Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Am J Physiol Heart Circ Physiol ; 323(6): H1296-H1310, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36367695

RESUMEN

RBM20 cardiomyopathy is an arrhythmogenic form of dilated cardiomyopathy caused by mutations in the splicing factor RBM20. A recent study found a more severe phenotype in male patients with RBM20 cardiomyopathy patients than in female patients. Here, we aim to determine sex differences in an animal model of RBM20 cardiomyopathy and investigate potential underlying mechanisms. In addition, we aim to determine sex and gender differences in clinical parameters in a novel RBM20 cardiomyopathy patient cohort. We characterized an Rbm20 knockout (KO) mouse model, and show that splicing of key RBM20 targets, cardiac function, and arrhythmia susceptibility do not differ between sexes. Next, we performed deep phenotyping of these mice, and show that male and female Rbm20-KO mice possess transcriptomic and phosphoproteomic differences. Hypothesizing that these differences may influence the heart's ability to compensate for stress, we exposed Rbm20-KO mice to acute catecholaminergic stimulation and again found no functional differences. We also replicate the lack of functional differences in a mouse model with the Rbm20-R636Q mutation. Lastly, we present a patient cohort of 33 RBM20 cardiomyopathy patients and show that these patients do not possess sex and gender differences in disease severity. Current mouse models of RBM20 cardiomyopathy show more pronounced changes in gene expression and phosphorylation of cardiac proteins in male mice, but no sex differences in cardiac morphology and function. Moreover, other than reported before, male RBM20 cardiomyopathy patients do not present with worse cardiac function in a patient cohort from Germany and the Netherlands.NEW & NOTEWORTHY Optimal management of the cardiac disease is increasingly personalized, partly because of differences in outcomes between sexes. RBM20 cardiomyopathy has been described to be more severe in male patients, and this carries the risk that male patients are more scrutinized in the clinic than female patients. Our findings do not support this observation and suggest that treatment should not differ between male and female RBM20 cardiomyopathy patients, but instead should focus on the underlying disease mechanism.


Asunto(s)
Cardiomiopatías , Proteínas de Unión al ARN , Ratones , Masculino , Femenino , Animales , Proteínas de Unión al ARN/genética , Arritmias Cardíacas/genética , Mutación , Ratones Noqueados , Índice de Severidad de la Enfermedad
3.
Elife ; 102021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34372968

RESUMEN

Organ laterality refers to the left-right asymmetry in disposition and conformation of internal organs and is established during embryogenesis. The heart is the first organ to display visible left-right asymmetries through its left-sided positioning and rightward looping. Here, we present a new zebrafish loss-of-function allele for tbx5a, which displays defective rightward cardiac looping morphogenesis. By mapping individual cardiomyocyte behavior during cardiac looping, we establish that ventricular and atrial cardiomyocytes rearrange in distinct directions. As a consequence, the cardiac chambers twist around the atrioventricular canal resulting in torsion of the heart tube, which is compromised in tbx5a mutants. Pharmacological treatment and ex vivo culture establishes that the cardiac twisting depends on intrinsic mechanisms and is independent from cardiac growth. Furthermore, genetic experiments indicate that looping requires proper tissue patterning. We conclude that cardiac looping involves twisting of the chambers around the atrioventricular canal, which requires correct tissue patterning by Tbx5a.


Asunto(s)
Corazón/embriología , Organogénesis/genética , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Tipificación del Cuerpo , Embrión no Mamífero/embriología , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo
4.
Cell Rep ; 28(10): 2704-2714.e5, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31484079

RESUMEN

The human ether-a-go-go-related gene KCNH2 encodes the voltage-gated potassium channel underlying IKr, a current critical for the repolarization phase of the cardiac action potential. Mutations in KCNH2 that cause a reduction of the repolarizing current can result in cardiac arrhythmias associated with long-QT syndrome. Here, we investigate the regulation of KCNH2 and identify multiple active enhancers. A transcribed enhancer ∼85 kbp downstream of Kcnh2 physically contacts the promoters of two Kcnh2 isoforms in a cardiac-specific manner in vivo. Knockdown of its ncRNA transcript results in reduced expression of Kcnh2b and two neighboring mRNAs, Nos3 and Abcb8, in vitro. Genomic deletion of the enhancer, including the ncRNA transcription start site, from the mouse genome causes a modest downregulation of both Kcnh2a and Kcnh2b in the ventricles. These findings establish that the regulation of Kcnh2a and Kcnh2b is governed by a complex regulatory landscape that involves multiple partially redundantly acting enhancers.


Asunto(s)
Canal de Potasio ERG1/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Miocardio/metabolismo , Transcripción Genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Canal de Potasio ERG1/metabolismo , Femenino , Sitios Genéticos , Ventrículos Cardíacos/metabolismo , Humanos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Eliminación de Secuencia , Pez Cebra
5.
Sci Transl Med ; 8(354): 354ra115, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27582060

RESUMEN

Cardiac rhythm is extremely robust, generating 2 billion contraction cycles during the average human life span. Transcriptional control of cardiac rhythm is poorly understood. We found that removal of the transcription factor gene Tbx5 from the adult mouse caused primary spontaneous and sustained atrial fibrillation (AF). Atrial cardiomyocytes from the Tbx5-mutant mice exhibited action potential abnormalities, including spontaneous depolarizations, which were rescued by chelating free calcium. We identified a multitiered transcriptional network that linked seven previously defined AF risk loci: TBX5 directly activated PITX2, and TBX5 and PITX2 antagonistically regulated membrane effector genes Scn5a, Gja1, Ryr2, Dsp, and Atp2a2 In addition, reduced Tbx5 dose by adult-specific haploinsufficiency caused decreased target gene expression, myocardial automaticity, and AF inducibility, which were all rescued by Pitx2 haploinsufficiency in mice. These results defined a transcriptional architecture for atrial rhythm control organized as an incoherent feed-forward loop, driven by TBX5 and modulated by PITX2. TBX5/PITX2 interplay provides tight control of atrial rhythm effector gene expression, and perturbation of the co-regulated network caused AF susceptibility. This work provides a model for the molecular mechanisms underpinning the genetic implication of multiple AF genome-wide association studies loci and will contribute to future efforts to stratify patients for AF risk by genotype.


Asunto(s)
Redes Reguladoras de Genes , Frecuencia Cardíaca/genética , Proteínas de Homeodominio/genética , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Función Atrial/genética , Función Atrial/fisiología , Señalización del Calcio , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Haploinsuficiencia , Frecuencia Cardíaca/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Ratones Noqueados , Contracción Miocárdica/genética , Contracción Miocárdica/fisiología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología , Investigación Biomédica Traslacional , Proteína del Homeodomínio PITX2
6.
PLoS One ; 9(6): e99844, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24936875

RESUMEN

UNLABELLED: ChIP-seq has become a major tool for the genome-wide identification of transcription factor binding or histone modification sites. Most peak-calling algorithms require input control datasets to model the occurrence of background reads to account for local sequencing and GC bias. However, the GC-content of reads in Input-seq datasets deviates significantly from that in ChIP-seq datasets. Moreover, we observed that a commonly used peak calling program performed equally well when the use of a simulated uniform background set was compared to an Input-seq dataset. This contradicts the assumption that input control datasets are necessary to fatefully reflect the background read distribution. Because the GC-content of the abundant single reads in ChIP-seq datasets is similar to those of randomly sampled regions we designed a peak-calling algorithm with a background model based on overlapping single reads. The application, OccuPeak, uses the abundant low frequency tags present in each ChIP-seq dataset to model the background, thereby avoiding the need for additional datasets. Analysis of the performance of OccuPeak showed robust model parameters. Its measure of peak significance, the excess ratio, is only dependent on the tag density of a peak and the global noise levels. Compared to the commonly used peak-calling applications MACS and CisGenome, OccuPeak had the highest sensitivity in an enhancer identification benchmark test, and performed similar in an overlap tests of transcription factor occupation with DNase I hypersensitive sites and H3K27ac sites. Moreover, peaks called by OccuPeak were significantly enriched with cardiac disease-associated SNPs. OccuPeak runs as a standalone application and does not require extensive tweaking of parameters, making its use straightforward and user friendly. AVAILABILITY: http://occupeak.hfrc.nl.


Asunto(s)
Análisis de Secuencia de ADN , Programas Informáticos , Animales , Composición de Base , Secuencia de Bases , Inmunoprecipitación de Cromatina , Estudio de Asociación del Genoma Completo , Humanos , Modelos Genéticos , Polimorfismo de Nucleótido Simple
7.
Circ Res ; 115(4): 432-41, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24963028

RESUMEN

RATIONALE: The evolutionary conserved Tbx3/Tbx5 gene cluster encodes T-box transcription factors that play crucial roles in the development and homeostasis of the cardiac conduction system in human and mouse. Both genes are expressed in overlapping patterns and function in strictly tissue-specific and dose-dependent manners, yet, their regulation is poorly understood. OBJECTIVE: To analyze the mechanism underlying the complex regulation of the Tbx3/Tbx5 cluster. METHODS AND RESULTS: By probing the 3-dimensional architecture of the Tbx3/Tbx5 cluster using high-resolution circular chromosome conformation capture sequencing in vivo, we found that its regulatory landscape is in a preformed conformation similar in embryonic heart, limbs, and brain. Tbx3 and its flanking gene desert form a 1 Mbp loop between CCCTC-binding factor (CTCF)-binding sites that is separated from the neighboring Tbx5 loop. However, Ctcf inactivation did not result in transcriptional regulatory interaction between Tbx3 and Tbx5. Multiple sites within the Tbx3 locus contact the promoter, including sites corresponding to regions known to contain variations in the human genome influencing conduction. We identified an atrioventricular-specific enhancer and a pan-cardiac enhancer that contact the promoter and each other and synergize to activate transcription in the atrioventricular conduction system. CONCLUSIONS: We provide a high-resolution model of the 3-dimensional structure and function of the Tbx3/Tbx5 locus and show that the locus is organized in a preformed, permissive structure. The Tbx3 locus forms a CTCF-independent autonomous regulatory domain with multiple combinatorial regulatory elements that control the precise pattern of Tbx3 in the cardiac conduction system.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Sistema de Conducción Cardíaco/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Región de Flanqueo 3' , Animales , Sitios de Unión , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Factor de Unión a CCCTC , Cromosomas Artificiales Bacterianos , ADN Circular/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Sistema de Conducción Cardíaco/embriología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Familia de Multigenes , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Transcripción Genética , Activación Transcripcional
8.
Circ Cardiovasc Genet ; 7(4): 475-81, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850809

RESUMEN

BACKGROUND: The PR interval on the ECG reflects atrial depolarization and atrioventricular nodal delay which can be partially differentiated by P wave duration and PR segment, respectively. Genome-wide association studies have identified several genetic loci for PR interval, but it remains to be determined whether this is driven by P wave duration, PR segment, or both. METHODS AND RESULTS: We replicated 7 of the 9 known PR interval loci in 16 468 individuals of European ancestry. Four loci were unambiguously associated with PR segment, while the others were shared for P wave duration and PR segment. Next, we performed a genome-wide analysis on P wave duration and PR segment separately and identified 5 novel loci. Single-nucleotide polymorphisms in KCND3 (P=8.3×10(-11)) and FADS2 (P=2.7×10(-8)) were associated with P wave duration, whereas single-nucleotide polymorphisms near IL17D (P=2.3×10(-8)), in EFHA1 (P=3.3×10(-10)), and in LRCH1 (P=2.1×10(-8)) were associated with PR segment. Analysis on DNA elements indicated that genome-wide significant single-nucleotide polymorphisms were enriched at genomic regions suggesting active gene transcription in the human right atrium. Quantitative polymerase chain reaction showed that genes were significantly higher expressed in the right atrium and atrioventricular node compared with left ventricle (P=5.6×10(-6)). CONCLUSIONS: Genetic associations of PR interval seem to be mainly driven by genetic determinants of the PR segment. Some of the PR interval associations are strengthened by a directional consistent effect of genetic determinants of P wave duration. Through genome-wide association we also identified genetic variants specifically associated with P wave duration which might be relevant for cardiac biology.


Asunto(s)
Estudio de Asociación del Genoma Completo , Fallo Renal Crónico/genética , Nodo Atrioventricular/metabolismo , Proteínas de Unión al Calcio/genética , Estudios de Cohortes , Electrocardiografía , Ácido Graso Desaturasas/genética , Regulación de la Expresión Génica , Sitios Genéticos , Genotipo , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Humanos , Interleucina-17/genética , Fallo Renal Crónico/patología , Proteínas de Microfilamentos/genética , Polimorfismo de Nucleótido Simple , Canales de Potasio Shal/genética
9.
J Clin Invest ; 124(4): 1844-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24642470

RESUMEN

Variants in SCN10A, which encodes a voltage-gated sodium channel, are associated with alterations of cardiac conduction parameters and the cardiac rhythm disorder Brugada syndrome; however, it is unclear how SCN10A variants promote dysfunctional cardiac conduction. Here we showed by high-resolution 4C-seq analysis of the Scn10a-Scn5a locus in murine heart tissue that a cardiac enhancer located in Scn10a, encompassing SCN10A functional variant rs6801957, interacts with the promoter of Scn5a, a sodium channel-encoding gene that is critical for cardiac conduction. We observed that SCN5A transcript levels were several orders of magnitude higher than SCN10A transcript levels in both adult human and mouse heart tissue. Analysis of BAC transgenic mouse strains harboring an engineered deletion of the enhancer within Scn10a revealed that the enhancer was essential for Scn5a expression in cardiac tissue. Furthermore, the common SCN10A variant rs6801957 modulated Scn5a expression in the heart. In humans, the SCN10A variant rs6801957, which correlated with slowed conduction, was associated with reduced SCN5A expression. These observations establish a genomic mechanism for how a common genetic variation at SCN10A influences cardiac physiology and predisposes to arrhythmia.


Asunto(s)
Variación Genética , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Adulto , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Elementos de Facilitación Genéticos , Femenino , Regulación de la Expresión Génica , Corazón/embriología , Humanos , Masculino , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.5/fisiología , Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Polimorfismo de Nucleótido Simple , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Trends Cardiovasc Med ; 24(3): 99-104, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24360055

RESUMEN

The electrical activity of the heart depends on the correct interplay between key transcription factors and cis-regulatory elements, which together regulate the proper heterogeneous expression of genes encoding for ion channels and other proteins. Genome-wide association studies of ECG parameters implicated genetic variants in the genes for these factors and ion channels modulating conduction and depolarization. Here, we review recent insights into the regulation of localized expression of ion channel genes and the mechanism by which a single-nucleotide polymorphism (SNP) associated with alterations in cardiac conduction patterns in humans affects the transcriptional regulation of the sodium channel genes, SCN5A and SCN10A. The identification of regulatory elements of electrical activity genes helps to explain the impact of genetic variants in non-coding regulatory DNA sequences on regulation of cardiac conduction and the predisposition for cardiac arrhythmias.


Asunto(s)
Arritmias Cardíacas/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Polimorfismo de Nucleótido Simple/genética , Humanos
11.
J Clin Invest ; 122(7): 2519-30, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22706305

RESUMEN

The contraction pattern of the heart relies on the activation and conduction of the electrical impulse. Perturbations of cardiac conduction have been associated with congenital and acquired arrhythmias as well as cardiac arrest. The pattern of conduction depends on the regulation of heterogeneous gene expression by key transcription factors and transcriptional enhancers. Here, we assessed the genome-wide occupation of conduction system-regulating transcription factors TBX3, NKX2-5, and GATA4 and of enhancer-associated coactivator p300 in the mouse heart, uncovering cardiac enhancers throughout the genome. Many of the enhancers colocalized with ion channel genes repressed by TBX3, including the clustered sodium channel genes Scn5a, essential for cardiac function, and Scn10a. We identified 2 enhancers in the Scn5a/Scn10a locus, which were regulated by TBX3 and its family member and activator, TBX5, and are functionally conserved in humans. We also provided evidence that a SNP in the SCN10A enhancer associated with alterations in cardiac conduction patterns in humans disrupts TBX3/TBX5 binding and reduces the cardiac activity of the enhancer in vivo. Thus, the identification of key regulatory elements for cardiac conduction helps to explain how genetic variants in noncoding regulatory DNA sequences influence the regulation of cardiac conduction and the predisposition for cardiac arrhythmias.


Asunto(s)
Elementos de Facilitación Genéticos , Polimorfismo de Nucleótido Simple , Canales de Sodio/genética , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Secuencia de Consenso , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Variación Genética , Sistema de Conducción Cardíaco/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Canal de Sodio Activado por Voltaje NAV1.8 , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Análisis de Secuencia de ADN , Proteínas de Dominio T Box/metabolismo , Pez Cebra
12.
Cardiovasc Res ; 94(3): 439-49, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22419669

RESUMEN

AIM: Treatment of disorders of the sinus node or the atrioventricular node requires insights into the molecular mechanisms of development and homoeostasis of these pacemaker tissues. In the developing heart, transcription factor TBX3 is required for pacemaker and conduction system development. Here, we explore the role of TBX3 in the adult heart and investigate whether TBX3 is able to reprogramme terminally differentiated working cardiomyocytes into pacemaker cells. METHODS AND RESULTS: TBX3 expression was ectopically induced in cardiomyocytes of adult transgenic mice using tamoxifen. Expression analysis revealed an efficient switch from the working myocardial expression profile to that of the pacemaker myocardium. This included suppression of genes encoding gap junction subunits (Cx40, Cx43), the cardiac Na(+) channel (Na(V)1.5; I(Na)), and inwardly rectifying K(+) ion channels (K(ir) genes; I(K1)). Concordantly, we observed conduction slowing in these hearts and reductions in I(Na) and I(K1) in cardiomyocytes isolated from these hearts. The reduction in I(K1) resulted in a more depolarized maximum diastolic potential, thus enabling spontaneous diastolic depolarization. Neither ectopic pacemaker activity nor pacemaker current I(f) was observed. Lentiviral expression of TBX3 in ventricular cardiomyocytes resulted in conduction slowing and development of heterogeneous phenotypes, including depolarized and spontaneously active cardiomyocytes. CONCLUSIONS: TBX3 reprogrammes terminally differentiated working cardiomyocytes and induces important pacemaker properties. The ability of TBX3 to reduce intercellular coupling to overcome current-to-load mismatch and the ability to reduce I(K1) density to enable diastolic depolarization are promising TBX3 characteristics that may facilitate biological pacemaker formation strategies.


Asunto(s)
Relojes Biológicos/genética , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/genética
13.
Curr Top Dev Biol ; 100: 171-201, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22449844

RESUMEN

The heart is a structurally complex and functionally heterogeneous organ. The repertoire of genes active in a given cardiac cell defines its shapes and function. This process of localized or heterogeneous gene expression is regulated to a large extent at the level of transcription, dictating the degree particular genes in a cell are active. Therefore, errors in the regulation of localized gene expression are at the basis of misregulation of the delicate process of heart development and function. In this review, we provide an overview of the origin of the different components of the vertebrate heart, and discuss our current understanding of the regulation of localized gene expression in the developing heart. We will also discuss where future research may lead to gain more insight into this process, which should provide much needed insight into the dysregulation of heart development and function, and the etiology of congenital defects.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/crecimiento & desarrollo , Animales , Cromatina/genética , Humanos , Miocardio/citología , Miocardio/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...