Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Scand J Immunol ; 93(1): e12995, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33188653

RESUMEN

Autoimmune congenital heart block (CHB) may develop in foetuses of women carrying anti-Ro/SSA and La/SSB autoantibodies and is characterized by disruption of signal conduction at the atrioventricular (AV) node, resulting in partial or complete AV block. If not fatal in utero, complete CHB typically requires lifelong cardiac pacing. No treatment has so far been unequivocally demonstrated to prevent or treat autoimmune CHB, and the relatively low incidence (1%-5%) and recurrence (12%-16%) rates of second/third-degree AV block add to the complexity of managing pregnancies in women with anti-Ro/La antibodies. Altogether, a better understanding of events leading to development of autoimmune CHB is needed to improve surveillance and treatment strategies. In the past decade, studies have started to look beyond the role of maternal autoantibodies in disease pathogenesis to assess other contributing factors such as foetal genetics and, more recently, immune responses in foetuses and neonates of anti-Ro/La antibody-positive women. In this review, we provide an update on the epidemiology, clinical presentation and current treatment approaches of autoimmune CHB, summarize the previously proposed pathogenic mechanisms implicating maternal autoantibodies, and discuss the recent findings of type I interferon (IFN) and innate immune activation in foetuses with autoimmune CHB and in neonates of anti-Ro/La antibody-positive mothers, and how these may contribute to autoimmune CHB pathogenesis.


Asunto(s)
Autoinmunidad , Susceptibilidad a Enfermedades , Bloqueo Cardíaco/congénito , Inmunidad Innata , Interferones/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Fibrosis , Bloqueo Cardíaco/diagnóstico , Bloqueo Cardíaco/epidemiología , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/metabolismo , Humanos , Inmunoterapia , Incidencia , Macrófagos/inmunología , Macrófagos/metabolismo , Recurrencia
2.
Am J Physiol Heart Circ Physiol ; 319(2): H396-H409, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32678707

RESUMEN

Myocardial ischemia leads to conduction slowing, cell-to-cell uncoupling, and arrhythmias. We previously demonstrated that varying perfusate sodium (Na+) and calcium (Ca2+) attenuates conduction slowing and arrhythmias during simulated ischemia with continuous perfusion. Cardioprotection was selectively associated with widening of the perinexus, a gap junction adjacent nanodomain important to ephaptic coupling. It is unknown whether perfusate composition affects the perinexus or ischemic conduction during nonsimulated ischemia, when coronary flow is reduced or halted. We hypothesized that altering preischemic perfusate composition could facilitate perinexal expansion and attenuate conduction slowing during global ischemia. To test this hypothesis, ex vivo guinea pig hearts (n = 49) were Langendorff perfused with 145 or 153 mM Na+ and 1.25 or 2.0 mM Ca2+ and optically mapped during 30 min of no-flow ischemia. Altering Na+ and Ca2+ did not substantially affect baseline conduction. Increasing Na+ and decreasing Ca2+ both lowered pacing thresholds, whereas increasing Ca2+ narrowed perinexal width (Wp). A least squares mean estimate revealed that reduced perfusate Na+ and Ca2+ resulted in the most severe conduction slowing during ischemia. Increasing Na+ alone modestly attenuated conduction slowing, yet significantly delayed the median time to conduction block (10 to 16 min). Increasing both Na+ and Ca2+ selectively widened Wp during ischemia (22.7 vs. 15.7 nm) and attenuated conduction slowing to the greatest extent. Neither repolarization nor levels of total or phosphorylated connexin43 correlated with conduction slowing or block. Thus, perfusate-dependent widening of the perinexus preserved ischemic conduction and may be an adaptive response to ischemic stress.NEW & NOTEWORTHY Conduction slowing during acute ischemia creates an arrhythmogenic substrate. We have shown that extracellular ionic concentrations can alter conduction by modulating ephaptic coupling. Here, we demonstrate increased extracellular sodium and calcium significantly attenuate conduction slowing during no-flow ischemia. This effect was associated with selective widening of the perinexus, an intercalated disc nanodomain and putative cardiac ephapse. These findings suggest that acute changes in ephaptic coupling may serve as an adaptive response to ischemic stress.


Asunto(s)
Bradicardia/prevención & control , Calcio/metabolismo , Bloqueo Cardíaco/prevención & control , Sistema de Conducción Cardíaco/metabolismo , Frecuencia Cardíaca , Isquemia Miocárdica/metabolismo , Sodio/metabolismo , Potenciales de Acción , Animales , Bradicardia/etiología , Bradicardia/metabolismo , Bradicardia/fisiopatología , Circulación Coronaria , Modelos Animales de Enfermedad , Cobayas , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Preparación de Corazón Aislado , Masculino , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/fisiopatología , Transducción de Señal , Factores de Tiempo
3.
Cell Physiol Biochem ; 54(4): 696-706, 2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32706220

RESUMEN

BACKGROUND/AIMS: Mutations of desmosomal genes are known to cause arrhythmogenic cardiomyopathy characterized by arrhythmias and sudden cardiac death. Previously, we described a novel genetic variant H1684R in desmoplakin gene (DSP), associated with a progressive cardiac conduction disease (PCCD). In the present study, we aimed to investigate an effect of the DSP-H1684R genetic variant on the activity of ion channels. METHODS: We used cardiomyocytes derived from induced pluripotent stem cells (iPSC cardiomyocytes) from a patient with DSP-H1684R genetic variant and from two healthy donors. Immunofluorescent staining and western blot analyses were used to characterize patient-specific cardiomyocytes. By the whole-cell voltage-clamp technique we estimated the activity of voltage-gated sodium, calcium, and potassium channels that are responsible for action potential generation and its shape. Action potentials' parameters were measured using whole-cell current-clamp technique. RESULTS: In patient-specific cardiomyocytes we observed both lower amplitudes of currents through sodium Nav1.5 channels and L-type calcium channels, but higher amplitude of current through transient-outward potassium channels in comparison to donor cardiomyocytes. Current-clamp measurements revealed shortening of action-potential in DSP-H1684R-carrying iPSC cardiomyocytes. Therefore, observed alterations in the channels activity might have a great impact on the properties of action potential and development of PCCD. CONCLUSION: Our results show that desmoplakin genetic variants, besides conduction slowing caused by structural heart remodeling, could affect multiple ion channel activity aggravating arrhythmia manifestation in PCCD.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/genética , Desmoplaquinas/genética , Bloqueo Cardíaco/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales Iónicos/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Canales de Calcio/fisiología , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Desmoplaquinas/metabolismo , Técnica del Anticuerpo Fluorescente , Bloqueo Cardíaco/metabolismo , Humanos , Canales Iónicos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Sodio Activados por Voltaje/fisiología
4.
Pediatrics ; 146(1)2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32561613

RESUMEN

Flecainide acetate is a Vaughan-Williams class IC antiarrhythmic drug prescribed for the treatment of supraventricular arrhythmias. It has a narrow therapeutic index and proarrhythmic effects even at therapeutic doses. Flecainide is metabolized by a CYP2D6 enzyme that exhibits polymorphism. In this case report, we present, to our best knowledge, the first case of toxicity contributed by genetic polymorphism in an infant. Our patient with recurrent supraventricular tachycardia was treated with a therapeutic dose of flecainide but developed heart block requiring extracorporeal membrane oxygenation support and subsequent treatment with lipid emulsion therapy. He was found to have supratherapeutic serum flecainide concentration, and gene testing revealed the patient to be an intermediate metabolizer. With this case report, we reinforce the importance of evaluating the CYP2D6 genotype before drug initiation in the neonatal population and recommend regular monitoring of serum flecainide levels and electrocardiograms in these patients.


Asunto(s)
Citocromo P-450 CYP2D6/genética , Electrocardiografía , Flecainida/efectos adversos , Bloqueo Cardíaco/inducido químicamente , Polimorfismo Genético , Taquicardia Supraventricular/tratamiento farmacológico , Citocromo P-450 CYP2D6/metabolismo , Genotipo , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/metabolismo , Humanos , Recién Nacido , Taquicardia Supraventricular/fisiopatología , Bloqueadores del Canal de Sodio Activado por Voltaje/efectos adversos
5.
PLoS One ; 15(6): e0233007, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32492036

RESUMEN

BACKGROUND: In humans, stillbirth describes the death of a fetus before birth after 28 weeks gestation, and accounts for approximately 2.6 million deaths worldwide annually. In high-income countries, up to half of stillbirths have an unknown cause and are described as "unexplained stillbirths"; this lack of understanding impairs efforts to prevent stillbirth. There are also few animal models of stillbirth, but those that have been described usually have significant placental abnormalities. This study describes a novel mutant murine model of fetal death with atrial conduction block due to an ErbB2 missense mutation which is not associated with abnormal placental morphology. METHODS: Phenotypic characterisation and histological analysis of the mutant mouse model was conducted. The mRNA distribution of the early cardiomyocyte marker Nkx2-5 was assessed via in situ hybridisation. Cardiac structure was quantified and cellular morphology evaluated by electron microscopy. Immunostaining was employed to quantify placental structure and cell characteristics on matched heterozygous and homozygous mutant placental samples. RESULTS: There were no structural abnormalities observed in hearts of mutant embryos. Comparable Nkx2-5 expression was observed in hearts of mutants and controls, suggesting normal cardiac specification. Additionally, there was no significant difference in the weight, placenta dimensions, giant cell characteristics, labyrinth tissue composition, levels of apoptosis, proliferation or vascularisation between placentas of homozygous mutant mice and controls. CONCLUSION: Embryonic lethality in the ErbB2 homozygous mutant mouse cannot be attributed to placental pathology. As such, we conclude the ErbB2M802R mutant is a model of stillbirth with a non-placental cause of death. The mechanism of the atrial block resulting from ErbB2 mutation and its role in embryonic death is still unclear. Studying this mutant mouse model could identify candidate genes involved in stillbirth associated with structural or functional cardiac defects.


Asunto(s)
Cardiopatías Congénitas/genética , Mutación Missense , Receptor ErbB-2/genética , Mortinato/genética , Animales , Modelos Animales de Enfermedad , Femenino , Bloqueo Cardíaco/congénito , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/patología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Heterocigoto , Proteína Homeótica Nkx-2.5/genética , Homocigoto , Humanos , Ratones , Ratones Mutantes , Miocardio/metabolismo , Miocardio/patología , Placenta/anomalías , Placenta/patología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
Eur Rev Med Pharmacol Sci ; 21(4 Suppl): 79-84, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29165759

RESUMEN

OBJECTIVE: Progressive cardiac conduction disease (PCCD) is a common pediatric heart conduction disorder. It is an autosomal inheritance of rare mutations, which leads to familial cases of PCCD. In these cases, the His-Purkinje system's conductive capacity is progressively deranged, involving either right or left bundle branch block. Also, QRS complexes display widening is an important characteristic that culminates in complete AV block, syncope, and sudden death. Mutations in TRPM4 gene that encodes for transient receptor potential melastatin 4 have recently been reported to cause familial cases of PCCD and heart block. TRPM4 conducts a Ca2+-activated non-selective monovalent cationic current leading to a negative plasma membrane potential. TRPM4 channels let Na+ ion influx, causing membrane depolarization, whereas, at positive membrane potentials, TRPM4 channels repolarize the membrane by facilitating K+ ion efflux from the cell. TRPM4 protein contains many regulatory motifs that confer voltage dependence, ATP/ADP sensitivity, and Ca2+ responsiveness. Mutational studies revealed the significance of the two-calmodulin binding sites at the N-terminus of for Ca2+ dependent activation of this channel. Mutations that reduce deSUMOylation increase the steady-state levels of active TRPM4 channels on the membrane without alteration of its sensitivity to Ca2+ or ATP or its voltage dependence of activation. Increased TRPM4 function interferes with cardiac conduction and eventually contributes to heart block. Both gain and loss of function mutations of TRPM4 are implicated in the cardiac block. Currently, the major therapeutic management of cardiac block due to TRPM4 mutations is implantation of a pacemaker to reinstate normal current propagation through AV node.


Asunto(s)
Bloqueo Cardíaco/patología , Canales Catiónicos TRPM/metabolismo , Adenosina Trifosfato/metabolismo , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/patología , Calcio/metabolismo , Niño , Bloqueo Cardíaco/tratamiento farmacológico , Bloqueo Cardíaco/metabolismo , Humanos , Potenciales de la Membrana/fisiología , Fenantrenos/uso terapéutico , Polimorfismo de Nucleótido Simple , Inhibidores de Proteínas Quinasas/uso terapéutico , Sumoilación , Canales Catiónicos TRPM/química , Canales Catiónicos TRPM/genética
7.
Biochem Biophys Res Commun ; 494(1-2): 346-351, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-28989025

RESUMEN

Malfunction of nodal pacemaker (Pm) cardiomyocytes (CMs) due to diseases or aging leads to rhythm generation disorders, necessitating electronic Pm implantation. We functionally reprogrammed human pluripotent stem cell (hPSC) derived-ventricular (V) CMs into -PmCMs via recombinant adeno-associated virus serotype 9 (rAAV9)-mediated overexpression of engineered HCN1 channel (HCN1ΔΔΔ) whose S3-S4 linker has been strategically deleted by design to promote cardiac pacemaking. rAAV9-HCN1ΔΔΔ-reprogrammed hPSC-PmCMs converted from -VCMs showed automaticity and action potential parameters typical of native nodal PmCMs. Implantation of rAAV9-HCN1ΔΔΔ-based BPm in a preclinical porcine model of complete heart block significantly reduced the dependence on device-supported pacing and generated spontaneous heart rhythms from the BPm. Collectively, these results have further laid the groundwork on BPm for future translation.


Asunto(s)
Dependovirus/metabolismo , Bloqueo Cardíaco/terapia , Ventrículos Cardíacos/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Canales de Potasio/metabolismo , Potenciales de Acción/fisiología , Animales , Diferenciación Celular , Reprogramación Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Miocitos Cardíacos/citología , Marcapaso Artificial , Células Madre Pluripotentes/citología , Canales de Potasio/genética , Porcinos
8.
Am J Physiol Heart Circ Physiol ; 313(3): H631-H640, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28626076

RESUMEN

The signature lesion of SSA/Ro autoantibody-associated congenital heart block (CHB) is fibrosis and a macrophage infiltrate, supporting an experimental focus on cues influencing the fibroblast component. The transcriptomes of human fetal cardiac fibroblasts were analyzed using two complementary approaches. Cardiac injury conditions were simulated in vitro by incubating human fetal cardiac fibroblasts with supernatants from macrophages transfected with the SSA/Ro-associated noncoding Y ssRNA. The top 10 upregulated transcripts in the stimulated fibroblasts reflected a type I interferon (IFN) response [e.g., IFN-induced protein 44-like (IFI44L), of MX dynamin-like GTPase (MX)1, MX2, and radical S-adenosyl methionine domain containing 2 (Rsad2)]. Within the fibrotic pathway, transcript levels of endothelin-1 (EDN1), phosphodiesterase (PDE)4D, chemokine (C-X-C motif) ligand (CXCL)2, and CXCL3 were upregulated, while others, including adenomedullin, RAP guanine nucleotide exchange factor 3 (RAPGEF3), tissue inhibitor of metalloproteinase (TIMP)1, TIMP3, and dual specificity phosphatase 1, were downregulated. Agnostic Database for Annotation, Visualization and Integrated Discovery analysis revealed a significant increase in inflammatory genes, including complement C3A receptor 1 (C3AR1), F2R-like thrombin/trypsin receptor 3, and neutrophil cytosolic factor 2. In addition, stimulated fibroblasts expressed high levels of phospho-MADS box transcription enhancer factor 2 [a substrate of MAPK5 (ERK5)], which was inhibited by BIX-02189, a specific inhibitor of ERK5. Translation to human disease leveraged an unprecedented opportunity to interrogate the transcriptome of fibroblasts freshly isolated and cell sorted without stimulation from a fetal heart with CHB and a matched healthy heart. Consistent with the in vitro data, five IFN response genes were among the top 10 most highly expressed transcripts in CHB fibroblasts. In addition, the expression of matrix-related genes reflected fibrosis. These data support the novel finding that cardiac injury in CHB may occur secondary to abnormal remodeling due in part to upregulation of type 1 IFN response genes.NEW & NOTEWORTHY Congenital heart block is a rare disease of the fetal heart associated with maternal anti-Ro autoantibodies which can result in death and for survivors, lifelong pacing. This study provides in vivo and in vitro transcriptome-support that injury may be mediated by an effect of Type I Interferon on fetal fibroblasts.


Asunto(s)
Anticuerpos Antinucleares/metabolismo , Corazón Fetal/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Bloqueo Cardíaco/congénito , Mediadores de Inflamación/metabolismo , Interferón Tipo I/metabolismo , Transcriptoma , Adulto , Anticuerpos Antinucleares/genética , Anticuerpos Antinucleares/inmunología , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Femenino , Corazón Fetal/inmunología , Corazón Fetal/patología , Fibroblastos/patología , Fibrosis , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/inmunología , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/patología , Humanos , Mediadores de Inflamación/inmunología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Miocardio , Comunicación Paracrina , Embarazo , Transfección
9.
Artículo en Inglés | MEDLINE | ID: mdl-27979911

RESUMEN

BACKGROUND: Heart block is associated with pulmonary hypertension, and the aim of the study was to test the hypothesis that the heart block is the result of a change in the ion channel transcriptome of the atrioventricular (AV) node. METHODS AND RESULTS: The most commonly used animal model of pulmonary hypertension, the monocrotaline-injected rat, was used. The functional consequences of monocrotaline injection were determined by echocardiography, ECG recording, and electrophysiological experiments on the Langendorff-perfused heart and isolated AV node. The ion channel transcriptome was measured by quantitative PCR, and biophysically detailed computer modeling was used to explore the changes observed. After monocrotaline injection, echocardiography revealed the pattern of pulmonary artery blood flow characteristic of pulmonary hypertension and right-sided hypertrophy and failure; the Langendorff-perfused heart and isolated AV node revealed dysfunction of the AV node (eg, 50% incidence of heart block in isolated AV node); and quantitative PCR revealed a widespread downregulation of ion channel and related genes in the AV node (eg, >50% downregulation of Cav1.2/3 and HCN1/2/4 channels). Computer modeling predicted that the changes in the transcriptome if translated into protein and function would result in heart block. CONCLUSIONS: Pulmonary hypertension results in a derangement of the ion channel transcriptome in the AV node, and this is the likely cause of AV node dysfunction in this disease.


Asunto(s)
Nodo Atrioventricular/metabolismo , Bloqueo Cardíaco/metabolismo , Hipertensión Pulmonar/metabolismo , Canales Iónicos/metabolismo , Transcriptoma , Animales , Nodo Atrioventricular/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ecocardiografía , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/fisiopatología , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/fisiopatología , Canales Iónicos/genética , Masculino , Monocrotalina , Reacción en Cadena de la Polimerasa , Ratas , Ratas Wistar
10.
J Autoimmun ; 67: 36-45, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26432597

RESUMEN

Based on the consistent demonstration of fibrosis of the atrioventricular node surrounded by macrophages and multinucleated giant cells in anti-Ro antibody exposed fetuses dying with heart block, this study focuses on macrophage signaling stimulated by ssRNA associated with the Ro60 protein and the impact of antagonizing innate cell drivers such as TLR7/8. Transcriptome and epigenetic modifications which affect transcription factors, NF-κB and STAT1, were selected to evaluate the phenotype of macrophages in which TLR7/8 was ligated following treatment with either anti-Ro60/Ro60/hY3 RNA immune complexes or transfection with hY3. Based on microarray, TNF and IL6 were among the most highly upregulated genes in both stimulated conditions, each of which was significantly inhibited by preincubation with hydroxychloroquine (HCQ). In contrast, following stimulation of macrophages with either TNF-α or IFN-α, which do not signal through TLR, the resultant gene expression was refractory to HCQ. Ligation of TLR7/8 resulted in increased histone methylation as measured by increased H3K4me2, a requirement for binding of NF-κB at certain promoters, specifically the kB1 region in the TNF promoter (ChIP-qPCR), which was significantly decreased by HCQ. In summary, these results support that the HCQ-sensitive phenotype of hY3 stimulated macrophages reflects the bifurcation of TLR downstream signals involving NF-κB and STAT 1 pathways and for the former dimethylation of H3K4. Accordingly, HCQ may act more as a preventive measure in downregulating the initial production of IFN-α or TNF-α and not affect the resultant autocoid stimulation reflected in TNF-α and IFN-α responsive genes. The beneficial scope of antimalarials in the prevention of organ damage, inclusive of heart block in an anti-Ro offspring or more broadly SLE, may include in part, a mechanism targeting TLR-dependent epigenetic modification.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Epigénesis Genética , Marcación de Gen , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Factores de Transcripción/genética , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Línea Celular , Endocitosis/inmunología , Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Unión Proteica , Ribonucleoproteínas/inmunología , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores
11.
Heart Rhythm ; 12(2): 397-408, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25444850

RESUMEN

BACKGROUND: Electrogram fractionation and atrial fibrosis are both thought to be pathophysiological hallmarks of evolving persistence of atrial fibrillation (AF), but recent studies in humans have shown that they do not colocalize. The interrelationship and relative roles of fractionation and fibrotic change in AF persistence therefore remain unclear. OBJECTIVE: The aim of the study was to examine the hypothesis that electrogram fractionation with increasing persistence of AF results from localized conduction slowing or block due to changes in atrial connexin distribution in the absence of fibrotic change. METHODS: Of 12 goats, atrial burst pacemakers maintained AF in 9 goats for up to 3 consecutive 4-week periods. After each 4-week period, 3 goats underwent epicardial mapping studies of the right atrium and examination of the atrial myocardium for immunodetection of connexins 43 and 40 (Cx43 and Cx40) and quantification of connective tissue. RESULTS: Despite refractoriness returning to normal in between each 4-week period of AF, there was a cumulative increase in the prevalence of fractionated atrial electrograms during both atrial pacing (control and 1, 2, and 3 months period of AF 0.3%, 1.3% ± 1.5%, 10.6% ± 2%, and 17% ± 5%, respectively; analysis of variance, P < .05) and AF (0.3% ± 0.1%, 2.3% ± 1.2%, 14% ± 2%, and 23% ± 3%; P < .05) caused by colocalized areas of conduction block during both pacing (local conduction velocity <10 cm/s: 0.1% ± 0.1%, 0.3% ± 0.6%, 6.5% ± 3%, and 6.9% ± 4%; P < .05) and AF (1.5% ± 0.5%, 2.7% ± 1.1%, 10.1% ± 1.2%, and 13.6% ± 0.4%; P < .05), associated with an increase in the heterogeneity of Cx40 and lateralization of Cx43 (lateralization scores: 1.75 ± 0.89, 1.44 ± 0.31, 2.85 ± 0.96, and 2.94 ± 0.31; P < .02), but not associated with change in connective tissue content or net conduction velocity. CONCLUSION: Electrogram fractionation with increasing persistence of AF results from slow localized conduction or block associated with changes in atrial connexin distribution in the absence of fibrotic change.


Asunto(s)
Fibrilación Atrial/fisiopatología , Conexinas/metabolismo , Técnicas Electrofisiológicas Cardíacas , Atrios Cardíacos/fisiopatología , Bloqueo Cardíaco/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Animales , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis , Cabras , Atrios Cardíacos/metabolismo , Bloqueo Cardíaco/diagnóstico , Bloqueo Cardíaco/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Imagen por Resonancia Cinemagnética , Pronóstico
13.
Cardiovasc Res ; 101(2): 326-34, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24296650

RESUMEN

BACKGROUND: A recent genome-wide association study identified a susceptibility locus for atrial fibrillation at the KCNN3 gene. Since the KCNN3 gene encodes for a small conductance calcium-activated potassium channel, we hypothesized that overexpression of the SK3 channel increases susceptibility to cardiac arrhythmias. METHODS AND RESULTS: We characterized the cardiac electrophysiological phenotype of a mouse line with overexpression of the SK3 channel. We generated homozygote (SK3(T/T)) and heterozygote (SK3(+/T)) mice with overexpression of the channel and compared them with wild-type (WT) controls. We observed a high incidence of sudden death among SK3(T/T) mice (7 of 19 SK3(T/T) mice). Ambulatory monitoring demonstrated that sudden death was due to heart block and bradyarrhythmias. SK3(T/T) mice displayed normal body weight, temperature, and cardiac function on echocardiography; however, histological analysis demonstrated that these mice have abnormal atrioventricular node morphology. Optical mapping demonstrated that SK3(T/T) mice have slower ventricular conduction compared with WT controls (SK3(T/T) vs. WT; 0.45 ± 0.04 vs. 0.60 ± 0.09 mm/ms, P = 0.001). Programmed stimulation in 1-month-old SK3(T/T) mice demonstrated inducible atrial arrhythmias (50% of SK3(T/T) vs. 0% of WT mice) and also a shorter atrioventricular nodal refractory period (SK3(T/T) vs. WT; 43 ± 6 vs. 52 ± 9 ms, P = 0.02). Three-month-old SK3(T/T) mice on the other hand displayed a trend towards a more prolonged atrioventricular nodal refractory period (SK3(T/T) vs. WT; 61 ± 1 vs. 52 ± 6 ms, P = 0.06). CONCLUSION: Overexpression of the SK3 channel causes an increased risk of sudden death associated with bradyarrhythmias and heart block, possibly due to atrioventricular nodal dysfunction.


Asunto(s)
Nodo Atrioventricular/metabolismo , Bradicardia/metabolismo , Muerte Súbita Cardíaca/etiología , Bloqueo Cardíaco/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción , Animales , Nodo Atrioventricular/anomalías , Nodo Atrioventricular/fisiopatología , Bradicardia/genética , Bradicardia/fisiopatología , Estimulación Cardíaca Artificial , Conexina 43/metabolismo , Electrocardiografía Ambulatoria , Predisposición Genética a la Enfermedad , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/fisiopatología , Heterocigoto , Homocigoto , Ratones , Ratones Transgénicos , Fenotipo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Factores de Tiempo , Regulación hacia Arriba , Imagen de Colorante Sensible al Voltaje
14.
Circ Heart Fail ; 6(2): 318-25, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23258573

RESUMEN

BACKGROUND: We investigated the contribution of inositol(1,4,5)-trisphosphate (Ins(1,4,5)P3 [IP3]) receptors (IP3-R) to disease progression in mouse models of dilated cardiomyopathy (DCM) and pressure overload hypertrophy. Mice expressing mammalian sterile 20-like kinase and dominant-negative phosphatidylinositol-3-kinase in heart (Mst1×dn-PI3K-2Tg; DCM-2Tg) develop severe DCM and conduction block, associated with increased expression of type 2 IP3-R (IP3-R(2)) and heightened generation of Ins(1,4,5)P3. Similar increases in Ins(1,4,5)P3 and IP3-R(2) are caused by transverse aortic constriction. METHODS AND RESULTS: To evaluate the contribution of IP3-R(2) to disease progression, the DCM-2Tg mice were further crossed with mice in which the type 2 IP3-R (IP3-R(2)-/-) had been deleted (DCM-2Tg×IP3-R(2)-/-) and transverse aortic constriction was performed on IP3-R(2)-/- mice. Hearts from DCM-2Tg mice and DCM-2Tg×IP3-R(2)-/- were similar in terms of chamber dilatation, atrial enlargement, and ventricular wall thinning. Electrophysiological changes were also similar in the DCM-2Tg mice, with and without IP3-R(2). Deletion of IP3-R(2) did not alter the progression of heart failure, because DCM-2Tg mice with and without IP3-R(2) had similarly reduced contractility, increased lung congestion, and atrial thrombus, and both strains died between 10 and 12 weeks of age. Loss of IP3-R(2) did not alter the progression of hypertrophy after transverse aortic constriction. CONCLUSIONS: We conclude that IP3-R(2) do not contribute to the progression of DCM or pressure overload hypertrophy, despite increased expression and heightened generation of the ligand, Ins(1,4,5)P3.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genotipo , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/deficiencia , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocardio/patología , Fenotipo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Tiempo , Función Ventricular Izquierda , Presión Ventricular , Remodelación Ventricular
15.
Circ Arrhythm Electrophysiol ; 5(1): 163-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22247482

RESUMEN

BACKGROUND: Progressive familial heart block type I (PFHBI) is a hereditary arrhythmia characterized by progressive conduction disturbances in the His-Purkinje system. PFHBI has been linked to genes such as SCN5A that influence cardiac excitability but not to genes that influence cell-to-cell communication. Our goal was to explore whether nucleotide substitutions in genes coding for connexin proteins would associate with clinical cases of PFHBI and if so, to establish a genotype-cell phenotype correlation for that mutation. METHODS AND RESULTS: We screened 156 probands with PFHBI. In addition to 12 sodium channel mutations, we found a germ line GJA5 (connexin40 [Cx40]) mutation (Q58L) in 1 family. Heterologous expression of Cx40-Q58L in connexin-deficient neuroblastoma cells resulted in marked reduction of junctional conductance (Cx40-wild type [WT], 22.2±1.7 nS, n=14; Cx40-Q58L, 0.56±0.34 nS, n=14; P<0.001) and diffuse localization of immunoreactive proteins in the vicinity of the plasma membrane without formation of gap junctions. Heteromeric cotransfection of Cx40-WT and Cx40-Q58L resulted in homogenous distribution of proteins in the plasma membrane rather than in membrane plaques in ≈50% of cells; well-defined gap junctions were observed in other cells. Junctional conductance values correlated with the distribution of gap junction plaques. CONCLUSIONS: Mutation Cx40-Q58L impairs gap junction formation at cell-cell interfaces. This is the first demonstration of a germ line mutation in a connexin gene that associates with inherited ventricular arrhythmias and emphasizes the importance of Cx40 in normal propagation in the specialized conduction system.


Asunto(s)
Fascículo Atrioventricular/metabolismo , Conexinas/genética , ADN/genética , Bloqueo Cardíaco/genética , Mutación , Biomarcadores/metabolismo , Western Blotting , Fascículo Atrioventricular/fisiopatología , Trastorno del Sistema de Conducción Cardíaco , Niño , Conexinas/metabolismo , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Predisposición Genética a la Enfermedad , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Frecuencia Cardíaca , Humanos , Inmunohistoquímica , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Pronóstico , Proteína alfa-5 de Unión Comunicante
16.
J Biol Chem ; 286(35): 30444-30454, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21730058

RESUMEN

Autoimmune associated congenital heart block (CHB) may result from pathogenic cross-talk between inflammatory and profibrosing pathways. Incubation of macrophages with immune complexes (IC) composed of Ro60, a target of the pathologic maternal autoantibodies necessary for CHB, hY3 ssRNA, and affinity-purified anti-Ro60 antibody induces the Toll-like receptor 7 (TLR7)-dependent generation of supernatants that provoke a fibrosing phenotype in human fetal cardiac fibroblasts. We show herein that these cells are a major source of TGFß and that endothelin-1 (ET-1) is one of the key components responsible for the profibrosing effects generated by stimulated macrophages. Supernatants from macrophages incubated with IC induced the fibroblast secretion of TGFß, which was inhibited by treating the macrophages with an antagonist of TLR7. Under the same conditions, the induced fibroblast secretion of TGFß was decreased by inhibitors of the ET-1 receptors ETa or ETb or by an anti-ET-1 antibody but not by an isotype control. Exogenous ET-1 induced a profibrosing phenotype, whereas fibroblasts transfected with either ETa or ETb siRNA were unresponsive to the profibrosing effects of the IC-generated macrophage supernatants. Immunohistochemistry of the hearts from two fetuses dying with CHB revealed the presence of ET-1-producing mononuclear cells in the septal region in areas of calcification and fibrosis. In conclusion, these data support a novel role of ET-1 in linking TLR7 inflammatory signaling to subsequent fibrosis and provide new insight in considering therapeutics for CHB.


Asunto(s)
Anticuerpos/química , Endotelina-1/fisiología , Fibrosis/metabolismo , Bloqueo Cardíaco/congénito , Inflamación , Ribonucleoproteínas/química , Receptor Toll-Like 7/metabolismo , Autoinmunidad , Femenino , Fibroblastos/metabolismo , Citometría de Flujo , Bloqueo Cardíaco/metabolismo , Humanos , Leucocitos Mononucleares/citología , Macrófagos/citología , Macrófagos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
17.
Circulation ; 123(20): 2192-203, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555709

RESUMEN

BACKGROUND: Electrical storm (ES), characterized by recurrent ventricular tachycardia/fibrillation, typically occurs in implantable cardioverter-defibrillator patients and adversely affects prognosis. However, the underlying molecular basis is poorly understood. In the present study, we report a new experimental model featuring repetitive episodes of implantable cardioverter-defibrillator firing for recurrent ventricular fibrillation (VF), in which we assessed involvement of Ca(2+)-related protein alterations in ES. METHODS AND RESULTS: We studied 37 rabbits with complete atrioventricular block for ≈80 days, all with implantable cardioverter-defibrillator implantation. All rabbits showed long-QT and VF episodes. Fifty-three percent of rabbits developed ES (≥3 VF episodes per 24-hour period; 103±23 VF episodes per rabbit). Expression/phosphorylation of Ca(2+)-handling proteins was assessed in left ventricular tissues from rabbits with the following: ES; VF episodes but not ES (non-ES); and controls. Left ventricular end-diastolic diameter increased comparably in ES and non-ES rabbits, but contractile dysfunction was significantly greater in ES than in non-ES rabbits. ES rabbits showed striking hyperphosphorylation of Ca(2+)/calmodulin-dependent protein kinase II, prominent phospholamban dephosphorylation, and increased protein phosphatase 1 and 2A expression versus control and non-ES rabbits. Ryanodine receptors were similarly hyperphosphorylated at Ser2815 in ES and non-ES rabbits, but ryanodine receptor Ser2809 and L-type Ca(2+) channel α-subunit hyperphosphorylation were significantly greater in ES versus non-ES rabbits. To examine direct effects of repeated VF/defibrillation, VF was induced 10 times in control rabbits. Repeated VF tissues showed autophosphorylated Ca(2+)/calmodulin-dependent protein kinase II upregulation and phospholamban dephosphorylation like those of ES rabbit hearts. Continuous infusion of a calmodulin antagonist (W-7) to ES rabbits reduced Ca(2+)/calmodulin-dependent protein kinase II hyperphosphorylation, suppressed ventricular tachycardia/fibrillation, and rescued left ventricular dysfunction. CONCLUSIONS: ES causes Ca(2+)/calmodulin-dependent protein kinase II activation and phospholamban dephosphorylation, which can explain the vicious cycle of arrhythmia promotion and mechanical dysfunction that characterizes ES.


Asunto(s)
Señalización del Calcio/fisiología , Modelos Animales de Enfermedad , Conejos , Taquicardia Ventricular/metabolismo , Fibrilación Ventricular/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Desfibriladores Implantables , Electrocardiografía , Inhibidores Enzimáticos/farmacología , Femenino , Bloqueo Cardíaco/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Recurrencia , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sulfonamidas/farmacología , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/terapia , Fibrilación Ventricular/diagnóstico , Fibrilación Ventricular/terapia
19.
Circ Cardiovasc Genet ; 3(4): 374-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20562447

RESUMEN

BACKGROUND: Isolated cardiac conduction block is a relatively common condition in young and elderly populations. Genetic predisposing factors have long been suspected because of numerous familial case reports. Deciphering genetic predisposing factors of conduction blocks may give a hint at stratifying conduction block carriers in a more efficient way. METHODS AND RESULTS: One Lebanese family and 2 French families with autosomal dominant isolated cardiac conduction blocks were used for linkage analysis. A maximum combined multipoint lod score of 10.5 was obtained on a genomic interval including more than 300 genes. After screening 12 genes of this interval for mutation, we found a heterozygous missense mutation of the TRPM4 gene in each family (p.Arg164Trp, p.Ala432Thr, and p.Gly844Asp). This gene encodes the TRPM4 channel, a calcium-activated nonselective cation channel of the transient receptor potential melastatin (TRPM) ion channel family. All 3 mutations result in an increased current density. This gain of function is due to an elevated TRPM4 channel density at the cell surface secondary to impaired endocytosis and deregulation of Small Ubiquitin MOdifier conjugation (SUMOylation). Furthermore, we showed by immunohistochemistry that TRPM4 channel signal level is higher in atrial cardiomyocytes than in common ventricular cells, but is highest in Purkinje fibers. Small bundles of highly TRPM4-positive cells were found in the subendocardium and in rare intramural bundles. CONCLUSIONS: the TRPM4 gene is a causative gene in isolated cardiac conduction disease with mutations resulting in a gain of function and TRPM4 channel being highly expressed in cardiac Purkinje fibers.


Asunto(s)
Canales Catiónicos TRPM/genética , Animales , Células CHO , Células COS , Trastorno del Sistema de Conducción Cardíaco , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Cricetulus , Familia , Femenino , Genes Dominantes , Ligamiento Genético , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/metabolismo , Humanos , Masculino , Mutación/fisiología , Linaje , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/fisiología , Transfección
20.
Indian J Exp Biol ; 45(11): 949-53, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18072538

RESUMEN

The kinins are implicated in the pathogenesis of scorpion envenomation. Therefore, this study was carried out to examine the involvement of kinins for the ECG abnormalities induced by M. tamulus concanesis, (BT) venom in anaesthetized rats. ECG was recorded using needle electrodes with limb lead II configuration. The PR interval, QRS wave pattern, QRS duration, ST segment and heart rate were examined in saline only, venom alone, and venom after aprotinin groups. BT venom (5 mg/kg) produced heart block of varying degree and ischemia-like changes in ECG wave pattern and the animals died within 30 min after exposure to venom. In aprotinin pretreated animals, the initial ECG changes produced by venom persisted, but after 15 min the ECG pattern improved and the animals survived for the entire period of observation (120 min). The results indicate that aprotinin protected the rats against the cardiotoxicity induced by BT venom.


Asunto(s)
Aprotinina/uso terapéutico , Bloqueo Cardíaco/prevención & control , Isquemia Miocárdica/prevención & control , Venenos de Escorpión/toxicidad , Animales , Aprotinina/administración & dosificación , Electrocardiografía , Bloqueo Cardíaco/inducido químicamente , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Cininas/antagonistas & inhibidores , Masculino , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Ratas , Escorpiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA