Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 103(3): 2271-2319, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36731030

RESUMEN

The intercalated disc (ID) is a highly specialized structure that connects cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery and an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance of normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.


Asunto(s)
Miocardio , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/fisiología , Miocardio/metabolismo , Uniones Comunicantes/metabolismo , Arritmias Cardíacas
2.
Europace ; 26(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38875491

RESUMEN

AIMS: Patients with mutations in SCN5A encoding NaV1.5 often display variable severity of electrical and structural alterations, but the underlying mechanisms are not fully elucidated. We here investigate the combined modulatory effect of genetic background and age on disease severity in the Scn5a1798insD/+ mouse model. METHODS AND RESULTS: In vivo electrocardiogram and echocardiograms, ex vivo electrical and optical mapping, and histological analyses were performed in adult (2-7 months) and aged (8-28 months) wild-type (WT) and Scn5a1798insD/+ (mutant, MUT) mice from the FVB/N and 129P2 inbred strains. Atrio-ventricular (AV) conduction, ventricular conduction, and ventricular repolarization are modulated by strain, genotype, and age. An aging effect was present in MUT mice, with aged MUT mice of both strains showing prolonged QRS interval and right ventricular (RV) conduction slowing. 129P2-MUT mice were severely affected, with adult and aged 129P2-MUT mice displaying AV and ventricular conduction slowing, prolonged repolarization, and spontaneous arrhythmias. In addition, the 129P2 strain appeared particularly susceptible to age-dependent electrical, functional, and structural alterations including RV conduction slowing, reduced left ventricular (LV) ejection fraction, RV dilatation, and myocardial fibrosis as compared to FVB/N mice. Overall, aged 129P2-MUT mice displayed the most severe conduction defects, RV dilatation, and myocardial fibrosis, in addition to the highest frequency of spontaneous arrhythmia and inducible arrhythmias. CONCLUSION: Genetic background and age both modulate disease severity in Scn5a1798insD/+ mice and hence may explain, at least in part, the variable disease expressivity observed in patients with SCN5A mutations. Age- and genetic background-dependent development of cardiac structural alterations furthermore impacts arrhythmia risk. Our findings therefore emphasize the importance of continued assessment of cardiac structure and function in patients carrying SCN5A mutations.


Asunto(s)
Arritmias Cardíacas , Modelos Animales de Enfermedad , Fibrosis , Predisposición Genética a la Enfermedad , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Animales , Canal de Sodio Activado por Voltaje NAV1.5/genética , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Factores de Edad , Índice de Severidad de la Enfermedad , Sistema de Conducción Cardíaco/fisiopatología , Potenciales de Acción , Electrocardiografía , Fenotipo , Antecedentes Genéticos , Ratones de la Cepa 129 , Masculino , Frecuencia Cardíaca/genética , Miocardio/patología , Envejecimiento/genética
3.
Int J Mol Sci ; 24(6)2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36982449

RESUMEN

Chronic kidney disease (CKD) is represented by a diminished filtration capacity of the kidneys. End-stage renal disease patients need dialysis treatment to remove waste and toxins from the circulation. However, endogenously produced uremic toxins (UTs) cannot always be filtered during dialysis. UTs are among the CKD-related factors that have been linked to maladaptive and pathophysiological remodeling of the heart. Importantly, 50% of the deaths in dialysis patients are cardiovascular related, with sudden cardiac death predominating. However, the mechanisms responsible remain poorly understood. The current study aimed to assess the vulnerability of action potential repolarization caused by exposure to pre-identified UTs at clinically relevant concentrations. We exposed human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and HEK293 chronically (48 h) to the UTs indoxyl sulfate, kynurenine, or kynurenic acid. We used optical and manual electrophysiological techniques to assess action potential duration (APD) in the hiPSC-CMs and recorded IKr currents in stably transfected HEK293 cells (HEK-hERG). Molecular analysis of KV11.1, the ion channel responsible for IKr, was performed to further understand the potential mechanism underlying the effects of the UTs. Chronic exposure to the UTs resulted in significant APD prolongation. Subsequent assessment of the repolarization current IKr, often most sensitive and responsible for APD alterations, showed decreased current densities after chronic exposure to the UTs. This outcome was supported by lowered protein levels of KV11.1. Finally, treatment with an activator of the IKr current, LUF7244, could reverse the APD prolongation, indicating the potential modulation of electrophysiological effects caused by these UTs. This study highlights the pro-arrhythmogenic potential of UTs and reveals a mode of action by which they affect cardiac repolarization.


Asunto(s)
Células Madre Pluripotentes Inducidas , Insuficiencia Renal Crónica , Humanos , Tóxinas Urémicas , Células HEK293 , Potenciales de Acción , Células Madre Pluripotentes Inducidas/metabolismo , Diálisis Renal , Miocitos Cardíacos , Insuficiencia Renal Crónica/metabolismo
4.
Int J Mol Sci ; 24(21)2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37958923

RESUMEN

Inherited forms of arrhythmogenic and dilated cardiomyopathy (ACM and DCM) are characterized by variable disease expression and age-related penetrance. Calcium (Ca2+) is crucially important for proper cardiac function, and dysregulation of Ca2+ homeostasis seems to underly cardiomyopathy etiology. A polymorphism, c.286T>G p.(Ser96Ala), in the gene encoding the histidine-rich Ca2+ binding (HRC) protein, relevant for sarcoplasmic reticulum Ca2+ cycling, has previously been associated with a marked increased risk of life-threatening arrhythmias among idiopathic DCM patients. Following this finding, we investigated whether p.(Ser96Ala) affects major cardiac disease manifestations in carriers of the phospholamban (PLN) c.40_42delAGA; p.(Arg14del) pathogenic variant (cohort 1); patients diagnosed with, or predisposed to, ACM (cohort 2); and DCM patients (cohort 3). We found that the allele frequency of the p.(Ser96Ala) polymorphism was similar across the general European-American population (control cohort, 40.3-42.2%) and the different cardiomyopathy cohorts (cohorts 1-3, 40.9-43.9%). Furthermore, the p.(Ser96Ala) polymorphism was not associated with life-threatening arrhythmias or heart failure-related events across various patient cohorts. We therefore conclude that there is a lack of evidence supporting the important role of the HRC p.(Ser96Ala) polymorphism as a modifier in cardiomyopathy, refuting previous findings. Further research is required to identify bona fide genomic predictors for the stratification of cardiomyopathy patients and their risk for life-threatening outcomes.


Asunto(s)
Cardiomiopatías , Cardiomiopatía Dilatada , Humanos , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/genética , Cardiomiopatía Dilatada/genética , Histidina/genética , Polimorfismo Genético
5.
Int J Mol Sci ; 23(1)2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-35008484

RESUMEN

In arrhythmogenic cardiomyopathy (ACM) pathogenic variants are found in genes encoding desmosomal proteins and in non-desmosomal genes, such as phospholamban (PLN, p.Arg14del variant). Previous research showed that plakoglobin protein levels and localization in the cardiac tissue of ACM patients, and PLN p.Arg14del patients diagnosed with an ACM phenotype, are disturbed. Moreover, the effects of pathogenic variants in desmosomal genes are reflected in non-cardiac tissues like buccal mucosa cells (BMC) which could serve as a promising new and non-invasive tool to support diagnosis. We collected the BMC of 33 ACM patients, 17 PLN p.Arg14del patients and 34 controls, labelled the BMC with anti-plakoglobin antibodies at different concentrations, and scored their membrane labelling. We found that plakoglobin protein levels were significantly reduced in BMC obtained from diagnosed ACM patients and preclinical variant carriers when compared to controls. This effect was independent from age and sex. Moderate to strong correlations were found with the revised 2010 Task Force Criteria score which is commonly used for ACM diagnosis (rs = -0.67, n = 64, p < 0.0001 and rs = -0.71, n = 64, p < 0.0001). In contrast, plakoglobin scores in PLN p.Arg14del patients were comparable to controls (p > 0.209), which suggests differences in underlying etiology. However, for the individual diagnosis of the 'classical' ACM patient, this method might not be discriminative enough to distinguish true patients from variant carriers and controls, because of the high interindividual variability.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/patología , Mucosa Bucal/patología , Adulto , Displasia Ventricular Derecha Arritmogénica/metabolismo , Proteínas de Unión al Calcio/metabolismo , Desmosomas/metabolismo , Desmosomas/patología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mucosa Bucal/metabolismo , gamma Catenina/metabolismo
6.
Int J Mol Sci ; 22(21)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34769252

RESUMEN

Involvement of the Toll-like receptor 4 (TLR4) in maladaptive cardiac remodeling and heart failure (HF) upon pressure overload has been studied extensively, but less is known about the role of TLR2. Interplay and redundancy of TLR4 with TLR2 have been reported in other organs but were not investigated during cardiac dysfunction. We explored whether TLR2 deficiency leads to less adverse cardiac remodeling upon chronic pressure overload and whether TLR2 and TLR4 additively contribute to this. We subjected 35 male C57BL/6J mice (wildtype (WT) or TLR2 knockout (KO)) to sham or transverse aortic constriction (TAC) surgery. After 12 weeks, echocardiography and electrocardiography were performed, and hearts were extracted for molecular and histological analysis. TLR2 deficiency (n = 14) was confirmed in all KO mice by PCR and resulted in less hypertrophy (heart weight to tibia length ratio (HW/TL), smaller cross-sectional cardiomyocyte area and decreased brain natriuretic peptide (BNP) mRNA expression, p < 0.05), increased contractility (QRS and QTc, p < 0.05), and less inflammation (e.g., interleukins 6 and 1ß, p < 0.05) after TAC compared to WT animals (n = 11). Even though TLR2 KO TAC animals presented with lower levels of ventricular TLR4 mRNA than WT TAC animals (13.2 ± 0.8 vs. 16.6 ± 0.7 mg/mm, p < 0.01), TLR4 mRNA expression was increased in animals with the largest ventricular mass, highest hypertrophy, and lowest ejection fraction, leading to two distinct groups of TLR2 KO TAC animals with variations in cardiac remodeling. This variation, however, was not seen in WT TAC animals even though heart weight/tibia length correlated with expression of TLR4 in these animals (r = 0.078, p = 0.005). Our data suggest that TLR2 deficiency ameliorates adverse cardiac remodeling and that ventricular TLR2 and TLR4 additively contribute to adverse cardiac remodeling during chronic pressure overload. Therefore, both TLRs may be therapeutic targets to prevent or interfere in the underlying molecular processes.


Asunto(s)
Presión Sanguínea , Cardiomegalia/metabolismo , Ventrículos Cardíacos/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Remodelación Ventricular , Animales , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Masculino , Ratones , Ratones Noqueados , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
7.
Europace ; 22(10): 1579-1589, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32778883

RESUMEN

AIMS: SCN5A mutations are associated with arrhythmia syndromes, including Brugada syndrome, long QT syndrome type 3 (LQT3), and cardiac conduction disease. Long QT syndrome type 3 patients display atrio-ventricular (AV) conduction slowing which may contribute to arrhythmogenesis. We here investigated the as yet unknown underlying mechanisms. METHODS AND RESULTS: We assessed electrophysiological and molecular alterations underlying AV-conduction abnormalities in mice carrying the Scn5a1798insD/+ mutation. Langendorff-perfused Scn5a1798insD/+ hearts showed prolonged AV-conduction compared to wild type (WT) without changes in atrial and His-ventricular (HV) conduction. The late sodium current (INa,L) inhibitor ranolazine (RAN) normalized AV-conduction in Scn5a1798insD/+ mice, likely by preventing the mutation-induced increase in intracellular sodium ([Na+]i) and calcium ([Ca2+]i) concentrations. Indeed, further enhancement of [Na+]i and [Ca2+]i by the Na+/K+-ATPase inhibitor ouabain caused excessive increase in AV-conduction time in Scn5a1798insD/+ hearts. Scn5a1798insD/+ mice from the 129P2 strain displayed more severe AV-conduction abnormalities than FVB/N-Scn5a1798insD/+ mice, in line with their larger mutation-induced INa,L. Transverse aortic constriction (TAC) caused excessive prolongation of AV-conduction in FVB/N-Scn5a1798insD/+ mice (while HV-intervals remained unchanged), which was prevented by chronic RAN treatment. Scn5a1798insD/+-TAC hearts showed decreased mRNA levels of conduction genes in the AV-nodal region, but no structural changes in the AV-node or His bundle. In Scn5a1798insD/+-TAC mice deficient for the transcription factor Nfatc2 (effector of the calcium-calcineurin pathway), AV-conduction and conduction gene expression were restored to WT levels. CONCLUSIONS: Our findings indicate a detrimental role for enhanced INa,L and consequent calcium dysregulation on AV-conduction in Scn5a1798insD/+ mice, providing evidence for a functional mechanism underlying AV-conduction disturbances secondary to gain-of-function SCN5A mutations.


Asunto(s)
Calcio , Síndrome de QT Prolongado , Animales , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/terapia , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.5/genética , Sodio/metabolismo
8.
Biophys J ; 117(12): 2303-2315, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31623886

RESUMEN

Human-induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) are a virtually endless source of human cardiomyocytes that may become a great tool for safety pharmacology; however, their electrical phenotype is immature: they show spontaneous action potentials (APs) and an unstable and depolarized resting membrane potential (RMP) because of lack of IK1. Such immaturity hampers their application in assessing drug safety. The electronic overexpression of IK1 (e.g., through the dynamic clamp (DC) technique) is an option to overcome this deficit. In this computational study, we aim to estimate how much IK1 is needed to bring hiPSC-CMs to a stable and hyperpolarized RMP and which mathematical description of IK1 is most suitable for DC experiments. We compared five mature IK1 formulations (Bett, Dhamoon, Ishihara, O'Hara-Rudy, and ten Tusscher) with the native one (Paci), evaluating the main properties (outward peak, degree of rectification), and we quantified their effects on AP features (RMP, V˙max, APD50, APD90 (AP duration at 50 and 90% of repolarization), and APD50/APD90) after including them in the hiPSC-CM mathematical model by Paci. Then, we automatically identified the critical conductance for IK1 ( GK1, critical), the minimally required amount of IK1 suppressing spontaneous activity. Preconditioning the cell model with depolarizing/hyperpolarizing prepulses allowed us to highlight time dependency of the IK1 formulations. Simulations showed that inclusion of mature IK1 formulations resulted in hyperpolarized RMP and higher V˙max, and observed GK1, critical and the effect on AP duration strongly depended on IK1 formulation. Finally, the Ishihara IK1 led to shorter (-16.3%) and prolonged (+6.5%) APD90 in response to hyperpolarizing and depolarizing prepulses, respectively, whereas other models showed negligible effects. Fine-tuning of GK1 is an important step in DC experiments. Our computational work proposes a procedure to automatically identify how much IK1 current is required to inject to stop the spontaneous activity and suggests the use of the Ishihara IK1 model to perform DC experiments in hiPSC-CMs.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Modelos Cardiovasculares , Miocitos Cardíacos/citología , Péptidos Cíclicos/metabolismo , Potenciales de Acción , Células Madre Pluripotentes Inducidas/metabolismo , Potenciales de la Membrana , Miocitos Cardíacos/metabolismo
9.
J Mol Cell Cardiol ; 126: 86-95, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30452906

RESUMEN

BACKGROUND: The intercalated disc (ID) is important for cardiac remodeling and has become a subject of intensive research efforts. However, as yet the composition of the ID has still not been conclusively resolved and the role of many proteins identified in the ID, like Flotillin-2, is often unknown. The Flotillin proteins are known to be involved in the stabilization of cadherins and desmosomes in the epidermis and upon cancer development. However, their role in the heart has so far not been investigated. Therefore, in this study, we aimed at identifying the role of Flotillin-1 and Flotillin-2 in the cardiac ID. METHODS: Location of Flotillins in human and murine cardiac tissue was evaluated by fluorescent immunolabeling and co-immunoprecipitation. In addition, the effect of Flotillin knockout (KO) on proteins of the ID and in electrical excitation and conduction was investigated in cardiac samples of wildtype (WT), Flotillin-1 KO, Flotilin-2 KO and Flotilin-1/2 double KO mice. Consequences of Flotillin knockdown (KD) on cardiac function were studied (patch clamp and Multi Electrode Array (MEA)) in neonatal rat cardiomyocytes (NRCMs) transfected with siRNAs against Flotillin-1 and/or Flotillin-2. RESULTS: First, we confirmed presence in the ID and mutual binding of Flotillin-1 and Flotillin-2 in murine and human cardiac tissue. Flotillin KO mice did not show cardiac fibrosis, nor hypertrophy or changes in expression of the desmosomal ID proteins. However, protein expression of the cardiac sodium channel NaV1.5 was significantly decreased in Flotillin-1 and Flotillin-1/2 KO mice compared to WT mice. In addition, sodium current density showed a significant decrease upon Flotillin-1/2 KD in NRCMs as compared to scrambled siRNA-transfected NRCMs. MEA recordings of Flotillin-2 KD NRCM cultures showed a significantly decreased spike amplitude and a tendency of a reduced spike slope when compared to control and scrambled siRNA-transfected cultures. CONCLUSIONS: In this study, we demonstrate the presence of Flotillin-1, in addition to Flotillin-2 in the cardiac ID. Our findings indicate a modulatory role of Flotillins on NaV1.5 expression at the ID, with potential consequences for cardiac excitation.


Asunto(s)
Proteínas de la Membrana/metabolismo , Miocardio/metabolismo , Animales , Animales Recién Nacidos , Conexina 43/metabolismo , Humanos , Activación del Canal Iónico , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Ratas Wistar
11.
Eur Heart J ; 39(31): 2898-2907, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-29718149

RESUMEN

Aims: Management of patients with inherited cardiac ion channelopathy is hindered by variability in disease severity and sudden cardiac death (SCD) risk. Here, we investigated the modulatory role of hypertrophy on arrhythmia and SCD risk in sodium channelopathy. Methods and results: Follow-up data was collected from 164 individuals positive for the SCN5A-1795insD founder mutation and 247 mutation-negative relatives. A total of 38 (obligate) mutation-positive patients died suddenly or suffered life-threatening ventricular arrhythmia. Of these, 18 were aged >40 years, a high proportion of which had a clinical diagnosis of hypertension and/or cardiac hypertrophy. While pacemaker implantation was highly protective in preventing bradycardia-related SCD in young mutation-positive patients, seven of them aged >40 experienced life-threatening arrhythmic events despite pacemaker treatment. Of these, six had a diagnosis of hypertension/hypertrophy, pointing to a modulatory role of this co-morbidity. Induction of hypertrophy in adult mice carrying the homologous mutation (Scn5a1798insD/+) caused SCD and excessive conduction disturbances, confirming a modulatory effect of hypertrophy in the setting of the mutation. The deleterious effects of the interaction between hypertrophy and the mutation were prevented by genetically impairing the pro-hypertrophic response and by pharmacological inhibition of the enhanced late sodium current associated with the mutation. Conclusion: This study provides the first evidence for a modulatory effect of co-existing cardiac hypertrophy on arrhythmia risk and treatment efficacy in inherited sodium channelopathy. Our findings emphasize the need for continued assessment and rigorous treatment of this co-morbidity in SCN5A mutation-positive individuals.


Asunto(s)
Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/terapia , Cardiomegalia/complicaciones , Canalopatías/complicaciones , Canalopatías/terapia , Muerte Súbita Cardíaca/prevención & control , Hipertensión/complicaciones , Adulto , Factores de Edad , Anciano , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Estimulación Cardíaca Artificial , Canalopatías/genética , Canalopatías/fisiopatología , Muerte Súbita Cardíaca/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación , Canal de Sodio Activado por Voltaje NAV1.4/genética , Linaje , Factores de Riesgo , Resultado del Tratamiento
12.
Int J Mol Sci ; 20(17)2019 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-31438494

RESUMEN

Human variants in plakophilin-2 (PKP2) associate with most cases of familial arrhythmogenic cardiomyopathy (ACM). Recent studies show that PKP2 not only maintains intercellular coupling, but also regulates transcription of genes involved in Ca2+ cycling and cardiac rhythm. ACM penetrance is low and it remains uncertain, which genetic and environmental modifiers are crucial for developing the cardiomyopathy. In this study, heterozygous PKP2 knock-out mice (PKP2-Hz) were used to investigate the influence of exercise, pressure overload, and inflammation on a PKP2-related disease progression. In PKP2-Hz mice, protein levels of Ca2+-handling proteins were reduced compared to wildtype (WT). PKP2-Hz hearts exposed to voluntary exercise training showed right ventricular lateral connexin43 expression, right ventricular conduction slowing, and a higher susceptibility towards arrhythmias. Pressure overload increased levels of fibrosis in PKP2-Hz hearts, without affecting the susceptibility towards arrhythmias. Experimental autoimmune myocarditis caused more severe subepicardial fibrosis, cell death, and inflammatory infiltrates in PKP2-Hz hearts than in WT. To conclude, PKP2 haploinsufficiency in the murine heart modulates the cardiac response to environmental modifiers via different mechanisms. Exercise upon PKP2 deficiency induces a pro-arrhythmic cardiac remodeling, likely based on impaired Ca2+ cycling and electrical conduction, versus structural remodeling. Pathophysiological stimuli mainly exaggerate the fibrotic and inflammatory response.


Asunto(s)
Calcio/metabolismo , Cardiomiopatías/metabolismo , Haploinsuficiencia/fisiología , Enfermedad Autoinmune Experimental del Sistema Nervioso/etiología , Enfermedad Autoinmune Experimental del Sistema Nervioso/metabolismo , Placofilinas/metabolismo , Animales , Western Blotting , Cardiomiopatías/etiología , Cardiomiopatías/patología , Ecocardiografía , Electrocardiografía , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Haploinsuficiencia/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad Autoinmune Experimental del Sistema Nervioso/patología , Placofilinas/genética , Reacción en Cadena de la Polimerasa
13.
Molecules ; 24(1)2019 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-30621310

RESUMEN

Sustained pacemaker function is a challenge in biological pacemaker engineering. Human cardiomyocyte progenitor cells (CMPCs) have exhibited extended survival in the heart after transplantation. We studied whether lentivirally transduced CMPCs that express the pacemaker current If (encoded by HCN4) can be used as functional gene delivery vehicle in biological pacing. Human CMPCs were isolated from fetal hearts using magnetic beads coated with Sca-1 antibody, cultured in nondifferentiating conditions, and transduced with a green fluorescent protein (GFP)- or HCN4-GFP-expressing lentivirus. A patch-clamp analysis showed a large hyperpolarization-activated, time-dependent inward current (-20 pA/pF at -140 mV, n = 14) with properties typical of If in HCN4-GFP-expressing CMPCs. Gap-junctional coupling between CMPCs and neonatal rat ventricular myocytes (NRVMs) was demonstrated by efficient dye transfer and changes in spontaneous beating activity. In organ explant cultures, the number of preparations showing spontaneous beating activity increased from 6.3% in CMPC/GFP-injected preparations to 68.2% in CMPC/HCN4-GFP-injected preparations (P < 0.05). Furthermore, in CMPC/HCN4-GFP-injected preparations, isoproterenol induced a significant reduction in cycle lengths from 648 ± 169 to 392 ± 71 ms (P < 0.05). In sum, CMPCs expressing HCN4-GFP functionally couple to NRVMs and induce physiologically controlled pacemaker activity and may therefore provide an attractive delivery platform for sustained pacemaker function.


Asunto(s)
Técnicas de Transferencia de Gen , Ventrículos Cardíacos/trasplante , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/genética , Miocitos Cardíacos/trasplante , Canales de Potasio/genética , Células Madre/citología , Animales , Terapia Genética/métodos , Proteínas Fluorescentes Verdes/química , Ventrículos Cardíacos/patología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/uso terapéutico , Proteínas Musculares/uso terapéutico , Técnicas de Placa-Clamp , Canales de Potasio/uso terapéutico , Ratas , Trasplante de Células Madre
14.
J Mol Cell Cardiol ; 112: 58-63, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28823816

RESUMEN

Circadian rhythms are biorhythms with a 24-hour period that are regulated by molecular clocks. Several clinical and animal models have been developed to analyze the role of these rhythms in cardiovascular physiology, disease and therapy, but a convenient in vitro model that mimics both molecular and functional circadian effects of the heart is not available. Therefore, we established a neonatal rat cardiomyocyte model that recapitulates in vivo circadian rhythmicity, as measured by anti-phasic oscillatory mRNA expression of two core clock genes, Bmal1 and Per2 and that shows functional dependence on the clock as indicated by an oscillating response in apoptosis induced by doxorubicin, hydroperoxide or hypoxia. In addition, perturbation of the cardiac clock by the use of several compounds including Resveratrol and Ex-527 was found to result in loss of functional rhythmicity. This indicates that neonatal rat cardiomyocytes are a good model to investigate the cardiac circadian clock as well as a system that allows for fast and easy preclinical testing of the influence of compounds on circadian rhythmicity that might have crucial effects on cardiac health.


Asunto(s)
Ritmo Circadiano/fisiología , Corazón/fisiología , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Carbazoles/farmacología , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Doxorrubicina/farmacología , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ratas Wistar , Resveratrol , Estilbenos/farmacología
15.
Europace ; 19(4): 518-528, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28431070

RESUMEN

Cardiac disease is the leading cause of death in the developed world. Ventricular arrhythmias associated with myocardial ischaemia and/or infarction are a major contributor to cardiovascular mortality, and require improved prevention and treatment. Drugs, devices, and radiofrequency catheter ablation have made important inroads, but have significant limitations ranging from incomplete success to undesired toxicities and major side effects. These limitations derive from the nature of the intervention. Drugs are frequently ineffective, target the entire heart, and often do not deal with the specific arrhythmia trigger or substrate. Devices can terminate rapid rhythms but at best indirectly affect the underlying disease, while ablation, even when appropriately targeted, induces additional tissue damage. In contrast, exploration of gene and cell therapies are expected to provide a targeted, non-destructive, and potentially regenerative approach to ischaemia- and infarction-related arrhythmias. Although these approaches are in the early stages of development, they carry substantial potential to advance arrhythmia prevention and treatment.


Asunto(s)
Arritmias Cardíacas/genética , Arritmias Cardíacas/terapia , Trasplante de Células/tendencias , Terapia Genética/tendencias , Terapia Molecular Dirigida/tendencias , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Arritmias Cardíacas/etiología , Medicina Basada en la Evidencia , Predicción , Marcación de Gen/tendencias , Humanos , Infarto del Miocardio/complicaciones , Resultado del Tratamiento
16.
J Cardiovasc Electrophysiol ; 27(12): 1420-1428, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27572111

RESUMEN

INTRODUCTION: Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is associated with desmosomal mutations. Although desmosomal disruption affects both ventricles and atria, little is known about atrial involvement in ARVD/C. OBJECTIVE: To describe the extent and clinical significance of structural atrial involvement and atrial arrhythmias (AA) in ARVD/C stratified by genotype. METHODS: We included 71 patients who met ARVD/C Task Force Criteria and underwent cardiac magnetic resonance (CMR) imaging and molecular genetic analysis. Indexed atrial end-diastolic volume and area-length-ejection-fraction (ALEF) were evaluated on CMR and compared to controls with idiopathic right ventricular outflow tract tachycardia (n = 40). The primary outcome was occurrence of AA (atrial fibrillation or atrial flutter) during follow-up, recorded by 12-lead ECG, Holter monitoring or implantable cardioverter defibrillator (ICD) interrogation. RESULTS: Patients harbored a desmosomal plakophilin-2 (PKP2) (n = 37) or nondesmosomal phospholamban (PLN) (n = 14) mutation. In 20 subjects, no pathogenic mutation was identified. Compared to controls, right atrial (RA) volumes were reduced in PKP2 (P = 0.002) and comparable in PLN (P = 0.441) mutation carriers. In patients with no mutation identified, RA (P = 0.011) and left atrial (P = 0.034) volumes were increased. Bi-atrial ALEF showed no significant difference between the groups. AA were experienced by 27% of patients and occurred equally among PKP2 (30%) and no mutation identified patients (30%), but less among PLN mutation carriers (14%). CONCLUSION: Genotype influences atrial volume and occurrence of AA in ARVD/C. While the incidence of AA is similar in PKP2 mutation carriers and patients with no mutation identified, PKP2 mutation carriers have significantly smaller atria. This suggests a different arrhythmogenic mechanism.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Fibrilación Atrial/genética , Aleteo Atrial/genética , Función Atrial/genética , Proteínas de Unión al Calcio/genética , Atrios Cardíacos/fisiopatología , Mutación , Placofilinas/genética , Adulto , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/fisiopatología , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Aleteo Atrial/diagnóstico , Aleteo Atrial/fisiopatología , Estudios de Casos y Controles , Estudios Transversales , Análisis Mutacional de ADN , Electrocardiografía Ambulatoria , Femenino , Predisposición Genética a la Enfermedad , Atrios Cardíacos/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Fenotipo , Sistema de Registros , Factores de Riesgo
17.
J Mol Cell Cardiol ; 88: 82-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26410398

RESUMEN

BACKGROUND: One of the main contributors to maladaptive cardiac remodeling is fibrosis. Connective tissue growth factor (CTGF), a matricellular protein that is secreted into the cardiac extracellular matrix by both cardiomyocytes and fibroblasts, is often associated with development of fibrosis. However, recent studies have questioned the role of CTGF as a pro-fibrotic factor. Therefore, we aimed to investigate the effect of CTGF on cardiac fibrosis, and on functional, structural, and electrophysiological parameters in a mouse model of CTGF knockout (KO) and chronic pressure overload. METHODS AND RESULTS: A new mouse model of global conditional CTGF KO induced by tamoxifen-driven deletion of CTGF, was subjected to 16weeks of chronic pressure overload via transverse aortic constriction (TAC, control was sham surgery). CTGF KO TAC mice presented with hypertrophic hearts, and echocardiography revealed a decrease in contractility on a similar level as control TAC mice. Ex vivo epicardial mapping showed a low incidence of pacing-induced ventricular arrhythmias (2/12 in control TAC vs. 0/10 in CTGF KO TAC, n.s.) and a tendency towards recovery of the longitudinal conduction velocity of CTGF KO TAC hearts. Picrosirius Red staining on these hearts unveiled increased fibrosis at a similar level as control TAC hearts. Furthermore, genes related to fibrogenesis were also similarly upregulated in both TAC groups. Histological analysis revealed an increase in fibronectin and vimentin protein expression, a significant reduction in connexin43 (Cx43) protein expression, and no difference in NaV1.5 expression of CTGF KO ventricles as compared with sham treated animals. CONCLUSION: Conditional CTGF inhibition failed to prevent TAC-induced cardiac fibrosis and hypertrophy. Additionally, no large differences were found in other parameters between CTGF KO and control TAC mice. With no profound effect of CTGF on fibrosis formation, other factors or pathways are likely responsible for fibrosis development.


Asunto(s)
Síndrome de Brugada/genética , Cardiomegalia/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Miocardio/metabolismo , Remodelación Ventricular , Animales , Aorta/cirugía , Compuestos Azo , Síndrome de Brugada/etiología , Síndrome de Brugada/metabolismo , Síndrome de Brugada/patología , Trastorno del Sistema de Conducción Cardíaco , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Factor de Crecimiento del Tejido Conjuntivo/deficiencia , Conexina 43/genética , Conexina 43/metabolismo , Constricción Patológica/complicaciones , Constricción Patológica/cirugía , Ecocardiografía , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Cultivo de Órganos , Presión , Transducción de Señal , Vimentina/genética , Vimentina/metabolismo
18.
Physiology (Bethesda) ; 29(1): 72-83, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24382873

RESUMEN

Circadian rhythms are of major importance in mammalian physiology and disease. In this review, we give an overview of the present knowledge on origination of circadian rhythms. We discuss the development of both master and peripheral clocks and compare the origination of circadian rhythms in utero and in vitro.


Asunto(s)
Diferenciación Celular/fisiología , Ritmo Circadiano/fisiología , Animales , Humanos , Transducción de Señal/fisiología , Células Madre/fisiología
19.
Kidney Int ; 87(3): 557-63, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25229336

RESUMEN

Genetic loss-of-function defects of connexin 40 in renal juxtaglomerular cells are associated with renin-dependent hypertension. The dysregulation of renin secretion results from an intrarenal displacement of renin cells and an interruption of the negative feedback control of renin secretion by blood pressure. It is unknown whether this phenotype is secondary to developmental defects of juxtaglomerular renin cells due to connexin 40 malfunction, or whether acute functional defects of connexin 40 in the normal adult kidney can also lead to a similar dysregulation of renin secretion and hypertension. To address this question, we generated mice with an inducible deletion of connexin 40 in the adult kidney by crossing connexin 40-floxed mice with mice harboring a ubiquitously expressed tamoxifen-inducible Cre recombinase. Tamoxifen treatment in these mice strongly reduced connexin 40 mRNA and protein expression in the kidneys. These mice displayed persistent hypertension with renin expression shifted from the media layer of afferent arterioles to juxtaglomerular periglomerular cells. Control of renin secretion by the perfusion pressure was abolished in vitro, whereas in vivo plasma renin concentrations were increased. Thus, interruption of the connexin 40 gene in the adult kidney produced very similar changes in the renin system as had embryonic deletion. Hence, impairments of connexin 40 function in the normal adult kidney can cause renin-dependent hypertension.


Asunto(s)
Secuencia de Bases , Conexinas/genética , Hipertensión/genética , Hipertensión/fisiopatología , Renina/metabolismo , Eliminación de Secuencia , Animales , Presión Sanguínea/genética , Conexinas/metabolismo , Retroalimentación Fisiológica , Hipertensión/inducido químicamente , Integrasas/metabolismo , Aparato Yuxtaglomerular/metabolismo , Masculino , Ratones , ARN Mensajero/metabolismo , Renina/sangre , Tamoxifeno , Proteína alfa-5 de Unión Comunicante
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA