Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 103: 48-55, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28077321

RESUMEN

Deletion of Ca2+/calmodulin-dependent protein kinase II delta (CaMKIIδ) has been shown to protect against in vivo ischemia/reperfusion (I/R) injury. It remains unclear which CaMKIIδ isoforms and downstream mechanisms are responsible for the salutary effects of CaMKIIδ gene deletion. In this study we sought to compare the roles of the CaMKIIδB and CaMKIIδC subtypes and the mechanisms by which they contribute to ex vivo I/R damage. WT, CaMKIIδKO, and mice expressing only CaMKIIδB or δC were subjected to ex vivo global ischemia for 25min followed by reperfusion. Infarct formation was assessed at 60min reperfusion by triphenyl tetrazolium chloride (TTC) staining. Deletion of CaMKIIδ conferred significant protection from ex vivo I/R. Re-expression of CaMKIIδC in the CaMKIIδKO background reversed this effect and exacerbated myocardial damage and dysfunction following I/R, while re-expression of CaMKIIδB was protective. Selective activation of CaMKIIδC in response to I/R was evident in a subcellular fraction enriched for cytosolic/membrane proteins. Further studies demonstrated differential regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling and tumor necrosis factor alpha (TNF-α) expression by CaMKIIδB and CaMKIIδC. Selective activation of CaMKIIδC was also observed and associated with NF-κB activation in neonatal rat ventricular myocytes (NRVMs) subjected to oxidative stress. Pharmacological inhibition of NF-κB or TNF-α significantly ameliorated infarct formation in WT mice and those that re-express CaMKIIδC, demonstrating distinct roles for CaMKIIδ subtypes in I/R and implicating acute activation of CaMKIIδC and NF-κB in the pathogenesis of reperfusion injury.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Biopsia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Modelos Animales de Enfermedad , Ecocardiografía , Técnicas de Inactivación de Genes , Ratones , Ratones Transgénicos , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/etiología , Infarto del Miocardio/mortalidad , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/mortalidad , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Disfunción Ventricular
2.
J Mol Cell Cardiol ; 103: 1-10, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28017639

RESUMEN

Sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, is generated and released at sites of tissue injury in the heart and can act on S1P1, S1P2, and S1P3 receptor subtypes to affect cardiovascular responses. We established that S1P causes little phosphoinositide hydrolysis and does not induce hypertrophy indicating that it does not cause receptor coupling to Gq. We previously demonstrated that S1P confers cardioprotection against ischemia/reperfusion by activating RhoA and its downstream effector PKD. The S1P receptor subtypes and G proteins that regulate RhoA activation and downstream responses in the heart have not been determined. Using siRNA or pertussis toxin to inhibit different G proteins in NRVMs we established that S1P regulates RhoA activation through Gα13 but not Gα12, Gαq, or Gαi. Knockdown of the three major S1P receptors using siRNA demonstrated a requirement for S1P3 in RhoA activation and subsequent phosphorylation of PKD, and this was confirmed in studies using isolated hearts from S1P3 knockout (KO) mice. S1P treatment reduced infarct size induced by ischemia/reperfusion in Langendorff perfused wild-type (WT) hearts and this protection was abolished in the S1P3 KO mouse heart. CYM-51736, an S1P3-specific agonist, also decreased infarct size after ischemia/reperfusion to a degree similar to that achieved by S1P. The finding that S1P3 receptor- and Gα13-mediated RhoA activation is responsible for protection against ischemia/reperfusion suggests that selective targeting of S1P3 receptors could provide therapeutic benefits in ischemic heart disease.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proproteína Convertasas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Serina Endopeptidasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Lisofosfolípidos/metabolismo , Masculino , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Unión Proteica , Ratas , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Canales Catiónicos TRPP/metabolismo
3.
J Biol Chem ; 291(8): 4156-65, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26742842

RESUMEN

The fast transient outward potassium current (Ito,f) plays a critical role in the electrical and contractile properties of the myocardium. Ito,f channels are formed by the co-assembly of the pore-forming α-subunits, Kv4.2 and Kv4.3, together with the accessory ß-subunit KChIP2. Reductions of Ito,f are common in the diseased heart, which is also associated with enhanced stimulation of ß-adrenergic receptors (ß-ARs). We used cultured neonatal rat ventricular myocytes to examine how chronic ß-AR stimulation decreases Ito,f. To determine which downstream pathways mediate these Ito,f changes, adenoviral infections were used to inhibit CaMKIIδc, CaMKIIδb, calcineurin, or nuclear factor κB (NF-κB). We observed that chronic ß-AR stimulation with isoproterenol (ISO) for 48 h reduced Ito,f along with mRNA expression of all three of its subunits (Kv4.2, Kv4.3, and KChIP2). Inhibiting either CaMKIIδc nor CaMKIIδb did not prevent the ISO-mediated Ito,f reductions, even though CaMKIIδc and CaMKIIδb clearly regulated Ito,f and the mRNA expression of its subunits. Likewise, calcineurin inhibition did not prevent the Ito,f reductions induced by ß-AR stimulation despite strongly modulating Ito,f and subunit mRNA expression. In contrast, NF-κB inhibition partly rescued the ISO-mediated Ito,f reductions in association with restoration of KChIP2 mRNA expression. Consistent with these observations, KChIP2 promoter activity was reduced by p65 as well as ß-AR stimulation. In conclusion, NF-κB, and not CaMKIIδ or calcineurin, partly mediates the Ito,f reductions induced by chronic ß-AR stimulation. Both mRNA and KChIP2 promoter data suggest that the ISO-induced Ito,f reductions are, in part, mediated through reduced KChIP2 transcription caused by NF-κB activation.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Isoproterenol/farmacología , Proteínas de Interacción con los Canales Kv/metabolismo , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Calcineurina/genética , Calcineurina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Interacción con los Canales Kv/genética , FN-kappa B/genética , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/metabolismo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo
4.
J Mol Cell Cardiol ; 85: 282-91, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26080362

RESUMEN

Chronic activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has been implicated in the deleterious effects of ß-adrenergic receptor (ß-AR) signaling on the heart, in part, by enhancing RyR2-mediated sarcoplasmic reticulum (SR) Ca(2+) leak. We used CaMKIIδ knockout (CaMKIIδ-KO) mice and knock-in mice with an inactivated CaMKII site S2814 on the ryanodine receptor type 2 (S2814A) to investigate the involvement of these processes in ß-AR signaling and cardiac remodeling. Langendorff-perfused hearts from CaMKIIδ-KO mice showed inotropic and chronotropic responses to isoproterenol (ISO) that were similar to those of wild type (WT) mice; however, in CaMKIIδ-KO mice, CaMKII phosphorylation of phospholamban and RyR2 was decreased and isolated myocytes from CaMKIIδ-KO mice had reduced SR Ca(2+) leak in response to isoproterenol (ISO). Chronic catecholamine stress with ISO induced comparable increases in relative heart weight and other measures of hypertrophy from day 9 through week 4 in WT and CaMKIIδ-KO mice, but the development of cardiac fibrosis was prevented in CaMKIIδ-KO animals. A 4-week challenge with ISO resulted in reduced cardiac function and pulmonary congestion in WT, but not in CaMKIIδ-KO or S2814A mice, implicating CaMKIIδ-dependent phosphorylation of RyR2-S2814 in the cardiomyopathy, independent of hypertrophy, induced by prolonged ß-AR stimulation.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Procesamiento Proteico-Postraduccional , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio , Cardiomegalia/enzimología , Cardiomiopatías/enzimología , Células Cultivadas , Fibrosis , Isoproterenol/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Fosforilación , Retículo Sarcoplasmático/metabolismo , Remodelación Ventricular
5.
Circ Res ; 116(5): e28-39, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25605649

RESUMEN

RATIONALE: Sustained activation of Gαq transgenic (Gq) signaling during pressure overload causes cardiac hypertrophy that ultimately progresses to dilated cardiomyopathy. The molecular events that drive hypertrophy decompensation are incompletely understood. Ca(2+)/calmodulin-dependent protein kinase II δ (CaMKIIδ) is activated downstream of Gq, and overexpression of Gq and CaMKIIδ recapitulates hypertrophy decompensation. OBJECTIVE: To determine whether CaMKIIδ contributes to hypertrophy decompensation provoked by Gq. METHODS AND RESULTS: Compared with Gq mice, compound Gq/CaMKIIδ knockout mice developed a similar degree of cardiac hypertrophy but exhibited significantly improved left ventricular function, less cardiac fibrosis and cardiomyocyte apoptosis, and fewer ventricular arrhythmias. Markers of oxidative stress were elevated in mitochondria from Gq versus wild-type mice and respiratory rates were lower; these changes in mitochondrial function were restored by CaMKIIδ deletion. Gq-mediated increases in mitochondrial oxidative stress, compromised membrane potential, and cell death were recapitulated in neonatal rat ventricular myocytes infected with constitutively active Gq and attenuated by CaMKII inhibition. Deep RNA sequencing revealed altered expression of 41 mitochondrial genes in Gq hearts, with normalization of ≈40% of these genes by CaMKIIδ deletion. Uncoupling protein 3 was markedly downregulated in Gq or by Gq expression in neonatal rat ventricular myocytes and reversed by CaMKIIδ deletion or inhibition, as was peroxisome proliferator-activated receptor α. The protective effects of CaMKIIδ inhibition on reactive oxygen species generation and cell death were abrogated by knock down of uncoupling protein 3. Conversely, restoration of uncoupling protein 3 expression attenuated reactive oxygen species generation and cell death induced by CaMKIIδ. Our in vivo studies further demonstrated that pressure overload induced decreases in peroxisome proliferator-activated receptor α and uncoupling protein 3, increases in mitochondrial protein oxidation, and hypertrophy decompensation, which were attenuated by CaMKIIδ deletion. CONCLUSIONS: Mitochondrial gene reprogramming induced by CaMKIIδ emerges as an important mechanism contributing to mitotoxicity in decompensating hypertrophy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Cardiomegalia/enzimología , Cardiomiopatía Dilatada/etiología , Insuficiencia Cardíaca/etiología , Mitocondrias Cardíacas/fisiología , Acetilcisteína/farmacología , Animales , Apoptosis , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/deficiencia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Cardiomegalia/fisiopatología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Dilatada/prevención & control , Células Cultivadas , Progresión de la Enfermedad , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Canales Iónicos/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , PPAR alfa/biosíntesis , PPAR alfa/genética , Mutación Puntual , Presión , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Ratas , Especies Reactivas de Oxígeno , Análisis de Secuencia de ARN , Sulfonamidas/farmacología , Transfección , Proteína Desacopladora 3
6.
Front Pharmacol ; 5: 15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24575042

RESUMEN

In this review we discuss the localization and function of the known subtypes of calcium/calmodulin dependent protein kinase IIδ (CaMKIIδ) and their role in cardiac physiology and pathophysiology. The CaMKII holoenzyme is comprised of multiple subunits that are encoded by four different genes called CaMKIIα, ß, γ, and δ. While these four genes have a high degree of sequence homology, they are expressed in different tissues. CaMKIIα and ß are expressed in neuronal tissue while γ and δ are present throughout the body, including in the heart. Both CaMKIIγ and δ are alternatively spliced in the heart to generate multiple subtypes. CaMKIIδ is the predominant cardiac isoform and is alternatively spliced in the heart to generate the CaMKIIδB subtype or the slightly less abundant δC subtype. The CaMKIIδB mRNA sequence contains a 33bp insert not present in δC that codes for an 11-amino acid nuclear localization sequence. This review focuses on the localization and function of the CaMKIIδ subtypes δB and δC and the role of these subtypes in arrhythmias, contractile dysfunction, gene transcription, and the regulation of Ca(2+) handling.

7.
Circ Res ; 112(6): 935-44, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23388157

RESUMEN

RATIONALE: Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has been implicated as a maladaptive mediator of cardiac ischemic injury. We hypothesized that the inflammatory response associated with in vivo ischemia/reperfusion (I/R) is initiated through CaMKII signaling. OBJECTIVE: To assess the contribution of CaMKIIδ to the development of inflammation, infarct, and ventricular dysfunction after in vivo I/R and define early cardiomyocyte-autonomous events regulated by CaMKIIδ using cardiac-specific knockout mice. METHODS AND RESULTS: Wild-type and CaMKIIδ knockout mice were subjected to in vivo I/R by occlusion of the left anterior descending artery for 1 hour followed by reperfusion for various times. CaMKIIδ deletion protected the heart against I/R damage as evidenced by decreased infarct size, attenuated apoptosis, and improved functional recovery. CaMKIIδ deletion also attenuated I/R-induced inflammation and upregulation of nuclear factor-κB (NF-κB) target genes. Further studies demonstrated that I/R rapidly increases CaMKII activity, leading to NF-κB activation within minutes of reperfusion. Experiments using cyclosporine A and cardiac-specific CaMKIIδ knockout mice indicate that NF-κB activation is initiated independent of necrosis and within cardiomyocytes. Expression of activated CaMKII in cardiomyocytes leads to IκB kinase phosphorylation and concomitant increases in nuclear p65. Experiments using an IκB kinase inhibitor support the conclusion that this is a proximal site of CaMKII-mediated NF-κB activation. CONCLUSIONS: This is the first study demonstrating that CaMKIIδ mediates NF-κB activation in cardiomyocytes after in vivo I/R and suggests that CaMKIIδ serves to trigger, as well as to sustain subsequent changes in inflammatory gene expression that contribute to myocardial I/R damage.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Daño por Reperfusión Miocárdica/etiología , FN-kappa B/metabolismo , Animales , Apoptosis/fisiología , Ciclosporina/farmacología , Perfilación de la Expresión Génica , Corazón , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Fosforilación , Recuperación de la Función/fisiología , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba
8.
Circ Res ; 109(12): 1354-62, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21998325

RESUMEN

RATIONALE: Differential effects of δ(B) and δ(C) subtypes of Ca²âº/calmodulin-dependent protein kinase (CaMKII) on cardiomyocyte Ca²âº handling and survival have been suggested to result from their respective nuclear versus cytosolic localizations. CaMKIIδ subtype localization and its relationship to enzyme activation and target phosphorylation have not, however, been systematically evaluated. OBJECTIVE: To determine whether CaMKIIδ subtypes are restricted to a particular subcellular location and assess the relationship of localization to enzyme activation and function. METHODS AND RESULTS: CaMKIIδ is highly expressed in mouse heart and cardiomyocytes and concentrated in sarcoplasmic reticulum (SR)/membrane and nuclear fractions. CaMKIIδ(B) and δ(C) subtypes differ by a nuclear localization sequence, but both are present in nuclear and SR/membrane fractions. Nonselective subtype distribution is also seen in mice overexpressing CaMKIIδ(B) or δ(C), even in a CaMKIIδ null background. Fluorescently tagged CaMKIIδ(B) expressed in cardiomyocytes concentrates in nuclei whereas δ(C) concentrates in cytosol, but neither localization is exclusive. Mouse hearts exposed to phenylephrine show selective CaMKIIδ activation in the nuclear (versus SR) compartment, whereas caffeine selectively activates CaMKIIδ in SR (versus nuclei), independent of subtype. Compartmentalized activation extends to functional differences in target phosphorylation at CaMKII sites: phenylephrine increases histone deacetylase 5 phosphorylation (Ser498) but not phospholamban (Thr17), whereas the converse holds for caffeine. CONCLUSIONS: These studies demonstrate that CaMKIIδ(B) and δ(C) are not exclusively restricted to the nucleus and cytosol and that spatial and functional specificity in CaMKIIδ activation is elicited by mobilization of different Ca²âº stores rather than by compartmentalized subtype localization.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/enzimología , Citoplasma/enzimología , Miocitos Cardíacos/metabolismo , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Cardiotónicos/farmacología , Núcleo Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Ratones , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Fenilefrina/farmacología , Fosforilación , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/enzimología
9.
J Neurosci ; 29(34): 10764-78, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19710327

RESUMEN

Dravet syndrome (also called severe myoclonic epilepsy of infancy) is one of the most severe forms of childhood epilepsy. Most patients have heterozygous mutations in SCN1A, encoding voltage-gated sodium channel Na(v)1.1 alpha subunits. Sodium channels are modulated by beta1 subunits, encoded by SCN1B, a gene also linked to epilepsy. Here we report the first patient with Dravet syndrome associated with a recessive mutation in SCN1B (p.R125C). Biochemical characterization of p.R125C in a heterologous system demonstrated little to no cell surface expression despite normal total cellular expression. This occurred regardless of coexpression of Na(v)1.1 alpha subunits. Because the patient was homozygous for the mutation, these data suggest a functional SCN1B null phenotype. To understand the consequences of the lack of beta1 cell surface expression in vivo, hippocampal slice recordings were performed in Scn1b(-/-) versus Scn1b(+/+) mice. Scn1b(-/-) CA3 neurons fired evoked action potentials with a significantly higher peak voltage and significantly greater amplitude compared with wild type. However, in contrast to the Scn1a(+/-) model of Dravet syndrome, we found no measurable differences in sodium current density in acutely dissociated CA3 hippocampal neurons. Whereas Scn1b(-/-) mice seize spontaneously, the seizure susceptibility of Scn1b(+/-) mice was similar to wild type, suggesting that, like the parents of this patient, one functional SCN1B allele is sufficient for normal control of electrical excitability. We conclude that SCN1B p.R125C is an autosomal recessive cause of Dravet syndrome through functional gene inactivation.


Asunto(s)
Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/fisiopatología , Polimorfismo de Nucleótido Simple/genética , Canales de Sodio/genética , Animales , Arginina/genética , Biofisica , Línea Celular Transformada , Cisteína/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Estimulación Eléctrica , Epilepsias Mioclónicas/mortalidad , Femenino , Proteínas Fluorescentes Verdes/genética , Hipocampo/patología , Humanos , Técnicas In Vitro , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/deficiencia , Oocitos , Canales de Sodio/deficiencia , Temperatura , Transfección , Gemelos , Subunidad beta-1 de Canal de Sodio Activado por Voltaje , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...