers have abnormally reduced Na+ current densities. This explains impaired ventricular conduction in the dystrophic heart.


Asunto(s)
Arritmias Cardíacas/metabolismo , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Ramos Subendocárdicos/metabolismo , Canales de Sodio/metabolismo , Potenciales de Acción/fisiología , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/fisiopatología , Trastorno del Sistema de Conducción Cardíaco/complicaciones , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Masculino , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/fisiopatología , Sodio/metabolismo
6.
J Inherit Metab Dis ; 43(5): 1046-1055, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32441337

RESUMEN

Plasmalogens (Pls) are a class of membrane phospholipids which serve a number of essential biological functions. Deficiency of Pls is associated with common disorders such as Alzheimer's disease or ischemic heart disease. A complete lack of Pls due to genetically determined defective biosynthesis gives rise to rhizomelic chondrodysplasia punctata (RCDP), characterized by a number of severe disabling pathologic features and death in early childhood. Frequent cardiac manifestations of RCDP include septal defects, mitral valve prolapse, and patent ductus arteriosus. In a mouse model of RCDP, reduced nerve conduction velocity was partially rescued by dietary oral supplementation of the Pls precursor batyl alcohol (BA). Here, we examine the impact of Pls deficiency on cardiac impulse conduction in a similar mouse model (Gnpat KO). In-vivo electrocardiographic recordings showed that the duration of the QRS complex was significantly longer in Gnpat KO mice than in age- and sex-matched wild-type animals, indicative of reduced cardiac conduction velocity. Oral supplementation of BA for 2 months resulted in normalization of cardiac Pls levels and of the QRS duration in Gnpat KO mice but not in untreated animals. BA treatment had no effect on the QRS duration in age-matched wild-type mice. These data suggest that Pls deficiency is associated with increased ventricular conduction time which can be rescued by oral BA supplementation.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Condrodisplasia Punctata Rizomélica/tratamiento farmacológico , Éteres de Glicerilo/farmacología , Plasmalógenos/biosíntesis , Administración Oral , Animales , Arritmias Cardíacas/etiología , Condrodisplasia Punctata Rizomélica/fisiopatología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Electrocardiografía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Éteres Fosfolípidos/farmacología
7.
Proc Natl Acad Sci U S A ; 114(18): 4811-4815, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28373535

RESUMEN

Malignant hyperthermia (MH) is a clinical syndrome of skeletal muscle that presents as a hypermetabolic response to volatile anesthetic gases, where susceptible persons may develop lethally high body temperatures. Genetic predisposition mainly arises from mutations on the skeletal muscle ryanodine receptor (RyR). Dantrolene is administered to alleviate MH symptoms, but its mechanism of action and its influence on the Ca2+ transients elicited by MH triggers are unknown. Here, we show that Ca2+ release in the absence of Mg2+ is unaffected by the presence of dantrolene but that dantrolene becomes increasingly effective as cytoplasmic-free [Mg2+] (free [Mg2+]cyto) passes mM levels. Furthermore, we found in human muscle susceptible to MH that dantrolene was ineffective at reducing halothane-induced repetitive Ca2+ waves in the presence of resting levels of free [Mg2+]cyto (1 mM). However, an increase of free [Mg2+]cyto to 1.5 mM could increase the period between Ca2+ waves. These results reconcile previous contradictory reports in muscle fibers and isolated RyRs, where Mg2+ is present or absent, respectively, and define the mechanism of action of dantrolene is to increase the Mg2+ affinity of the RyR (or "stabilize" the resting state of the channel) and suggest that the accumulation of the metabolite Mg2+ from MgATP hydrolysis is required to make dantrolene administration effective in arresting an MH episode.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Dantroleno/farmacología , Magnesio/farmacología , Hipertermia Maligna , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Adulto , Animales , Femenino , Halotano/farmacología , Humanos , Masculino , Hipertermia Maligna/tratamiento farmacológico , Hipertermia Maligna/metabolismo , Hipertermia Maligna/patología , Músculo Esquelético/patología , Ratas , Ratas Wistar
8.
Proc Natl Acad Sci U S A ; 114(40): E8372-E8381, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28916735

RESUMEN

The mammalian heart undergoes maturation during postnatal life to meet the increased functional requirements of an adult. However, the key drivers of this process remain poorly defined. We are currently unable to recapitulate postnatal maturation in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), limiting their potential as a model system to discover regenerative therapeutics. Here, we provide a summary of our studies, where we developed a 96-well device for functional screening in human pluripotent stem cell-derived cardiac organoids (hCOs). Through interrogation of >10,000 organoids, we systematically optimize parameters, including extracellular matrix (ECM), metabolic substrate, and growth factor conditions, that enhance cardiac tissue viability, function, and maturation. Under optimized maturation conditions, functional and molecular characterization revealed that a switch to fatty acid metabolism was a central driver of cardiac maturation. Under these conditions, hPSC-CMs were refractory to mitogenic stimuli, and we found that key proliferation pathways including ß-catenin and Yes-associated protein 1 (YAP1) were repressed. This proliferative barrier imposed by fatty acid metabolism in hCOs could be rescued by simultaneous activation of both ß-catenin and YAP1 using genetic approaches or a small molecule activating both pathways. These studies highlight that human organoids coupled with higher-throughput screening platforms have the potential to rapidly expand our knowledge of human biology and potentially unlock therapeutic strategies.


Asunto(s)
Factores Biológicos/metabolismo , Puntos de Control del Ciclo Celular , Miocitos Cardíacos/metabolismo , Organoides/metabolismo , Células Madre Pluripotentes/metabolismo , Regeneración/fisiología , Adulto , Animales , Diferenciación Celular , Daño del ADN , Humanos , Masculino , Miocitos Cardíacos/citología , Organoides/citología , Células Madre Pluripotentes/citología , Ratas Sprague-Dawley
9.
Cell Physiol Biochem ; 53(1): 36-48, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31169990

RESUMEN

BACKGROUND/AIMS: Ivabradine lowers the heart rate by inhibition of hyperpolarisation-activated cyclic nucleotide-gated (HCN) channels mediating the 'funny' pacemaker current If in the sinoatrial node. It is clinically approved for the treatment of heart failure and angina pectoris. Due to its proposed high selectivity for If administration of ivabradine is not associated with the side effects commonly observed following the application of other heart rate lowering agents. Recent evidence, however, has shown significant affinity of ivabradine towards Kv11.1 (ether-a-go-go related gene, ERG) potassium channels. Despite the inhibition of Kv11.1 channels by ivabradine, cardiac action potential (AP) duration and heart rate corrected QT interval (QTc) of the human electrocardiogram (ECG) were not prolonged. We thus surmised that compensatory mechanisms might counteract the drug's inhibitory action on Kv11.1. METHODS: The effects of ivabradine on human Kv11.1 and Kv7.1 potassium, Cav1.2 calcium, and Nav1.5 sodium channels, heterologously expressed in tsA-201 cells, were studied in the voltage-clamp mode of the whole cell patch clamp technique. In addition, changes in action potential parameters of human induced pluripotent stem cell (iPSC) derived cardiomyocytes upon application of ivabradine were studied with current-clamp experiments. RESULTS: Here we show that ivabradine exhibits significant affinity towards cardiac ion channels other than HCN. We demonstrate for the first time inhibition of human voltage-gated Nav1.5 sodium channels at therapeutically relevant concentrations. Within this study we also confirm recent findings of human Kv11.1 inhibition by low µM concentrations of ivabradine and observed no prolongation of ventricular-like APs in cardiomyocytes derived from iPSCs. CONCLUSION: Our results provide an explanation why ivabradine, despite its affinity for Kv11.1 channels, does not prolong the cardiac AP and QTc interval. Furthermore, our results suggest the inhibition of voltage-gated Nav1.5 sodium channels to underlie the recent observations of slowed atrioventricular conduction by increased atrial-His bundle intervals upon administration of ivabradine.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Fármacos Cardiovasculares/farmacología , Canales Iónicos/metabolismo , Ivabradina/farmacología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/metabolismo , Línea Celular , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Canales Iónicos/antagonistas & inhibidores , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/química , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp
10.
Int J Mol Sci ; 19(11)2018 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-30360568

RESUMEN

Mutations in the gene encoding for the intracellular protein dystrophin cause severe forms of muscular dystrophy. These so-called dystrophinopathies are characterized by skeletal muscle weakness and degeneration. Dystrophin deficiency also gives rise to considerable complications in the heart, including cardiomyopathy development and arrhythmias. The current understanding of the pathomechanisms in the dystrophic heart is limited, but there is growing evidence that dysfunctional voltage-dependent ion channels in dystrophin-deficient cardiomyocytes play a significant role. Herein, we summarize the current knowledge about abnormalities in voltage-dependent sarcolemmal ion channel properties in the dystrophic heart, and discuss the potentially underlying mechanisms, as well as their pathophysiological relevance.


Asunto(s)
Cardiomiopatías/metabolismo , Distrofina/deficiencia , Distrofina/metabolismo , Canales Iónicos/metabolismo , Sarcolema/metabolismo , Animales , Arritmias Cardíacas , Humanos , Canales de Potasio/metabolismo , Canales de Sodio/metabolismo
11.
J Physiol ; 595(3): 759-776, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27621207

RESUMEN

KEY POINTS: Phosphatidylinositol-4,5-bisphosphate (PIP2 ) is a key regulator of many membrane proteins, including voltage-gated Kv7.2 channels. In this study, we identified the residues in five phosphorylation sites and their corresponding protein kinases, the former being clustered within one of four putative PIP2 -binding domains in Kv7.2. Dephosphorylation of these residues reduced the sensitivity of Kv7.2 channels towards PIP2 . Dephosphorylation of Kv7.2 affected channel inhibition via M1 muscarinic receptors, but not via bradykinin receptors. Our data indicated that phosphorylation of the Kv7.2 channel was necessary to maintain its low affinity for PIP2 , thereby ensuring the tight regulation of the channel via G protein-coupled receptors. ABSTRACT: The function of numerous ion channels is tightly controlled by G protein-coupled receptors (GPCRs). The underlying signalling mechanisms may involve phosphorylation of channel proteins and participation of phosphatidylinositol-4,5-bisphosphate (PIP2 ). Although the roles of both mechanisms have been investigated extensively, thus far only little has been reported on their interaction in channel modulation. GPCRs govern Kv7 channels, the latter playing a major role in the regulation of neuronal excitability by determining the levels of PIP2 and through phosphorylation. Using liquid chromatography-coupled mass spectrometry for Kv7.2 immunoprecipitates of rat brain membranes and transfected cells, we mapped a cluster of five phosphorylation sites in one of the PIP2-binding domains. To evaluate the effect of phosphorylation on PIP2 -mediated Kv7.2 channel regulation, a quintuple alanine mutant of these serines (S427/S436/S438/S446/S455; A5 mutant) was generated to mimic the dephosphorylated state. Currents passing through these mutated channels were less sensitive towards PIP2 depletion via the voltage-sensitive phosphatase Dr-VSP than were wild-type channels. In vitro phosphorylation assays with the purified C-terminus of Kv7.2 revealed that CDK5, p38 MAPK, CaMKIIα and PKA were able to phosphorylate the five serines. Inhibition of these protein kinases reduced the sensitivity of wild-type but not mutant Kv7.2 channels towards PIP2 depletion via Dr-VSP. In superior cervical ganglion neurons, the protein kinase inhibitors attenuated Kv7 current regulation via M1 receptors, but left unaltered the control by B2 receptors. Our results revealed that the phosphorylation status of serines located within a putative PIP2 -binding domain determined the phospholipid sensitivity of Kv7.2 channels and supported GPCR-mediated channel regulation.


Asunto(s)
Canal de Potasio KCNQ2/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Neuronas/fisiología , Fosforilación , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología
12.
Toxicol Appl Pharmacol ; 329: 309-317, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28641963

RESUMEN

Retigabine, currently used as antiepileptic drug, has a wide range of potential medical uses. Administration of the drug in patients can lead to QT interval prolongation in the electrocardiogram and to cardiac arrhythmias in rare cases. This suggests that the drug may perturb the electrical properties of the heart, and the underlying mechanisms were investigated here. Effects of retigabine on currents through human cardiac ion channels, heterologously expressed in tsA-201 cells, were studied in whole-cell patch-clamp experiments. In addition, the drug's impact on the cardiac action potential was tested. This was done using ventricular cardiomyocytes isolated from Langendorff-perfused guinea pig hearts and cardiomyocytes derived from human induced pluripotent stem cells. Further, to unravel potential indirect effects of retigabine on the heart which might involve the autonomic nervous system, membrane potential and noradrenaline release from sympathetic ganglionic neurons were measured in the absence and presence of the drug. Retigabine significantly inhibited currents through hKv11.1 potassium, hNav1.5 sodium, as well as hCav1.2 calcium channels, but only in supra-therapeutic concentrations. In a similar concentration range, the drug shortened the action potential in both guinea pig and human cardiomyocytes. Therapeutic concentrations of retigabine, on the other hand, were sufficient to inhibit the activity of sympathetic ganglionic neurons. We conclude that retigabine- induced QT interval prolongation, and the reported cases of cardiac arrhythmias after application of the drug in a typical daily dose range, cannot be explained by a direct modulatory effect on cardiac ion channels. They are rather mediated by indirect actions at the level of the autonomic nervous system.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Anticonvulsivantes/toxicidad , Arritmias Cardíacas/inducido químicamente , Carbamatos/toxicidad , Ganglios Simpáticos/efectos de los fármacos , Bloqueadores Ganglionares/toxicidad , Sistema de Conducción Cardíaco/efectos de los fármacos , Canales Iónicos/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Fenilendiaminas/toxicidad , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Bloqueadores de los Canales de Calcio/toxicidad , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/metabolismo , Ganglios Simpáticos/metabolismo , Ganglios Simpáticos/fisiopatología , Cobayas , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Preparación de Corazón Aislado , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Norepinefrina/metabolismo , Bloqueadores de los Canales de Potasio/toxicidad , Ratas Sprague-Dawley , Medición de Riesgo , Factores de Tiempo , Transfección , Bloqueadores del Canal de Sodio Activado por Voltaje/toxicidad
13.
Mol Pharmacol ; 88(5): 866-79, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26358763

RESUMEN

The clinically important suppression of high-frequency discharges of excitable cells by local anesthetics (LA) is largely determined by drug-induced prolongation of the time course of repriming (recovery from inactivation) of voltage-gated Na(+) channels. This prolongation may result from periodic drug-binding to a high-affinity binding site during the action potentials and subsequent slow dissociation from the site between action potentials ("dissociation hypothesis"). For many drugs it has been suggested that the fast inactivated state represents the high-affinity binding state. Alternatively, LAs may bind with high affinity to a native slow-inactivated state, thereby accelerating the development of this state during action potentials ("stabilization hypothesis"). In this case, slow recovery between action potentials occurs from enhanced native slow inactivation. To test these two hypotheses we produced serial cysteine mutations of domain IV segment 6 in rNav1.4 that resulted in constructs with varying propensities to enter fast- and slow-inactivated states. We tested the effect of the LA lidocaine on the time course of recovery from short and long depolarizing prepulses, which, under drug-free conditions, recruited mainly fast- and slow-inactivated states, respectively. Among the tested constructs the mutation-induced changes in native slow recovery induced by long depolarizations were not correlated with the respective lidocaine-induced slow recovery after short depolarizations. On the other hand, for long depolarizations the mutation-induced alterations in native slow recovery were significantly correlated with the kinetics of lidocaine-induced slow recovery. These results favor the "dissociation hypothesis" for short depolarizations but the "stabilization hypothesis" for long depolarizations.


Asunto(s)
Anestésicos Locales/farmacología , Lidocaína/farmacología , Proteínas Musculares/antagonistas & inhibidores , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Proteínas Musculares/fisiología , Mutagénesis , Ratas , Canales de Sodio/fisiología , Relación Estructura-Actividad
14.
Mol Pharmacol ; 88(1): 12-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25873594

RESUMEN

The membrane transporters for the monoamines serotonin (SERT) and dopamine (DAT) are prominent targets of various psychoactive substances, including competitive inhibitors, such as tricyclic antidepressants, methylphenidate, and cocaine. Upon rapid application of a substrate, SERT and DAT display an inwardly directed current comprised of a peak and a steady-state component. Binding of a competitive inhibitor to the transporter leads to reduction of the peak current amplitude because occupancy of the transporter by an inhibitor prevents the induction of the peak current by the substrate. We show that the inhibitory effect on the peak current can be used to study the association rate constant (k(on)), dissociation rate constant (k(off)), and equilibrium dissociation constant (K(D)) of chemically distinct SERT and DAT inhibitors, with high temporal precision and without the need of high-affinity radioligands as surrogates. We exemplify our approach by measuring the kinetics of cocaine, methylphenidate, and desipramine binding to SERT and DAT. Our analysis revealed that the selectivity of methylphenidate and desipramine for DAT and SERT, respectively, can be accounted for by their rate of association and not by the residence time in their respective binding sites.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inhibidores de la Captación de Neurotransmisores/farmacocinética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sitios de Unión , Cocaína/farmacocinética , Desipramina/farmacocinética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/química , Células HEK293 , Humanos , Metilfenidato/farmacocinética , Técnicas de Placa-Clamp/métodos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química
15.
J Biol Chem ; 289(31): 21770-81, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24947510

RESUMEN

Despite the availability of several crystal structures of bacterial voltage-gated Na(+) channels, the structure of eukaryotic Na(+) channels is still undefined. We used predictions from available homology models and crystal structures to modulate an external access pathway for the membrane-impermeant local anesthetic derivative QX-222 into the internal vestibule of the mammalian rNaV1.4 channel. Potassium channel-based homology models predict amino acid Ile-1575 in domain IV segment 6 to be in close proximity to Lys-1237 of the domain III pore-loop selectivity filter. The mutation K1237E has been shown previously to increase the diameter of the selectivity filter. We found that an access pathway for external QX-222 created by mutations of Ile-1575 was abolished by the additional mutation K1237E, supporting the notion of a close spatial relationship between sites 1237 and 1575. Crystal structures of bacterial voltage-gated Na(+) channels predict that the side chain of rNaV1.4 Trp-1531 of the domain IV pore-loop projects into the space between domain IV segment 6 and domain III pore-loop and, therefore, should obstruct the putative external access pathway. Indeed, mutations W1531A and W1531G allowed for exceptionally rapid access of QX-222. In addition, W1531G created a second non-selective ion-conducting pore, bypassing the outer vestibule but probably merging into the internal vestibule, allowing for control by the activation gate. These data suggest a strong structural similarity between bacterial and eukaryotic voltage-gated Na(+) channels.


Asunto(s)
Anestésicos Locales/farmacología , Activación del Canal Iónico , Canales de Sodio/efectos de los fármacos , Animales , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp , Conformación Proteica , Canales de Sodio/química , Canales de Sodio/genética , Xenopus laevis
16.
Cell Physiol Biochem ; 36(3): 1049-58, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26112643

RESUMEN

BACKGROUND/AIMS: Dysferlin plays a decisive role in calcium-dependent membrane repair in myocytes. Mutations in the encoding DYSF gene cause a number of myopathies, e.g. limb-girdle muscular dystrophy type 2B (LGMD2B). Besides skeletal muscle degenerative processes, dysferlin deficiency is also associated with cardiac complications. Thus, both LGMD2B patients and dysferlin-deficient mice develop a dilated cardiomyopathy. We and others have recently reported that dystrophin-deficient ventricular cardiomyocytes from mouse models of Duchenne muscular dystrophy show significant abnormalities in voltage-dependent ion channels, which may contribute to the pathophysiology in dystrophic cardiomyopathy. The aim of the present study was to investigate if dysferlin, like dystrophin, is a regulator of cardiac ion channels. METHODS AND RESULTS: By using the whole cell patch-clamp technique, we compared the properties of voltage-dependent calcium and sodium channels, as well as action potentials in ventricular cardiomyocytes isolated from the hearts of normal and dysferlin-deficient (dysf) mice. In contrast to dystrophin deficiency, the lack of dysferlin did not impair the ion channel properties and left action potential parameters unaltered. In connection with normal ECGs in dysf mice these results suggest that dysferlin deficiency does not perturb cardiac electrophysiology. CONCLUSION: Our study demonstrates that dysferlin does not regulate cardiac voltage-dependent ion channels, and implies that abnormalities in cardiac ion channels are not a universal characteristic of all muscular dystrophy types.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo T/metabolismo , Proteínas de la Membrana/deficiencia , Miocitos Cardíacos/fisiología , Canales de Sodio/metabolismo , Potenciales de Acción/fisiología , Animales , Bario/metabolismo , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo T/genética , Cationes Bivalentes , Cationes Monovalentes , Disferlina , Femenino , Expresión Génica , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Transporte Iónico , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Cultivo Primario de Células , Sodio/metabolismo , Canales de Sodio/genética
17.
Epilepsia ; 56(4): 647-57, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25779225

RESUMEN

OBJECTIVE: Within its range of therapeutic plasma concentrations, the anticonvulsant retigabine (ezogabine) is believed to selectively act on Kv7 channels. Here, the contribution of specific γ-aminobutyric acid (GABA)A receptor subtypes to the antiseizure effects of retigabine was investigated. METHODS: Using patch-clamp recordings, seizure-like activity, tonic currents, and GABA-induced currents in hippocampal neurons were tested for their sensitivity toward retigabine, as were recombinant GABAA receptors expressed in tsA 201 cells. RESULTS: Retigabine reduced seizure-like activity elicited by low Mg(2+) in a concentration-dependent manner with half maximal inhibition at 1 µm. Seizure-like activity triggered by blocking either Kv7 channels or GABAA receptors was equally reduced by retigabine, but when these channels/receptors were blocked simultaneously, the inhibition was lost. Retigabine (10 µm) enhanced bicuculline-sensitive tonic currents in hippocampal neurons, but failed to affect GABA-evoked currents. However, when receptors involved in phasic GABAergic inhibition were blocked by penicillin, retigabine did enhance GABA-evoked currents. In tsA 201 cells expressing various combinations of GABAA receptor subunits, 10 µm retigabine enhanced currents through α1ß2δ, α4ß2δ, α4ß3δ, and α6ß2δ receptors, but left currents through α1ß2γ2S, α4ß3γ2S, α5ß3γ2S, and α6ß2γ2S receptors unaltered. With αß receptors, retigabine diminished currents through α1ß2 and α4ß3, but increased currents through α6ß2 receptors. The enhancement of currents through α1ß2δ receptors by retigabine was concentration dependent and became significant at 1 µm. SIGNIFICANCE: These results demonstrate that retigabine is a subtype selective modulator of GABAA receptors with preference for extrasynaptic δ-containing receptors; this property may contribute to its broad antiepileptic effectiveness and explain its lack of effect on absence seizures.


Asunto(s)
Anticonvulsivantes/farmacología , Carbamatos/farmacología , Moduladores del GABA/farmacología , Fenilendiaminas/farmacología , Receptores de GABA-A/fisiología , Animales , Anticonvulsivantes/uso terapéutico , Carbamatos/uso terapéutico , Células Cultivadas , Relación Dosis-Respuesta a Droga , Moduladores del GABA/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Fenilendiaminas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología
18.
Molecules ; 20(2): 2208-28, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25642835

RESUMEN

The plant indole alkaloid ibogaine has shown promising anti-addictive properties in animal studies. Ibogaine is also anti-addictive in humans as the drug alleviates drug craving and impedes relapse of drug use. Although not licensed as therapeutic drug and despite safety concerns, ibogaine is currently used as an anti-addiction medication in alternative medicine in dozens of clinics worldwide. In recent years, alarming reports of life-threatening complications and sudden death cases, temporally associated with the administration of ibogaine, have been accumulating. These adverse reactions were hypothesised to be associated with ibogaine's propensity to induce cardiac arrhythmias. The aim of this review is to recapitulate the current knowledge about ibogaine's effects on the heart and the cardiovascular system, and to assess the cardiac risks associated with the use of this drug in anti- addiction therapy. The actions of 18-methoxycoronaridine (18-MC), a less toxic ibogaine congener with anti-addictive properties, are also considered.


Asunto(s)
Alucinógenos/efectos adversos , Ibogaína/efectos adversos , Animales , Bloqueadores de los Canales de Calcio/efectos adversos , Bloqueadores de los Canales de Calcio/farmacocinética , Bloqueadores de los Canales de Calcio/uso terapéutico , Alucinógenos/farmacocinética , Alucinógenos/uso terapéutico , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Ibogaína/farmacocinética , Ibogaína/uso terapéutico , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Factores de Riesgo , Trastornos Relacionados con Sustancias/tratamiento farmacológico
19.
Am J Physiol Heart Circ Physiol ; 306(4): H564-H573, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337461

RESUMEN

Duchenne muscular dystrophy (DMD), induced by mutations in the gene encoding for the cytoskeletal protein dystrophin, is an inherited disease characterized by progressive muscle weakness. Besides the relatively well characterized skeletal muscle degenerative processes, DMD is also associated with cardiac complications. These include cardiomyopathy development and cardiac arrhythmias. The current understanding of the pathomechanisms in the heart is very limited, but recent research indicates that dysfunctional ion channels in dystrophic cardiomyocytes play a role. The aim of the present study was to characterize abnormalities in L-type calcium channel function in adult dystrophic ventricular cardiomyocytes. By using the whole cell patch-clamp technique, the properties of currents through calcium channels in ventricular cardiomyocytes isolated from the hearts of normal and dystrophic adult mice were compared. Besides the commonly used dystrophin-deficient mdx mouse model for human DMD, we also used mdx-utr mice, which are both dystrophin- and utrophin-deficient. We found that calcium channel currents were significantly increased, and channel inactivation was reduced in dystrophic cardiomyocytes. Both effects enhance the calcium influx during an action potential (AP). Whereas the AP in dystrophic mouse cardiomyocytes was nearly normal, implementation of the enhanced dystrophic calcium conductance in a computer model of a human ventricular cardiomyocyte considerably prolonged the AP. Finally, the described dystrophic calcium channel abnormalities entailed alterations in the electrocardiograms of dystrophic mice. We conclude that gain of function in cardiac L-type calcium channels may disturb the electrophysiology of the dystrophic heart and thereby cause arrhythmias.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Corazón/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Animales , Cardiomiopatías/complicaciones , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Simulación por Computador , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos mdx , Modelos Cardiovasculares , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/metabolismo , Miocitos Cardíacos/metabolismo
20.
Cell Physiol Biochem ; 33(1): 205-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24481283

RESUMEN

BACKGROUND/AIMS: Cell transplantation into the heart is a new therapy after myocardial infarction. Its success, however, is impeded by poor donor cell survival and by limited transdifferentiation of the transplanted cells into functional cardiomyocytes. A promising strategy to overcome these problems is the induction of cardiomyogenic properties in donor cells by small molecules. METHODS: Here we studied cardiomyogenic effects of the small molecule compound cardiogenol C (CgC), and structural derivatives thereof, on lineage-committed progenitor cells by various molecular biological, biochemical, and functional assays. RESULTS: Treatment with CgC up-regulated cardiac marker expression in skeletal myoblasts. Importantly, the compound also induced cardiac functional properties: first, cardiac-like sodium currents in skeletal myoblasts, and secondly, spontaneous contractions in cardiovascular progenitor cell-derived cardiac bodies. CONCLUSION: CgC induces cardiomyogenic function in lineage-committed progenitor cells, and can thus be considered a promising tool to improve cardiac repair by cell therapy.


Asunto(s)
Compuestos de Anilina/farmacología , Biomarcadores/metabolismo , Linaje de la Célula/efectos de los fármacos , Miocardio/metabolismo , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Compuestos de Anilina/química , Animales , Factor Natriurético Atrial/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ratones , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pirimidinas/química , Ratas , Bibliotecas de Moléculas Pequeñas/química , Células Madre , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA