Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cardiovasc Res ; 114(8): 1082-1097, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522173

RESUMEN

Aims: Arrhythmogenic cardiomyopathy (AC) is an inherited heart disease characterized by life-threatening ventricular arrhythmias and fibro-fatty replacement of the myocardium. More than 60% of AC patients show pathogenic mutations in genes encoding for desmosomal proteins. By focusing our attention on the AC8 form, linked to the junctional protein desmoplakin (DSP), we present here a zebrafish model of DSP deficiency, exploited to identify early changes of cell signalling in the cardiac region. Methods and results: To obtain an embryonic model of Dsp deficiency, we first confirmed the orthologous correspondence of zebrafish Dsp genes (dspa and dspb) to the human DSP counterpart. Then, we verified their cardiac expression, at embryonic and adult stages, and subsequently we targeted them by antisense morpholino strategy, confirming specific and disruptive effects on desmosomes, like those identified in AC patients. Finally, we exploited our Dsp-deficient models for an in vivo cell signalling screen, using pathway-specific reporter transgenes. Out of nine considered, three pathways (Wnt/ß-catenin, TGFß/Smad3, and Hippo/YAP-TAZ) were significantly altered, with Wnt as the most dramatically affected. Interestingly, under persistent Dsp deficiency, Wnt signalling is rescuable both by a genetic and a pharmacological approach. Conclusion: Our data point to Wnt/ß-catenin as the final common pathway underlying different desmosomal AC forms and support the zebrafish as a suitable model for detecting early signalling pathways involved in the pathogenesis of DSP-associated diseases, possibly responsive to pharmacological or genetic rescue.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/metabolismo , Desmoplaquinas/metabolismo , Miocardio/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/patología , Desmoplaquinas/deficiencia , Desmoplaquinas/genética , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Indoles/farmacología , Maleimidas/farmacología , Morfogénesis , Miocardio/ultraestructura , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
2.
Cardiovasc Res ; 111(3): 274-86, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27097650

RESUMEN

AIMS: Current mechanisms driving cardiac pacemaker function have focused on ion channel and gap junction channel function, which are essential for action potential generation and propagation between pacemaker cells. However, pacemaker cells also harbour desmosomes that structurally anchor pacemaker cells to each other in tissue, but their role in pacemaker function remains unknown. METHODS AND RESULTS: To determine the role of desmosomes in pacemaker function, we generated a novel mouse model harbouring cardiac conduction-specific ablation (csKO) of the central desmosomal protein, desmoplakin (DSP) using the Hcn4-Cre-ERT2 mouse line. Hcn4-Cre targets cells of the adult mouse sinoatrial node (SAN) and can ablate DSP expression in the adult DSP csKO SAN resulting in specific loss of desmosomal proteins and structures. Dysregulation of DSP via loss-of-function (adult DSP csKO mice) and mutation (clinical case of a patient harbouring a pathogenic DSP variant) in mice and man, respectively, revealed that desmosomal dysregulation is associated with a primary phenotype of increased sinus pauses/dysfunction in the absence of cardiomyopathy. Underlying defects in beat-to-beat regulation were also observed in DSP csKO mice in vivo and intact atria ex vivo. DSP csKO SAN exhibited migrating lead pacemaker sites associated with connexin 45 loss. In vitro studies exploiting ventricular cardiomyocytes that harbour DSP loss and concurrent early connexin loss phenocopied the loss of beat-to-beat regulation observed in DSP csKO mice and atria, extending the importance of DSP-associated mechanisms in driving beat-to-beat regulation of working cardiomyocytes. CONCLUSION: We provide evidence of a mechanism that implicates an essential role for desmosomes in cardiac pacemaker function, which has broad implications in better understanding mechanisms underlying beat-to-beat regulation as well as sinus node disease and dysfunction.


Asunto(s)
Relojes Biológicos , Desmosomas , Frecuencia Cardíaca , Síndrome del Seno Enfermo/fisiopatología , Nodo Sinoatrial/fisiopatología , Potenciales de Acción , Factores de Edad , Animales , Función Atrial , Células Cultivadas , Conexinas/metabolismo , Desmoplaquinas/deficiencia , Desmoplaquinas/genética , Desmosomas/metabolismo , Desmosomas/ultraestructura , Predisposición Genética a la Enfermedad , Humanos , Ratones Noqueados , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Fenotipo , Síndrome del Seno Enfermo/genética , Síndrome del Seno Enfermo/metabolismo , Síndrome del Seno Enfermo/patología , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/ultraestructura , Factores de Tiempo
3.
Hum Mol Genet ; 23(5): 1134-50, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24108106

RESUMEN

Arrhythmogenic right ventricular cardiomyopathy (ARVC) termed a 'disease of the desmosome' is an inherited cardiomyopathy that recently underwent reclassification owing to the identification of left-dominant and biventricular disease forms. Homozygous loss-of-function mutations in the desmosomal component, desmoplakin, are found in patients exhibiting a biventricular form of ARVC; however, no models recapitulate the postnatal hallmarks of the disease as seen in these patients. To gain insights into the homozygous loss-of-function effects of desmoplakin in the heart, we generated cardiomyocyte-specific desmoplakin-deficient mice (DSP-cKO) using ventricular myosin light chain-2-Cre mice. Homozygous DSP-cKO mice are viable but display early ultrastructural defects in desmosomal integrity leading to a cardiomyopathy reminiscent of a biventricular form of ARVC, which includes cell death and fibro-fatty replacement within the ventricle leading to biventricular dysfunction, failure and premature death. DSP-cKO mice also exhibited ventricular arrhythmias that are exacerbated with exercise and catecholamine stimulation. Furthermore, DSP-cKO hearts exhibited right ventricular conduction defects associated with loss of connexin 40 expression and electrical wavefront propagation defects associated with loss of connexin 43 expression. Dose-dependent assessment of the effects of loss of desmoplakin in neonatal ventricular cardiomyocytes revealed primary loss of connexin 43 levels, phosphorylation and function independent of the molecular dissociation of the mechanical junction complex and fibro-fatty manifestation associated with ARVC, suggesting a role for desmoplakin as a primary stabilizer of connexin integrity. In summary, we provide evidence for a novel mouse model, which is reminiscent of the postnatal onset of ARVC while highlighting mechanisms underlying a biventricular form of human ARVC.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Conexinas/deficiencia , Animales , Animales Recién Nacidos , Arritmias Cardíacas/genética , Displasia Ventricular Derecha Arritmogénica/diagnóstico , Displasia Ventricular Derecha Arritmogénica/mortalidad , Síndrome de Brugada , Trastorno del Sistema de Conducción Cardíaco , Catecolaminas/farmacología , Conexina 43/deficiencia , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Desmoplaquinas/deficiencia , Modelos Animales de Enfermedad , Electrocardiografía , Expresión Génica , Corazón/efectos de los fármacos , Sistema de Conducción Cardíaco/anomalías , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Fosforilación , Condicionamiento Físico Animal/efectos adversos , Proteína alfa-5 de Unión Comunicante
4.
Eur Heart J ; 33(15): 1942-53, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22240500

RESUMEN

AIMS: Anecdotal observations suggest that sub-clinical electrophysiological manifestations of arrhythmogenic right ventricular cardiomyopathy (ARVC) develop before detectable structural changes ensue on cardiac imaging. To test this hypothesis, we investigated a murine model with conditional cardiac genetic deletion of one desmoplakin allele (DSP ±) and compared the findings to patients with non-diagnostic features of ARVC who carried mutations in desmoplakin. METHODS AND RESULTS: Murine: the DSP (±) mice underwent electrophysiological, echocardiographic, and immunohistochemical studies. They had normal echocardiograms but delayed conduction and inducible ventricular tachycardia associated with mislocalization and reduced intercalated disc expression of Cx43. Sodium current density and myocardial histology were normal at 2 months of age. Human: ten patients with heterozygous mutations in DSP without overt structural heart disease (DSP+) and 12 controls with supraventricular tachycardia were studied by high-density electrophysiological mapping of the right ventricle. Using a standard S(1)-S(2) protocol, restitution curves of local conduction and repolarization parameters were constructed. Significantly greater mean increases in delay were identified particularly in the outflow tract vs. controls (P< 0.01) coupled with more uniform wavefront progression. The odds of a segment with a maximal activation-repolarization interval restitution slope >1 was 99% higher (95% CI: 13%; 351%, P = 0.017) in DSP+ vs. controls. Immunostaining revealed Cx43 mislocalization and variable Na channel distribution. CONCLUSION: Desmoplakin disease causes connexin mislocalization in the mouse and man preceding any overt histological abnormalities resulting in significant alterations in conduction-repolarization kinetics prior to morphological changes detectable on conventional cardiac imaging. Haploinsufficiency of desmoplakin is sufficient to cause significant Cx43 mislocalization. Changes in sodium current density and histological abnormalities may contribute to a worsening phenotype or disease but are not necessary to generate an arrhythmogenic substrate. This has important implications for the earlier diagnosis of ARVC and risk stratification.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Desmoplaquinas/genética , Mutación/genética , Adulto , Anciano , Animales , Estudios de Casos y Controles , Desmoplaquinas/deficiencia , Electrocardiografía , Femenino , Eliminación de Gen , Sistema de Conducción Cardíaco/fisiología , Heterocigoto , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Sodio/fisiología , Adulto Joven
5.
J Cell Biol ; 194(4): 631-42, 2011 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-21844209

RESUMEN

Desmosomes are cell-cell adhesion structures that integrate cytoskeletal networks. In addition to binding intermediate filaments, the desmosomal protein desmoplakin (DP) regulates microtubule reorganization in the epidermis. In this paper, we identify a specific subset of centrosomal proteins that are recruited to the cell cortex by DP upon epidermal differentiation. These include Lis1 and Ndel1, which are centrosomal proteins that regulate microtubule organization and anchoring in other cell types. This recruitment was mediated by a region of DP specific to a single isoform, DPI. Furthermore, we demonstrate that the epidermal-specific loss of Lis1 results in dramatic defects in microtubule reorganization. Lis1 ablation also causes desmosomal defects, characterized by decreased levels of desmosomal components, decreased attachment of keratin filaments, and increased turnover of desmosomal proteins at the cell cortex. This contributes to loss of epidermal barrier activity, resulting in completely penetrant perinatal lethality. This work reveals essential desmosome-associated components that control cortical microtubule organization and unexpected roles for centrosomal proteins in epidermal function.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Desmosomas/metabolismo , Epidermis/metabolismo , Queratinocitos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/deficiencia , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Desmoplaquinas/deficiencia , Desmoplaquinas/genética , Epidermis/embriología , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Permeabilidad , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Transfección , alfa Catenina/deficiencia , alfa Catenina/genética
6.
Circ Res ; 104(9): 1076-84, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19359597

RESUMEN

The phenotypic hallmark of arrhythmogenic right ventricular cardiomyopathy, a genetic disease of desmosomal proteins, is fibroadipocytic replacement of the right ventricle. Cellular origin of excess adipocytes, the responsible mechanism(s) and the basis for predominant involvement of the right ventricle are unknown. We generated 3 sets of lineage tracer mice regulated by cardiac lineage promoters alpha-myosin heavy chain (alphaMyHC), Nkx2.5, or Mef2C. We conditionally expressed the reporter enhanced yellow fluorescent protein while concomitantly deleting the desmosomal protein desmoplakin in cardiac myocyte lineages using the Cre-LoxP technique. Lineage tracer mice showed excess fibroadiposis and increased numbers of adipocytes in the hearts. Few adipocytes in the hearts of alphaMyHC-regulated lineage tracer mice, but the majority of adipocytes in the hearts of Nkx2.5- and Mef2C-regulated lineage tracer mice, expressed enhanced yellow fluorescent protein. In addition, rare cells coexpressed adipogenic transcription factors and the second heart field markers Isl1 and Mef2C in the lineage tracer mouse hearts and in human myocardium from patients with arrhythmogenic right ventricular cardiomyopathy. To delineate the responsible mechanism, we generated transgenic mice expressing desmosomal protein plakoglobin in myocyte lineages. Transgene plakoglobin translocated to nucleus, detected by immunoblotting and immunofluorescence staining and coimmunoprecipitated with Tcf7l2, a canonical Wnt signaling transcription factor. Expression levels of canonical Wnt/Tcf7l2 targets bone morphogenetic protein 7 and Wnt5b, which promote adipogenesis, were increased and expression level of connective tissue growth factor, an inhibitor of adipogenesis, was decreased. We conclude adipocytes in arrhythmogenic right ventricular cardiomyopathy originate from the second heart field cardiac progenitors, which switch to an adipogenic fate because of suppressed canonical Wnt signaling by nuclear plakoglobin.


Asunto(s)
Adipocitos/patología , Adipogénesis/genética , Displasia Ventricular Derecha Arritmogénica/genética , Linaje de la Célula/genética , Miocardio/patología , Células Madre/patología , Adipocitos/metabolismo , Adulto , Animales , Displasia Ventricular Derecha Arritmogénica/metabolismo , Displasia Ventricular Derecha Arritmogénica/patología , Proteína Morfogenética Ósea 7/metabolismo , Desmoplaquinas/deficiencia , Modelos Animales de Enfermedad , Ecocardiografía Doppler , Fibrosis , Genotipo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Transducción de Señal , Células Madre/metabolismo , Factores de Transcripción TCF/metabolismo , Proteína 2 Similar al Factor de Transcripción 7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , gamma Catenina/genética , gamma Catenina/metabolismo
7.
J Med Genet ; 43(2): e5, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16467215

RESUMEN

BACKGROUND: Desmosomes are cellular junctions important for intercellular adhesion and anchoring the intermediate filament (IF) cytoskeleton to the cell membrane. Desmoplakin (DSP) is the most abundant desmosomal protein with 2 isoforms produced by alternative splicing. METHODS: We describe a patient with a recessively inherited arrhythmogenic dilated cardiomyopathy with left and right ventricular involvement, epidermolytic palmoplantar keratoderma, and woolly hair. The patient showed a severe heart phenotype with an early onset and rapid progression to heart failure at 4 years of age. RESULTS: A homozygous nonsense mutation, R1267X, was found in exon 23 of the desmoplakin gene, which results in an isoform specific truncation of the larger DSPI isoform. The loss of most of the DSPI specific rod domain and C-terminal area was confirmed by Western blotting and immunofluorescence. We further showed that the truncated DSPI transcript is unstable, leading to a loss of DSPI. DSPI is reported to be an obligate constituent of desmosomes and the only isoform present in cardiac tissue. To address this, we reviewed the expression of DSP isoforms in the heart. Our data suggest that DSPI is the major cardiac isoform but we also show that specific compartments of the heart have detectable DSPII expression. CONCLUSIONS: This is the first description of a phenotype caused by a mutation affecting only one DSP isoform. Our findings emphasise the importance of desmoplakin and desmosomes in epidermal and cardiac function and additionally highlight the possibility that the different isoforms of desmoplakin may have distinct functional properties within the desmosome.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Desmoplaquinas/deficiencia , Desmoplaquinas/genética , Edad de Inicio , Cardiomiopatías/epidemiología , Preescolar , Análisis Mutacional de ADN , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Haplotipos/genética , Humanos , Masculino , Miocardio/metabolismo , Linaje , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Piel/metabolismo , Síndrome , gamma Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...