Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Prog Biophys Mol Biol ; 138: 20-31, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30036562

RESUMEN

The molecular mechanism essential for the formation of heart valves involves complex interactions of signaling molecules and transcription factors. The Mediator Complex (MC) functions as multi-subunit machinery to orchestrate gene transcription, especially for tissue-specific fine-tuning of transcriptional processes during development, also in the heart. Here, we analyzed the role of the MC subunit Med12 during atrioventricular canal (AVC) development and endocardial cushion formation, using the Med12-deficient zebrafish mutant trapped (tpd). Whereas primary heart formation was only slightly affected in tpd, we identified defects in AVC development and cardiac jelly formation. We found that although misexpression of bmp4 and versican in tpd hearts can be restored by overexpression of a modified version of the Sox9b transcription factor (harboring VP16 transactivation domain) that functions independent of its co-activator Med12, endocardial cushion development in tpd was not reconstituted. Interestingly, expression of tbx2b and its target hyaluronan synthase 2 (has2) - the synthase of hyaluronan (HA) in the heart - was absent in both uninjected and Sox9b-VP16 overexpressing tpd hearts. HA is a major ECM component of the cardiac jelly and required for endocardial cushion formation. Furthermore, we found secreted phosphoprotein 1 (spp1), an endocardial marker of activated AV endocardial cells, completely absent in tpd hearts, suggesting that crucial steps of the transformation of AV endocardial cells into endocardial cushions is blocked. We demonstrate that Med12 controls cardiac jelly formation Sox9-independently by regulating tbx2b and has2 expression and therefore the production of the glycosaminoglycan HA at the AVC to guarantee proper endocardial cushion development.


Asunto(s)
Válvulas Cardíacas/crecimiento & desarrollo , Corazón/crecimiento & desarrollo , Complejo Mediador/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Animales , Endocardio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hialuronano Sintasas/metabolismo , Complejo Mediador/deficiencia , Complejo Mediador/genética , Mutación , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
2.
J Mol Cell Cardiol ; 120: 42-52, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29750993

RESUMEN

The genetic underpinnings that orchestrate the vertebrate heart rate are not fully understood yet, but of high clinical importance, since diseases of cardiac impulse formation and propagation are common and severe human arrhythmias. To identify novel regulators of the vertebrate heart rate, we deciphered the pathogenesis of the bradycardia in the homozygous zebrafish mutant hiphop (hip) and identified a missense-mutation (N851K) in Na+/K+-ATPase α1-subunit (atp1a1a.1). N851K affects zebrafish Na+/K+-ATPase ion transport capacity, as revealed by in vitro pump current measurements. Inhibition of the Na+/K+-ATPase in vivo indicates that hip rather acts as a hypomorph than being a null allele. Consequently, reduced Na+/K+-ATPase function leads to prolonged QT interval and refractoriness in the hip mutant heart, as shown by electrocardiogram and in vivo electrical stimulation experiments. We here demonstrate for the first time that Na+/K+-ATPase plays an essential role in heart rate regulation by prolonging myocardial repolarization.


Asunto(s)
Bradicardia/genética , Frecuencia Cardíaca/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Pez Cebra/genética , Potenciales de Acción , Alelos , Animales , Bloqueo Atrioventricular/genética , Estimulación Eléctrica , Electrocardiografía , Genes Modificadores , Células HEK293 , Humanos , Bombas Iónicas , Transporte Iónico , Mutación Missense , Miocitos Cardíacos/metabolismo , Polimorfismo de Nucleótido Simple , Estadísticas no Paramétricas
3.
PLoS One ; 11(12): e0167306, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27907103

RESUMEN

The molecular mechanisms that regulate cardiomyocyte proliferation during embryonic heart growth are not completely deciphered yet. In a forward genetic N-ethyl-N-nitrosourea (ENU) mutagenesis screen, we identified the recessive embryonic-lethal zebrafish mutant line weiches herz (whz). Homozygous mutant whz embryos display impaired heart growth due to diminished embryonic cardiomyocyte proliferation resulting in cardiac hypoplasia and weak cardiac contraction. By positional cloning, we found in whz mutant zebrafish a missense mutation within the T-box 20 (Tbx20) transcription factor gene leading to destabilization of Tbx20 protein. Morpholino-mediated knock-down of Tbx20 in wild-type zebrafish embryos phenocopies whz, indicating that the whz phenotype is due to loss of Tbx20 function, thereby leading to significantly reduced cardiomyocyte numbers by impaired proliferation of heart muscle cells. Ectopic overexpression of wild-type Tbx20 in whz mutant embryos restored cardiomyocyte proliferation and heart growth. Interestingly, ectopic overexpression of Tbx20 in wild-type zebrafish embryos resulted, similar to the situation in the embryonic mouse heart, in significantly reduced proliferation rates of ventricular cardiomyocytes, suggesting that Tbx20 activity needs to be tightly fine-tuned to guarantee regular cardiomyocyte proliferation and embryonic heart growth in vivo.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Organogénesis/genética , Proteínas de Dominio T Box/genética , Proteínas de Pez Cebra/genética , Animales , Proliferación Celular , Expresión Génica Ectópica , Técnicas de Silenciamiento del Gen , Mutación , Miocitos Cardíacos/metabolismo , Fenotipo , Pez Cebra/genética
4.
Biochem Biophys Res Commun ; 477(4): 581-588, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27343557

RESUMEN

In search for novel key regulators of cardiac valve formation, we isolated the zebrafish cardiac valve mutant ping pong (png). We find that an insertional promoter mutation within the zebrafish mediator complex subunit 10 (med10) gene is leading to impaired heart valve formation. Expression of the T-box transcription factor 2b (Tbx2b), known to be essential in cardiac valve development, is severely reduced in png mutant hearts. We demonstrate here that transient reconstitution of Tbx2b expression rescues AV canal development in png mutant zebrafish. By contrast, overexpression of Forkhead box N4 (Foxn4), a known upstream regulator of Tbx2b, is not capable to reconstitute tbx2b expression and heart valve formation in Med10-deficient png mutant hearts. Interestingly, hyaluronan synthase 2 (has2), a known downstream target of Tbx2 and producer of hyaluronan (HA) - a major ECM component of the cardiac jelly and critical for proper heart valve development - is completely absent in ping pong mutant hearts. We propose here a rather unique role of Med10 in orchestrating cardiac valve formation by mediating Foxn4 dependent tbx2b transcription, expression of Has2 and subsequently proper development of the cardiac jelly.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Válvulas Cardíacas/embriología , Complejo Mediador/fisiología , Proteínas de Dominio T Box/fisiología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Factores de Transcripción Forkhead/metabolismo , Válvulas Cardíacas/metabolismo , Hialuronano Sintasas , Mutación , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Pez Cebra/genética
5.
Basic Res Cardiol ; 111(3): 36, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27138930

RESUMEN

Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia with a strong genetic component. Molecular pathways involving the homeodomain transcription factor Shox2 control the development and function of the cardiac conduction system in mouse and zebrafish. Here we report the analysis of human SHOX2 as a potential susceptibility gene for early-onset AF. To identify causal variants and define the underlying mechanisms, results from 378 patients with early-onset AF before the age of 60 years were analyzed and compared to 1870 controls or reference datasets. We identified two missense mutations (p.G81E, p.H283Q), that were predicted as damaging. Transactivation studies using SHOX2 targets and phenotypic rescue experiments in zebrafish demonstrated that the p.H283Q mutation severely affects SHOX2 pacemaker function. We also demonstrate an association between a 3'UTR variant c.*28T>C of SHOX2 and AF (p = 0.00515). Patients carrying this variant present significantly longer PR intervals. Mechanistically, this variant creates a functional binding site for hsa-miR-92b-5p. Circulating hsa-miR-92b-5p plasma levels were significantly altered in AF patients carrying the 3'UTR variant (p = 0.0095). Finally, we demonstrate significantly reduced SHOX2 expression levels in right atrial appendages of AF patients compared to patients with sinus rhythm. Together, these results suggest a genetic contribution of SHOX2 in early-onset AF.


Asunto(s)
Fibrilación Atrial/genética , Predisposición Genética a la Enfermedad/genética , Proteínas de Homeodominio/genética , Adolescente , Animales , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación Missense , Reacción en Cadena de la Polimerasa , Transfección , Adulto Joven , Pez Cebra
6.
Sci Signal ; 8(369): ra30, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25805888

RESUMEN

Physiologically, Notch signal transduction plays a pivotal role in differentiation; pathologically, Notch signaling contributes to the development of cancer. Transcriptional activation of Notch target genes involves cleavage of the Notch receptor in response to ligand binding, production of the Notch intracellular domain (NICD), and NICD migration into the nucleus and assembly of a coactivator complex. Posttranslational modifications of the NICD are important for its transcriptional activity and protein turnover. Deregulation of Notch signaling and stabilizing mutations of Notch1 have been linked to leukemia development. We found that the methyltransferase CARM1 (coactivator-associated arginine methyltransferase 1; also known as PRMT4) methylated NICD at five conserved arginine residues within the C-terminal transactivation domain. CARM1 physically and functionally interacted with the NICD-coactivator complex and was found at gene enhancers in a Notch-dependent manner. Although a methylation-defective NICD mutant was biochemically more stable, this mutant was biologically less active as measured with Notch assays in embryos of Xenopus laevis and Danio rerio. Mathematical modeling indicated that full but short and transient Notch signaling required methylation of NICD.


Asunto(s)
Arginina/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Arginina/genética , Sitios de Unión/genética , Western Blotting , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Metilación , Ratones , Datos de Secuencia Molecular , Mutación , Proteína-Arginina N-Metiltransferasas/genética , Interferencia de ARN , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Activación Transcripcional , Xenopus laevis/embriología , Xenopus laevis/genética , Xenopus laevis/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
7.
Basic Res Cardiol ; 110(2): 14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25697682

RESUMEN

The genetic underpinnings of heart rate regulation are only poorly understood. In search for genetic regulators of cardiac pacemaker activity, we isolated in a large-scale mutagenesis screen the embryonic lethal, recessive zebrafish mutant schneckentempo (ste). Homozygous ste mutants exhibit a severely reduced resting heart rate with normal atrio-ventricular conduction and contractile function. External electrical pacing reveals that defective excitation generation in cardiac pacemaker cells underlies bradycardia in ste (-/-) mutants. By positional cloning and gene knock-down analysis we find that loss of dihydrolipoyl succinyltransferase (DLST) function causes the ste phenotype. The mitochondrial enzyme DLST is an essential player in the citric acid cycle that warrants proper adenosine-tri-phosphate (ATP) production. Accordingly, ATP levels are significantly diminished in ste (-/-) mutant embryos, suggesting that limited energy supply accounts for reduced cardiac pacemaker activity in ste (-/-) mutants. We demonstrate here for the first time that the mitochondrial enzyme DLST plays an essential role in the modulation of the vertebrate heart rate by controlling ATP production in the heart.


Asunto(s)
Aciltransferasas/metabolismo , Frecuencia Cardíaca/fisiología , Aciltransferasas/genética , Animales , Técnicas de Silenciamiento del Gen , Reacción en Cadena en Tiempo Real de la Polimerasa , Pez Cebra
8.
J Biol Chem ; 289(38): 26119-26130, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25104355

RESUMEN

G protein-coupled receptor kinases 2 (GRK2) and 5 (GRK5) are fundamental regulators of cardiac performance in adults but are less well characterized for their function in the hearts of embryos. GRK2 and -5 belong to different subfamilies and function as competitors in the control of certain receptors and signaling pathways. In this study, we used zebrafish to investigate whether the fish homologs of GRK2 and -5, Grk2/3 and Grk5, also have unique, complementary, or competitive roles during heart development. We found that they differentially regulate the heart rate of early embryos and equally facilitate heart function in older embryos and that both are required to develop proper cardiac morphology. A loss of Grk2/3 results in dilated atria and hypoplastic ventricles, and the hearts of embryos depleted in Grk5 present with a generalized atrophy. This Grk5 morphant phenotype was associated with an overall decrease of early cardiac progenitors as well as a reduction in the area occupied by myocardial progenitor cells. In the case of Grk2/3, the progenitor decrease was confined to a subset of precursor cells with a committed ventricular fate. We attempted to rescue the GRK loss-of-function heart phenotypes by downstream activation of Hedgehog signaling. The Grk2/3 loss-of-function embryos were rescued by this approach, but Grk5 embryos failed to respond. In summary, we found that GRK2 and GRK5 control cardiac function as well as morphogenesis during development although with different morphological outcomes.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Corazón/embriología , Proteínas de Pez Cebra/fisiología , Animales , Proliferación Celular , Células Madre Embrionarias/fisiología , Técnicas de Silenciamiento del Gen , Corazón/crecimiento & desarrollo , Frecuencia Cardíaca , Proteínas Hedgehog/metabolismo , Morfolinos/genética , Contracción Miocárdica , Miocardio/citología , Miocardio/enzimología , Neovascularización Fisiológica , Tamaño de los Órganos , Organogénesis , Transducción de Señal , Pez Cebra
9.
Basic Res Cardiol ; 108(2): 339, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23455426

RESUMEN

The heart's rhythm is initiated and regulated by a group of specialized cells in the sinoatrial node (SAN), the primary pacemaker of the heart. Abnormalities in the development of the SAN can result in irregular heart rates (arrhythmias). Although several of the critical genes important for SAN formation have been identified, our understanding of the transcriptional network controlling SAN development remains at a relatively early stage. The homeodomain transcription factor Shox2 is involved in the specification and patterning of the SAN. While the Shox2 knockout in mice results in embryonic lethality due to severe cardiac defects including improper SAN development, Shox2 knockdown in zebrafish causes a reduced heart rate (bradycardia). In order to gain deeper insight into molecular pathways involving Shox2, we compared gene expression levels in right atria of wildtype and Shox2 (-/-) hearts using microarray experiments and identified the LIM homeodomain transcription factor Islet1 (Isl1) as one of its putative target genes. The downregulation of Isl1 expression in Shox2 (-/-) hearts was confirmed and the affected region narrowed down to the SAN by whole-mount in situ hybridization. Using luciferase reporter assays and EMSA studies, we identified two specific SHOX2 binding sites within intron 2 of the ISL1 locus. We also provide functional evidence for Isl1 as a transcriptional target of Shox2 by rescuing the Shox2-mediated bradycardia phenotype with Isl1 using zebrafish as a model system. Our findings demonstrate a novel epistatic relationship between Shox2 and Isl1 in the heart with important developmental consequences for SAN formation and heart beat.


Asunto(s)
Bradicardia/genética , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Animales , Bradicardia/metabolismo , Bradicardia/fisiopatología , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Redes Reguladoras de Genes , Inmunohistoquímica , Hibridación in Situ , Ratones , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Nodo Sinoatrial/fisiología , Transcripción Genética , Pez Cebra
10.
Circ Res ; 111(12): 1504-16, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22972877

RESUMEN

RATIONALE: The emerging role of the ubiquitin-proteasome system in cardiomyocyte function and homeostasis implies the necessity of tight regulation of protein degradation. However, little is known about cardiac components of this machinery. OBJECTIVE: We sought to determine whether molecules exist that control turnover of cardiac-specific proteins. METHODS AND RESULTS: Using a bioinformatic approach to identify novel cardiac-enriched sarcomere proteins, we identified F-box and leucine-rich repeat protein 22 (Fbxl22). Tissue-specific expression was confirmed by multiple tissue Northern and Western Blot analyses as well as quantitative reverse-transcriptase polymerase chain reaction on a human cDNA library. Immunocolocalization experiments in neonatal and adult rat ventricular cardiomyocytes as well as murine heart tissue located Fbxl22 to the sarcomeric z-disc. To detect cardiac protein interaction partners, we performed a yeast 2-hybrid screen using Fbxl22 as bait. Coimmunoprecipitation confirmed the identified interactions of Fbxl22 with S-phase kinase-associated protein 1 and Cullin1, 2 critical components of SCF (Skp1/Cul1/F-box) E3- ligases. Moreover, we identified several potential substrates, including the z-disc proteins α-actinin and filamin C. Consistently, in vitro overexpression of Fbxl22-mediated degradation of both substrates in a dose-dependent fashion, whereas proteasome inhibition with MG-132 markedly attenuated degradation of both α-actinin and filamin C. Finally, targeted knockdown of Fbxl22 in rat cardiomyocytes as well as zebrafish embryos results in the accumulation of α-actinin associated with severely impaired contractile function and cardiomyopathy in vivo. CONCLUSIONS: These findings reveal the previously uncharacterized cardiac-specific F-box protein Fbxl22 as a component of a novel cardiac E3 ligase. Fbxl22 promotes the proteasome-dependent degradation of key sarcomeric proteins, such as α-actinin and filamin C, and is essential for maintenance of normal contractile function in vivo.


Asunto(s)
Proteínas F-Box/fisiología , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Sarcómeros/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Células Cultivadas , Células HEK293 , Humanos , Datos de Secuencia Molecular , Miocitos Cardíacos/fisiología , Transporte de Proteínas/fisiología , Ratas , Sarcómeros/fisiología
11.
J Cell Sci ; 124(Pt 18): 3127-36, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21852424

RESUMEN

Assembly, maintenance and renewal of sarcomeres require highly organized and balanced folding, transport, modification and degradation of sarcomeric proteins. However, the molecules that mediate these processes are largely unknown. Here, we isolated the zebrafish mutant flatline (fla), which shows disturbed sarcomere assembly exclusively in heart and fast-twitch skeletal muscle. By positional cloning we identified a nonsense mutation within the SET- and MYND-domain-containing protein 1 gene (smyd1) to be responsible for the fla phenotype. We found SMYD1 expression to be restricted to the heart and fast-twitch skeletal muscle cells. Within these cell types, SMYD1 localizes to both the sarcomeric M-line, where it physically associates with myosin, and the nucleus, where it supposedly represses transcription through its SET and MYND domains. However, although we found transcript levels of thick filament chaperones, such as Hsp90a1 and UNC-45b, to be severely upregulated in fla, its histone methyltransferase activity - mainly responsible for the nuclear function of SMYD1 - is dispensable for sarcomerogenesis. Accordingly, sarcomere assembly in fla mutant embryos can be reconstituted by ectopically expressing histone methyltransferase-deficient SMYD1. By contrast, ectopic expression of myosin-binding-deficient SMYD1 does not rescue fla mutants, implicating an essential role for the SMYD1-myosin interaction in cardiac and fast-twitch skeletal muscle thick filament assembly.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Músculo Esquelético/enzimología , Miocardio/enzimología , Miosinas/metabolismo , Sarcómeros/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Clonación Molecular , Citoesqueleto/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Análisis por Micromatrices , Contracción Muscular/fisiología , Músculo Esquelético/ultraestructura , Mutación/genética , Miocardio/ultraestructura , Unión Proteica , Sarcómeros/genética , Transgenes/genética , Pez Cebra , Proteínas de Pez Cebra/genética
12.
Circulation ; 124(3): 324-34, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730303

RESUMEN

BACKGROUND: The molecular mechanisms that guide heart valve formation are not well understood. However, elucidation of the genetic basis of congenital heart disease is one of the prerequisites for the development of tissue-engineered heart valves. METHODS AND RESULTS: We isolated here a mutation in zebrafish, bungee (bng(jh177)), which selectively perturbs valve formation in the embryonic heart by abrogating endocardial Notch signaling in cardiac cushions. We found by positional cloning that the bng phenotype is caused by a missense mutation (Y849N) in zebrafish protein kinase D2 (pkd2). The bng mutation selectively impairs PKD2 kinase activity and hence Histone deacetylase 5 phosphorylation, nuclear export, and inactivation. As a result, the expression of Histone deacetylase 5 target genes Krüppel-like factor 2a and 4a, transcription factors known to be pivotal for heart valve formation and to act upstream of Notch signaling, is severely downregulated in bungee (bng) mutant embryos. Accordingly, the expression of Notch target genes, such as Hey1, Hey2, and HeyL, is severely decreased in bng mutant embryos. Remarkably, downregulation of Histone deacetylase 5 activity in homozygous bng mutant embryos can rescue the mutant phenotype and reconstitutes notch1b expression in atrioventricular endocardial cells. CONCLUSIONS: We demonstrate for the first time that proper heart valve formation critically depends on Protein kinase D2-Histone deacetylase 5-Krüppel-like factor signaling.


Asunto(s)
Desarrollo Embrionario/fisiología , Válvulas Cardíacas/embriología , Histona Desacetilasas/fisiología , Proteínas Quinasas/fisiología , Pez Cebra/embriología , Animales , Embrión no Mamífero/fisiología , Desarrollo Embrionario/genética , Histona Desacetilasas/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Modelos Animales , Mutación Missense/genética , Proteína Quinasa D2 , Proteínas Quinasas/genética , Receptor Notch1/fisiología , Transducción de Señal/fisiología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
13.
Biochem Biophys Res Commun ; 408(2): 218-24, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21458413

RESUMEN

Inherited cardiac arrhythmias are caused by genetic defects in ion channels and associated proteins. Mutations in these channels often do not affect their biophysical properties, but rather interfere with their trafficking to the cell membrane. Accordingly, strategies that could reroute the mutated channels to the membrane should be sufficient to restore the electrical properties of the affected cells, thereby suppressing the underlying arrhythmia. We identified here both, embryonic and adult zebrafish breakdance (bre) as a valuable model for human Long-QT syndrome. Electrocardiograms of adult homozygous bre mutants exhibit significant QT prolongation caused by delayed repolarization of the ventricle. We further show that the bre mutation (zERG(I59S)) disrupts ERG protein trafficking, thereby reducing the amount of active potassium channels on the cell membrane. Interestingly, improvement of channel trafficking by cisapride or dimethylsulfoxid is sufficient to reconstitute ERG channels on the cell membrane in a manner that suffices to suppress the Long-QT induced arrhythmia in breakdance mutant zebrafish. In summary, we show for the first time that therapeutic intervention can cure protein trafficking defects and the associated cardiac arrhythmia in vivo.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Síndrome de QT Prolongado/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Modelos Animales de Enfermedad , Canales de Potasio Éter-A-Go-Go/genética , Células HEK293 , Frecuencia Cardíaca/genética , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Mutación , Transporte de Proteínas/genética , Disfunción Ventricular/genética , Disfunción Ventricular/fisiopatología , Pez Cebra , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...