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 ; 106(1): 43-54, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25661081

RESUMEN

AIMS: Foetal growth has been proposed to influence cardiovascular health in adulthood, a process referred to as foetal programming. Indeed, intrauterine growth restriction in animal models alters heart size and cardiomyocyte number in the perinatal period, yet the consequences for the adult or challenged heart are largely unknown. The aim of this study was to elucidate postnatal myocardial growth pattern, left ventricular function, and stress response in the adult heart after neonatal cardiac hypoplasia in mice. METHODS AND RESULTS: Utilizing a new mouse model of impaired cardiac development leading to fully functional but hypoplastic hearts at birth, we show that myocardial mass is normalized until early adulthood by accelerated physiological cardiomyocyte hypertrophy. Compensatory hypertrophy, however, cannot be maintained upon ageing, resulting in reduced organ size without maladaptive myocardial remodelling. Angiotensin II stress revealed aberrant cardiomyocyte growth kinetics in adult hearts after neonatal hypoplasia compared with normally developed controls, characterized by reversible overshooting hypertrophy. This exaggerated growth mainly depends on STAT3, whose inhibition during angiotensin II treatment reduces left ventricular mass in both groups but causes contractile dysfunction in developmentally impaired hearts only. Whereas JAK/STAT3 inhibition reduces cardiomyocyte cross-sectional area in the latter, it prevents fibrosis in control hearts, indicating fundamentally different mechanisms of action. CONCLUSION: Impaired prenatal development leading to neonatal cardiac hypoplasia alters postnatal cardiac growth and stress response in vivo, thereby linking foetal programming to organ size control in the heart.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Desarrollo Embrionario/fisiología , Desarrollo Fetal/fisiología , Corazón/embriología , Corazón/fisiopatología , Estrés Fisiológico/fisiología , Envejecimiento/fisiología , Angiotensina II/farmacología , Animales , Femenino , Corazón/efectos de los fármacos , Hipertrofia , Liasas/deficiencia , Liasas/genética , Liasas/fisiología , Ratones , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Tamaño de los Órganos/fisiología , Factor de Transcripción STAT3/fisiología
2.
Hypertension ; 64(5): 1032-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25185127

RESUMEN

Angiotensin (Ang) II is a potent mediator of both hypertension and cardiac damage; however, the mechanisms by which this occur remain unclear. B-cell lymphoma/leukemia 10 (Bcl10) is a member of the CBM signalosome, which links Ang II and nuclear factor-κB signaling. We hypothesized that Bcl10 is pivotal in the pathogenesis of Ang II-induced cardiac damage. Ang II infusion in mice lacking Bcl10 resulted in reduced cardiac fibrosis, less cellular infiltration, and improved arrhythmogenic electric remodeling, despite a similar degree of hypertension or cardiac hypertrophy. Adoptive transfer of bone marrow (BM), whereby Bcl10 knockout or wildtype BM was transferred to their opposite genotype recipients, revealed the dual importance of Bcl10 within both cardiac and immune cells. Loss of Bcl10 in cardiac cells resulted in reduced expression of genes important for the adhesion and recruitment of immune cells. In vitro experiments demonstrated that adhesion of monocytes to Ang II-treated endothelial cells also required Bcl10. Additionally, Bcl10 deficiency in macrophages reduced their intrinsic migratory ability. To address the role of BM-derived fibroblasts in the formation of cardiac fibrosis, we explored whether Bcl10 is also important for the infiltration of BM-derived (myo)fibroblasts into the heart. The transfer of green fluorescent protein positive wildtype BM into Bcl10 knockout recipient mice revealed a reduced number of noncardiac (myo)fibroblasts compared with those wildtype recipients. Our results demonstrate the significant role of Bcl10 in multiple cell types important for the generation of Ang II-induced cardiac damage and electric remodeling and may provide a new avenue for therapeutic intervention.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Angiotensina II/efectos adversos , Remodelación Atrial/fisiología , Cardiopatías/inducido químicamente , Cardiopatías/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 10 de la LLC-Linfoma de Células B , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Fibrosis , Cardiopatías/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Miocardio/metabolismo , Miocardio/patología , FN-kappa B/metabolismo
3.
J Am Coll Cardiol ; 63(6): 549-59, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24291282

RESUMEN

OBJECTIVES: The aim of this study was to investigate the modulatory effect of the coxsackie and adenovirus receptor (CAR) on ventricular conduction and arrhythmia vulnerability in the setting of myocardial ischemia. BACKGROUND: A heritable component in the risk of ventricular fibrillation during myocardial infarction has been well established. A recent genome-wide association study of ventricular fibrillation during acute myocardial infarction led to the identification of a locus on chromosome 21q21 (rs2824292) in the vicinity of the CXADR gene. CXADR encodes the CAR, a cell adhesion molecule predominantly located at the intercalated disks of the cardiomyocyte. METHODS: The correlation between CAR transcript levels and rs2824292 genotype was investigated in human left ventricular samples. Electrophysiological studies and molecular analyses were performed using CAR haploinsufficient (CAR⁺/⁻) mice. RESULTS: In human left ventricular samples, the risk allele at the chr21q21 genome-wide association study locus was associated with lower CXADR messenger ribonucleic acid levels, suggesting that decreased cardiac levels of CAR predispose to ischemia-induced ventricular fibrillation. Hearts from CAR⁺/⁻ mice displayed slowing of ventricular conduction in addition to an earlier onset of ventricular arrhythmias during the early phase of acute myocardial ischemia after ligation of the left anterior descending artery. Expression and distribution of connexin 43 were unaffected, but CAR⁺/⁻ hearts displayed increased arrhythmia susceptibility on pharmacological electrical uncoupling. Patch-clamp analysis of isolated CAR⁺/⁻ myocytes showed reduced sodium current magnitude specifically at the intercalated disk. Moreover, CAR coprecipitated with NaV1.5 in vitro, suggesting that CAR affects sodium channel function through a physical interaction with NaV1.5. CONCLUSIONS: CAR is a novel modifier of ventricular conduction and arrhythmia vulnerability in the setting of myocardial ischemia. Genetic determinants of arrhythmia susceptibility (such as CAR) may constitute future targets for risk stratification of potentially lethal ventricular arrhythmias in patients with coronary artery disease.


Asunto(s)
Arritmias Cardíacas/etiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/fisiología , Sistema de Conducción Cardíaco/fisiopatología , Isquemia Miocárdica/metabolismo , Función Ventricular , Animales , Carbenoxolona , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/fisiopatología , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo
4.
Hypertension ; 62(5): 966-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24001898

RESUMEN

Alzheimer disease features amyloid-ß (Aß) peptide deposition in brain and blood vessels and is associated with hypertension. Aß peptide can cause vasoconstriction and endothelial dysfunction. We observed that Aß peptides exert a chronotropic effect in neonatal cardiomyocytes, similar to α1-adrenergic receptor autoantibodies that we described earlier. Recently, it was shown that α1-adrenergic receptor could impair blood-brain flow. We hypothesized that Aß peptides might elicit a signal transduction pathway in vascular cells, induced by α1-adrenergic receptor activation. Aß (25-35) and Aß (10-35) induced a positive chronotropic effect in the cardiac contraction assay (28.75±1.15 and 29.40±0.98 bpm), which was attenuated by α1-adrenergic receptor blockers (urapidil, 1.53±1.17 bpm; prazosin, 0.30±0.96 bpm). Both Aß peptides induced an intracellular calcium release in vascular smooth muscle cells. Chronotropic activity and calcium response elicited by Aß (25-35) were blocked with peptides corresponding to the first extracellular loop of the α1-adrenergic receptor. We observed an induction of extracellular-regulated kinase 1/2 phosphorylation by Aß (25-35) in Chinese hamster ovary cells overexpressing α1-adrenergic receptor, vascular smooth muscle cells, and cardiomyocytes. We generated an activation-state-sensitive α1-adrenergic receptor antibody and visualized activation of the α1-adrenergic receptor by Aß peptide. Aß (25-35) induced vasoconstriction of mouse aortic rings and in coronary arteries in Langendorff-perfused rat hearts that resulted in decreased coronary flow. Both effects could be reversed by α1-adrenergic receptor blockade. Our data are relevant to the association between Alzheimer disease and hypertension. They may explain impairment of vascular responses by Aß and could have therapeutic implications.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Péptidos beta-Amiloides/farmacología , Miocitos Cardíacos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Corazón/efectos de los fármacos , Masculino , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
PLoS One ; 8(8): e73490, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023684

RESUMEN

Maladaptive cardiac hypertrophy predisposes one to arrhythmia and sudden death. Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) promote anti-inflammatory and antiapoptotic mechanisms, and are involved in the regulation of cardiac Ca(2+)-, K(+)- and Na(+)-channels. To test the hypothesis that enhanced cardiac EET biosynthesis counteracts hypertrophy-induced electrical remodeling, male transgenic mice with cardiomyocyte-specific overexpression of the human epoxygenase CYP2J2 (CYP2J2-TG) and wildtype littermates (WT) were subjected to chronic pressure overload (transverse aortic constriction, TAC) or ß-adrenergic stimulation (isoproterenol infusion, ISO). TAC caused progressive mortality that was higher in WT (42% over 8 weeks after TAC), compared to CYP2J2-TG mice (6%). In vivo electrophysiological studies, 4 weeks after TAC, revealed high ventricular tachyarrhythmia inducibility in WT (47% of the stimulation protocols), but not in CYP2J2-TG mice (0%). CYP2J2 overexpression also enhanced ventricular refractoriness and protected against TAC-induced QRS prolongation and delocalization of left ventricular connexin-43. ISO for 14 days induced high vulnerability for atrial fibrillation in WT mice (54%) that was reduced in CYP-TG mice (17%). CYP2J2 overexpression also protected against ISO-induced reduction of atrial refractoriness and development of atrial fibrosis. In contrast to these profound effects on electrical remodeling, CYP2J2 overexpression only moderately reduced TAC-induced cardiac hypertrophy and did not affect the hypertrophic response to ß-adrenergic stimulation. These results demonstrate that enhanced cardiac EET biosynthesis protects against electrical remodeling, ventricular tachyarrhythmia, and atrial fibrillation susceptibility during maladaptive cardiac hypertrophy.


Asunto(s)
Arritmias Cardíacas/etiología , Arritmias Cardíacas/prevención & control , Cardiomegalia/complicaciones , Cardiomegalia/enzimología , Sistema Enzimático del Citocromo P-450/metabolismo , Animales , Estenosis de la Válvula Aórtica/complicaciones , Estenosis de la Válvula Aórtica/enzimología , Estenosis de la Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/fisiopatología , Arritmias Cardíacas/enzimología , Arritmias Cardíacas/fisiopatología , Biomarcadores/metabolismo , Cardiomegalia/fisiopatología , Enfermedad Crónica , Conexina 43/metabolismo , Citocromo P-450 CYP2J2 , Susceptibilidad a Enfermedades/complicaciones , Susceptibilidad a Enfermedades/enzimología , Susceptibilidad a Enfermedades/patología , Susceptibilidad a Enfermedades/fisiopatología , Fenómenos Electrofisiológicos , Fibrosis Endomiocárdica/complicaciones , Fibrosis Endomiocárdica/enzimología , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/fisiopatología , Humanos , Masculino , Ratones , Ratones Transgénicos , Presión , Receptores Adrenérgicos beta/metabolismo , Análisis de Supervivencia , Remodelación Ventricular
6.
Hypertension ; 60(6): 1430-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23108651

RESUMEN

Angiotensin (Ang) II induces vascular injury in part by activating innate and adaptive immunity; however, the mechanisms are unclear. We investigated the role of interferon (IFN)-γ and interleukin (IL)-23 signaling. We infused Ang II into IFN-γ receptor (IFN-γR) knockout mice and wild-type controls, as well as into mice treated with neutralizing antibodies against IL-23 receptor and IL-17A. Ang II-treated IFN-γR knockout mice exhibited reduced cardiac hypertrophy, reduced cardiac macrophage and T-cell infiltration, less fibrosis, and less arrhythmogenic electric remodeling independent of blood pressure changes. In contrast, IL-23 receptor antibody treatment did not reduce cardiac hypertrophy, fibrosis, or electric remodeling despite mildly reduced inflammation. IL-17A antibody treatment behaved similarly. In the kidney, IFN-γR deficiency reduced inflammation and tubulointerstitial damage and improved glomerular filtration rate. Nonetheless, albuminuria was increased compared with Ang II-treated wild-type controls. The glomeruli of Ang II-treated IFN-γR knockout mice exhibited fewer podocytes, less nephrin and synaptopodin staining, and impaired podocyte autophagy. Thus, IFN-γ blockade, but not IL-23 receptor antibody treatment, protects from Ang II-induced cardiac damage and electric remodeling. In the kidney, IFN-γ signaling acts in a cell type-specific manner. Glomerular filtration rate is preserved in the absence of the IFN-γR, whereas podocytes may require the IFN-γR in the presence of Ang II for normal integrity and function.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/metabolismo , Corazón/efectos de los fármacos , Interferón gamma/metabolismo , Miocardio/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Fibrosis , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-23/genética , Interleucina-23/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Miocardio/patología , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Podocitos/patología , Transducción de Señal/efectos de los fármacos
7.
Nat Med ; 18(5): 766-73, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22466703

RESUMEN

Alternative splicing has a major role in cardiac adaptive responses, as exemplified by the isoform switch of the sarcomeric protein titin, which adjusts ventricular filling. By positional cloning using a previously characterized rat strain with altered titin mRNA splicing, we identified a loss-of-function mutation in the gene encoding RNA binding motif protein 20 (Rbm20) as the underlying cause of pathological titin isoform expression. The phenotype of Rbm20-deficient rats resembled the pathology seen in individuals with dilated cardiomyopathy caused by RBM20 mutations. Deep sequencing of the human and rat cardiac transcriptome revealed an RBM20-dependent regulation of alternative splicing. In addition to titin (TTN), we identified a set of 30 genes with conserved splicing regulation between humans and rats. This network is enriched for genes that have previously been linked to cardiomyopathy, ion homeostasis and sarcomere biology. Our studies emphasize the key role of post-transcriptional regulation in cardiac function and provide mechanistic insights into the pathogenesis of human heart failure.


Asunto(s)
Cardiomiopatía Dilatada/genética , Proteínas Musculares/genética , Proteínas Quinasas/genética , Empalme del ARN , Proteínas de Unión al ARN/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Secuencia de Bases , Conectina , Humanos , Proteínas con Dominio LIM/genética , Datos de Secuencia Molecular , Mutación , Proteínas de Unión al ARN/fisiología , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...