Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Gen Physiol ; 152(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32761048

RESUMEN

Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ influx mechanism triggered by depletion of Ca2+ stores from the endoplasmic/sarcoplasmic reticulum (ER/SR). We recently reported that acute exercise in WT mice drives the formation of Ca2+ entry units (CEUs), intracellular junctions that contain STIM1 and Orai1, the two key proteins mediating SOCE. The presence of CEUs correlates with increased constitutive- and store-operated Ca2+ entry, as well as sustained Ca2+ release and force generation during repetitive stimulation. Skeletal muscle from mice lacking calsequestrin-1 (CASQ1-null), the primary Ca2+-binding protein in the lumen of SR terminal cisternae, exhibits significantly reduced total Ca2+ store content and marked SR Ca2+ depletion during high-frequency stimulation. Here, we report that CEUs are constitutively assembled in extensor digitorum longus (EDL) and flexor digitorum brevis (FDB) muscles of sedentary CASQ1-null mice. The higher density of CEUs in EDL (39.6 ± 2.1/100 µm2 versus 2.0 ± 0.3/100 µm2) and FDB (16.7 ± 1.0/100 µm2 versus 2.7 ± 0.5/100 µm2) muscles of CASQ1-null compared with WT mice correlated with enhanced constitutive- and store-operated Ca2+ entry and increased expression of STIM1, Orai1, and SERCA. The higher ability to recover Ca2+ ions via SOCE in CASQ1-null muscle served to promote enhanced maintenance of peak Ca2+ transient amplitude, increased dependence of luminal SR Ca2+ replenishment on BTP-2-sensitive SOCE, and increased maintenance of contractile force during repetitive, high-frequency stimulation. Together, these data suggest that muscles from CASQ1-null mice compensate for the lack of CASQ1 and reduction in total releasable SR Ca2+ content by assembling CEUs to promote constitutive and store-operated Ca2+ entry.


Asunto(s)
Calcio , Calsecuestrina , Músculo Esquelético , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio , Calsecuestrina/fisiología , Iones , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/fisiología , Proteína ORAI1 , Molécula de Interacción Estromal 1
2.
Int J Mol Sci ; 21(15)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32751833

RESUMEN

: Experimental evidence highlights the involvement of the endoplasmic reticulum (ER)-mediated Ca2+ signals in modulating synaptic plasticity and spatial memory formation in the hippocampus. Ca2+ release from the ER mainly occurs through two classes of Ca2+ channels, inositol 1,4,5-trisphosphate receptors (InsP3Rs) and ryanodine receptors (RyRs). Calsequestrin (CASQ) and calreticulin (CR) are the most abundant Ca2+-binding proteins allowing ER Ca2+ storage. The hippocampus is one of the brain regions expressing CASQ, but its role in neuronal activity, plasticity, and the learning processes is poorly investigated. Here, we used knockout mice lacking both CASQ type-1 and type-2 isoforms (double (d)CASQ-null mice) to: a) evaluate in adulthood the neuronal electrophysiological properties and synaptic plasticity in the hippocampal Cornu Ammonis 1 (CA1) field and b) study the performance of knockout mice in spatial learning tasks. The ablation of CASQ increased the CA1 neuron excitability and improved the long-term potentiation (LTP) maintenance. Consistently, (d)CASQ-null mice performed significantly better than controls in the Morris Water Maze task, needing a shorter time to develop a spatial preference for the goal. The Ca2+ handling analysis in CA1 pyramidal cells showed a decrement of Ca2+ transient amplitude in (d)CASQ-null mouse neurons, which is consistent with a decrease in afterhyperpolarization improving LTP. Altogether, our findings suggest that CASQ deletion affects activity-dependent ER Ca2+ release, thus facilitating synaptic plasticity and spatial learning in post-natal development.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Proteínas de Unión al Calcio/fisiología , Calsecuestrina/fisiología , Plasticidad Neuronal , Aprendizaje Espacial , Animales , Región CA1 Hipocampal/citología , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Calsecuestrina/genética , Retículo Endoplásmico/metabolismo , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Piramidales/citología , Células Piramidales/metabolismo
3.
Drug Alcohol Depend ; 133(2): 344-51, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23876860

RESUMEN

BACKGROUND: Cocaine-related deaths are continuously rising and its overdose is often associated with lethal cardiotoxic effects. METHODS AND RESULTS: Our approach, employing isothermal titration calorimetry (ITC) and light scattering in parallel, has confirmed the significant affinity of human cardiac calsequestrin (CASQ2) for cocaine. Calsequestrin (CASQ) is a major Ca(2+)-storage protein within the sarcoplasmic reticulum (SR) of both cardiac and skeletal muscles. CASQ acts as a Ca(2+) buffer and Ca(2+)-channel regulator through its unique Ca(2+)-dependent oligomerization. Equilibrium dialysis and atomic absorption spectroscopy experiments illustrated the perturbational effect of cocaine on CASQ2 polymerization, resulting in substantial reduction of its Ca(2+)-binding capacity. We also confirmed the accumulation of cocaine in rat heart tissue and the substantial effects cocaine has on cultured C2C12 cells. The same experiments were performed with methamphetamine as a control, which displayed neither affinity for CASQ2 nor any significant effects on its function. Since cocaine did not have any direct effect on the Ca(2+)-release channel judging from our single channel recordings, these studies provide new insights into how cocaine may interfere with the normal E-C coupling mechanism with lethal arrhythmogenic consequences. CONCLUSION: We propose that cocaine accumulates in SR through its affinity for CASQ2 and affects both SR Ca(2+) storage and release by altering the normal CASQ2 Ca(2+)-dependent polymerization. By this mechanism, cocaine use could produce serious cardiac problems, especially in people who have genetically-impaired CASQ2, defects in other E-C coupling components, or compromised cocaine metabolism and clearance.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Calsecuestrina/fisiología , Cocaína/efectos adversos , Corazón/fisiopatología , Animales , Arritmias Cardíacas/fisiopatología , Canales de Calcio/fisiología , Calorimetría , Calsecuestrina/metabolismo , Línea Celular , Cocaína/metabolismo , Diálisis , Luz , Ratones , Modelos Moleculares , Peso Molecular , Miocardio/citología , Miocardio/metabolismo , Unión Proteica , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Retículo Sarcoplasmático/metabolismo , Dispersión de Radiación , Espectrofotometría Atómica
4.
Proc Natl Acad Sci U S A ; 110(25): 10312-7, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23733959

RESUMEN

Dysregulated intracellular Ca(2+) signaling is implicated in a variety of cardiac arrhythmias, including catecholaminergic polymorphic ventricular tachycardia. Spontaneous diastolic Ca(2+) release (DCR) can induce arrhythmogenic plasma membrane depolarizations, although the mechanism responsible for DCR synchronization among adjacent myocytes required for ectopic activity remains unclear. We investigated the synchronization mechanism(s) of DCR underlying untimely action potentials and diastolic contractions (DCs) in a catecholaminergic polymorphic ventricular tachycardia mouse model with a mutation in cardiac calsequestrin. We used a combination of different approaches including single ryanodine receptor channel recording, optical imaging (Ca(2+) and membrane potential), and contractile force measurements in ventricular myocytes and intact cardiac muscles. We demonstrate that DCR occurs in a temporally and spatially uniform manner in both myocytes and intact myocardial tissue isolated from cardiac calsequestrin mutation mice. Such synchronized DCR events give rise to triggered electrical activity that results in synchronous DCs in the myocardium. Importantly, we establish that synchronization of DCR is a result of a combination of abbreviated ryanodine receptor channel refractoriness and the preceding synchronous stimulated Ca(2+) release/reuptake dynamics. Our study reveals how aberrant DCR events can become synchronized in the intact myocardium, leading to triggered activity and the resultant DCs in the settings of a cardiac rhythm disorder.


Asunto(s)
Señalización del Calcio/fisiología , Calsecuestrina/genética , Corazón/fisiología , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/fisiopatología , Animales , Calcio/metabolismo , Calsecuestrina/fisiología , Diástole/fisiología , Modelos Animales de Enfermedad , Ventrículos Cardíacos/citología , Masculino , Ratones , Ratones Mutantes , Mutación , Miocitos Cardíacos/fisiología , Músculos Papilares/citología , Músculos Papilares/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/fisiología , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo
6.
Cardiovasc Res ; 98(2): 169-76, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23400762

RESUMEN

This review highlights recent and ongoing discoveries that are transforming the previously held view of dyad structure and function. New data show that dyads vary greatly in both structure and in their associated molecules. Dyads can contain varying numbers of type 2 ryanodine receptor (RYR2) clusters that range in size from one to hundreds of tetramers and they can adopt numerous orientations other than the expected checkerboard. The association of Ca(v)1.2 with RYR2, which defines the couplon, is not absolute, leading to a number of scenarios such as dyads without couplons and those in which only a fraction of the clusters are in couplons. Different dyads also vary in the transporters and exchangers with which they are associated producing functional differences that amplify their structural diversity. The essential role of proteins, such as junctophilin-2, calsequestrin, triadin, and junctin that maintain both the functional and structural integrity of the dyad have recently been elucidated giving a new mechanistic understanding of heart diseases, such as arrhythmias, hypertension, failure, and sudden cardiac death.


Asunto(s)
Miocitos Cardíacos/ultraestructura , Sarcolema/ultraestructura , Retículo Sarcoplasmático/ultraestructura , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Calsecuestrina/fisiología , Acoplamiento Excitación-Contracción , Humanos , Proteínas de la Membrana/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Intercambiador de Sodio-Calcio/fisiología
7.
Cardiovasc Res ; 98(2): 240-7, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23408344

RESUMEN

The cardiac ryanodine receptor (RyR2), a Ca(2+) release channel on the membrane of the sarcoplasmic reticulum (SR), plays a key role in determining the strength of the heartbeat by supplying Ca(2+) required for contractile activation. Abnormal RyR2 function is recognized as an important part of the pathophysiology of heart failure (HF). While in the normal heart, the balance between the cytosolic and intra-SR Ca(2+) regulation of RyR2 function maintains the contraction-relaxation cycle, in HF, this behaviour is compromised by excessive post-translational modifications of the RyR2. Such modification of the Ca(2+) release channel impairs the ability of the RyR2 to properly deactivate leading to a spectrum of Ca(2+)-dependent pathologies that include cardiac systolic and diastolic dysfunction, arrhythmias, and structural remodelling. In this article, we present an overview of recent advances in our understanding of the underlying causes and pathological consequences of abnormal RyR2 function in the failing heart. We also discuss the implications of these findings for HF therapy.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Calsecuestrina/fisiología , Acoplamiento Excitación-Contracción , Humanos , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología
8.
Circ Res ; 112(4): 689-97, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23295832

RESUMEN

RATIONALE: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by mutations in cardiac ryanodine receptor (RyR2) or calsequestrin (Casq2) genes. Sinoatrial node dysfunction associated with CPVT may increase the risk for ventricular arrhythmia (VA). OBJECTIVE: To test the hypothesis that CPVT is suppressed by supraventricular overdrive stimulation. METHODS AND RESULTS: Using CPVT mouse models (Casq2(-/-) and RyR2(R4496C/+) mice), the effect of increasing sinus heart rate was tested by pretreatment with atropine and by atrial overdrive pacing. Increasing intrinsic sinus rate with atropine before catecholamine challenge suppressed ventricular tachycardia in 86% of Casq2(-/-) mice (6/7) and significantly reduced the VA score (atropine: 0.6±0.2 versus vehicle: 1.7±0.3; P<0.05). Atrial overdrive pacing completely prevented VA in 16 of 19 (84%) Casq2(-/-) and in 7 of 8 (88%) RyR2(R4496C/+) mice and significantly reduced ventricular premature beats in both CPVT models (P<0.05). Rapid pacing also prevented spontaneous calcium waves and triggered beats in isolated CPVT myocytes. In humans, heart rate dependence of CPVT was evaluated by screening a CPVT patient registry for antiarrhythmic drug-naïve individuals that reached >85% of their maximum-predicted heart rate during exercise testing. All 18 CPVT patients who fulfilled the inclusion criteria exhibited VA before reaching 87% of maximum heart rate. In 6 CPVT patients (33%), VA were paradoxically suppressed as sinus heart rates increased further with continued exercise. CONCLUSIONS: Accelerated supraventricular rates suppress VAs in 2 CPVT mouse models and in a subset of CPVT patients. Hypothetically, atrial overdrive pacing may be a therapy for preventing exercise-induced ventricular tachycardia in treatment-refractory CPVT patients.


Asunto(s)
Frecuencia Cardíaca , Adulto , Animales , Atropina/farmacología , Atropina/uso terapéutico , Bradicardia/genética , Bradicardia/fisiopatología , Cafeína/toxicidad , Señalización del Calcio/fisiología , Calsecuestrina/deficiencia , Calsecuestrina/genética , Calsecuestrina/fisiología , Estimulación Cardíaca Artificial , Prueba de Esfuerzo , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Isoproterenol/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Distribución Aleatoria , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Nodo Sinoatrial/fisiopatología , Simpatectomía Química , Taquicardia Ventricular , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiopatología , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/prevención & control
9.
Cardiovasc Res ; 98(2): 297-306, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23135969

RESUMEN

AIMS: Although aberrant Ca(2+) release (i.e. Ca(2+) 'leak') from the sarcoplasmic reticulum (SR) through cardiac ryanodine receptors (RyR2) is linked to heart failure (HF), it remains unknown whether and under what conditions SR-derived Ca(2+) can actually cause HF. We tested the hypothesis that combining dysregulated RyR2 function with facilitated Ca(2+) uptake into SR will exacerbate abnormal SR Ca(2+) release and induce HF. We also examined the mechanisms for these alterations. METHODS AND RESULTS: We crossbred mice deficient in expression of cardiac calsequestrin (CASQ2) with mice overexpressing the skeletal muscle isoform of SR Ca(2+)ATPase (SERCA1a). The new double-mutant strains displayed early mortality, congestive HF with left ventricular dilated hypertrophy, and decreased ejection fraction. Intact right ventricular muscle preparations from double-mutant mice preserved normal systolic contractile force but were susceptible to spontaneous contractions. Double-mutant cardiomyocytes while preserving normal amplitude of systolic Ca(2+) transients displayed marked disturbances in diastolic Ca(2+) handling in the form of multiple, periodic Ca(2+) waves and wavelets. Dysregulated myocyte Ca(2+) handling and structural and functional cardiac pathology in double-mutant mice were associated with increased rate of apoptotic cell death. Qualitatively similar results were obtained in a hybrid strain created by crossing CASQ2 knockout mice with mice deficient in phospholamban. CONCLUSION: We demonstrate that enhanced SR Ca(2+) uptake combined with dysregulated RyR2s results in sustained diastolic Ca(2+) release causing apoptosis, dilated cardiomyopathy, and early mortality. Our data also suggest that up-regulation of SERCA activity must be advocated with caution as a therapy for HF in the context of abnormal RyR2 function.


Asunto(s)
Calcio/metabolismo , Calsecuestrina/fisiología , Cardiomegalia/etiología , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Apoptosis , Arritmias Cardíacas/etiología , Calsecuestrina/deficiencia , Cardiomegalia/metabolismo , Diástole , Insuficiencia Cardíaca/etiología , Masculino , Ratones , Miocitos Cardíacos/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Regulación hacia Arriba , Remodelación Ventricular
10.
J Physiol ; 590(15): 3575-83, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22687611

RESUMEN

Dietary inorganic nitrate has profound effects on health and physiological responses to exercise. Here, we examined if nitrate, in doses readily achievable via a normal diet, could improve Ca(2+) handling and contractile function using fast- and slow-twitch skeletal muscles from C57bl/6 male mice given 1 mm sodium nitrate in water for 7 days. Age matched controls were provided water without added nitrate. In fast-twitch muscle fibres dissected from nitrate treated mice, myoplasmic free [Ca(2+)] was significantly greater than in Control fibres at stimulation frequencies from 20 to 150 Hz, which resulted in a major increase in contractile force at ≤ 50 Hz. At 100 Hz stimulation, the rate of force development was ∼35% faster in the nitrate group. These changes in nitrate treated mice were accompanied by increased expression of the Ca(2+) handling proteins calsequestrin 1 and the dihydropyridine receptor. No changes in force or calsequestrin 1 and dihydropyridine receptor expression were measured in slow-twitch muscles. In conclusion, these results show a striking effect of nitrate supplementation on intracellular Ca(2+) handling in fast-twitch muscle resulting in increased force production. A new mechanism is revealed by which nitrate can exert effects on muscle function with applications to performance and a potential therapeutic role in conditions with muscle weakness.


Asunto(s)
Calcio/fisiología , Contracción Muscular/efectos de los fármacos , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Nitratos/administración & dosificación , Animales , Canales de Calcio Tipo L/fisiología , Proteínas de Unión al Calcio/fisiología , Calsecuestrina/fisiología , Dieta , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares de Contracción Rápida/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología
11.
Wien Med Wochenschr ; 162(13-14): 292-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22733365

RESUMEN

Calcium is an important mediator in cardiac excitation and disorders in cardiac Ca(2+) homeostasis have great influence on the cardiac action potential. Therefore dysfunction in regulatory proteins that are involved in Ca(2+) handling can lead to the occurrence of severe arrhythmia. Although mutations in Ca(2+) regulating proteins are quite rare, they can offer general insights into arrhythmogenesis. Here, we briefly review some important aspects of arrhythmia-associated mutations in Ca(2+) regulating proteins with special emphasis to its associated pathophysiology.


Asunto(s)
Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/fisiología , Calcio/metabolismo , Mutación , Trastorno Autístico , Síndrome de Brugada/genética , Síndrome de Brugada/fisiopatología , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/fisiología , Calsecuestrina/genética , Calsecuestrina/fisiología , Citosol/fisiología , Diástole/fisiología , Electrocardiografía , Femenino , Homeostasis/genética , Homeostasis/fisiología , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Masculino , Miofibrillas/fisiología , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/fisiología , Sindactilia/genética , Sindactilia/fisiopatología , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatología
12.
J Thromb Haemost ; 10(1): 116-24, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22060633

RESUMEN

BACKGROUND: Altered Ca(2+) homeostasis contributes significantly to platelet hyperactivity in diabetes mellitus. Calsequestrin (CSQ), as a Ca(2+) buffer protein in the sarcoplasmic reticulum, also regulates the Ca(2+) release process in muscles. We hypothesized that CSQ may be expressed in platelets, but is altered and involved in diabetic platelet Ca(2+) abnormalities and hyperaggregability. METHODS: CSQ expression in platelets from streptozotocin-induced type 1 diabetes rats, type 2 diabetes volunteers and Goto-Kakizaki rats were analyzed by western blotting and RT-qPCR. Platelet Ca(2+) and aggregation were evaluated with Fura2 and an aggregometer, respectively. RESULTS: Platelets from diabetic patients and rats exhibited increased resting Ca(2+) levels, and hyperactive Ca(2+) and aggregation responses to agonists. This enhanced basal Ca(2+) was largely dependent on intracellular Ca(2+) and insensitive to inositol 1,4,5-trisphosphate receptor (IP(3)R) antagonism. Additionally, the expression of the skeletal CSQ isotype (CSQ-1) was detected in both rat and human platelets, but its levels were significantly lowered in diabetic platelets as compared with normal platelets. Impairment of CSQ by trifluoperazine caused concentration-dependent Ca(2+) release in normal platelets and HEK293 cells. Knocking down CSQ-1 in HEK293 cells resulted in increased leakage of Ca(2+), which was also insensitive to IP(3)R inhibition, and exaggerated Ca(2+) release following carbachol treatment. CONCLUSIONS: Downregulation of CSQ-1 in diabetic platelets and impairment of CSQ-1 in normal cells leads to disturbed Ca(2+) release, demonstrating a potential role for CSQ-1 in the regulation of the platelet Ca(2+) release process and a possible causal contribution to diabetic platelet hyperactivity.


Asunto(s)
Plaquetas/metabolismo , Calcio/metabolismo , Calsecuestrina/fisiología , Diabetes Mellitus/sangre , Homeostasis , Animales , Regulación hacia Abajo , Humanos , Activación Plaquetaria , Ratas
13.
Am J Physiol Heart Circ Physiol ; 301(4): H1706-15, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21803942

RESUMEN

Calcium flux into and out of the sarco(endo)plasmic reticulum is vitally important to cardiac function because the cycle of calcium entry and exit controls contraction and relaxation. Putative estrogen and androgen consensus binding sites near to a CpG island are present in the cardiac calsequestrin 2 (CSQ2) promoter. Cardiomyocytes express sex hormone receptors and respond to sex hormones. We hypothesized that sex hormones control CSQ2 expression in cardiomyocytes and so affect cardiac structure/function. Echocardiographic analysis of male and female C57bl6n mice identified thinner walled and lighter hearts in females and significant concentric remodeling after long-term gonadectomy. CSQ2 and sodium-calcium exchanger-1 (NCX1) expression was significantly increased in female compared with male hearts and decreased postovariectomy. NCX1, but not CSQ2, expression was increased postcastration. CSQ2 expression was reduced when H9c2 cells were cultured in hormone-deficient media; increased when estrogen receptor-α (ERα), estrogen receptor-ß (ERß), or androgen agonists were added; and increased in hearts from ERß-deficient mice. CSQ2 expression was reduced in mice fed a diet low in the methyl donor folic acid and in cells treated with 5-azadeoxycytidine suggesting an involvement of DNA methylation. DNA methylation in CpG in the CSQ2 CpG island was significantly different in males and females and was additionally changed postgonadectomy. Expression of DNA methyltransferases 1, 3a, and 3b was unchanged. These studies strongly link sex hormone-directed changes in CSQ2 expression to DNA methylation with changed expression correlated with altered left ventricular structure and function.


Asunto(s)
Hormonas Esteroides Gonadales/fisiología , Función Ventricular Izquierda/fisiología , Animales , Secuencia de Bases , Calcio/fisiología , Calsecuestrina/genética , Calsecuestrina/fisiología , Línea Celular , Metilación de ADN , Ecocardiografía , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/fisiología , Femenino , Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/biosíntesis , Hormonas Esteroides Gonadales/genética , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Miocardio/citología , Orquiectomía , Ovariectomía , Intercambiador de Sodio-Calcio/genética
14.
Cardiovasc Res ; 91(4): 598-605, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21613275

RESUMEN

AIMS: In heart cells, the mechanisms underlying refractoriness of the elementary units of sarcoplasmic reticulum (SR) Ca(2+) release, Ca(2+) sparks, remain unclear. We investigated local recovery of SR Ca(2+) release using experimental measurements and mathematical modelling. METHODS AND RESULTS: Repeated Ca(2+) sparks were induced from individual clusters of ryanodine receptors (RyRs) in quiescent rat ventricular myocytes, and we examined how changes in RyR gating influenced the time-dependent recovery of Ca(2+) spark amplitude and triggering probability. Repeated Ca(2+) sparks from individual sites were analysed in the presence of 50 nM ryanodine with: (i) no additional agents (control); (ii) 50 µM caffeine to sensitize RyRs; (iii) 50 µM tetracaine to inhibit RyRs; or (iv) 100 nM isoproterenol to activate ß-adrenergic receptors. Sensitization and inhibition of RyR clusters shortened and lengthened, respectively, the median interval between consecutive Ca(2+) sparks (caffeine 239 ms; control 280 ms; tetracaine 453 ms). Recovery of Ca(2+) spark amplitude, however, was exponential with a time constant of ∼100 ms in all cases. Isoproterenol both accelerated the recovery of Ca(2+) spark amplitude (τ = 58 ms) and shortened the median interval between Ca(2+) sparks (192 ms). The results were recapitulated by a mathematical model in which SR [Ca(2+)] depletion terminates Ca(2+) sparks, but not by an alternative model based on limited depletion and Ca(2+)-dependent inactivation of RyRs. CONCLUSION: Together, the results strongly suggest that: (i) local SR refilling controls Ca(2+) spark amplitude recovery; (ii) Ca(2+) spark triggering depends on both refilling and RyR sensitivity; and (iii) ß-adrenergic stimulation influences both processes.


Asunto(s)
Calcio/metabolismo , Miocardio/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/metabolismo , Animales , Calsecuestrina/fisiología , Simulación por Computador , Isoproterenol/farmacología , Masculino , Ratas , Rianodina/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología
15.
Biochem J ; 435(2): 391-9, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21265816

RESUMEN

CASQ (calsequestrin) is a Ca2+-buffering protein localized in the muscle SR (sarcoplasmic reticulum); however, it is unknown whether Ca2+ binding to CASQ2 is due to its location inside the SR rich in Ca2+ or due to its preference for Ca2+ over other ions. Therefore a major aim of the present study was to determine how CASQ2 selects Ca2+ over other metal ions by studying monomer folding and subsequent aggregation upon exposure to alkali (monovalent), alkaline earth (divalent) and transition (polyvalent) metals. We additionally investigated how CPVT (catecholaminergic polymorphic ventricular tachycardia) mutations affect CASQ2 structure and its molecular behaviour when exposed to different metal ions. Our results show that alkali and alkaline earth metals can initiate similar molecular compaction (folding), but only Ca2+ can promote CASQ2 to aggregate, suggesting that CASQ2 has a preferential binding to Ca2+ over all other metals. We additionally found that transition metals (having higher co-ordinated bonding ability than Ca2+) can also initiate folding and promote aggregation of CASQ2. These studies led us to suggest that folding and formation of higher-order structures depends on cationic properties such as co-ordinate bonding ability and ionic radius. Among the CPVT mutants studied, the L167H mutation disrupts the Ca2+-dependent folding and, when folding is achieved by Mn2+, L167H can undergo aggregation in a Ca2+-dependent manner. Interestingly, domain III mutants (D307H and P308L) lost their selectivity to Ca2+ and could be aggregated in the presence of Mg2+. In conclusion, these studies suggest that CPVT mutations modify CASQ2 behaviour, including folding, aggregation/polymerization and selectivity towards Ca2+.


Asunto(s)
Calsecuestrina/metabolismo , Cationes/metabolismo , Proteínas Mutantes/metabolismo , Miocardio/metabolismo , Taquicardia Ventricular/genética , Secuencia de Aminoácidos , Calcio/metabolismo , Calcio/farmacología , Calsecuestrina/química , Calsecuestrina/genética , Calsecuestrina/fisiología , Humanos , Metales Alcalinotérreos/metabolismo , Metales Alcalinotérreos/farmacología , Modelos Moleculares , Técnicas de Sonda Molecular , Datos de Secuencia Molecular , Proteínas Mutantes/análisis , Mutación Missense/fisiología , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína , Multimerización de Proteína/genética , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Especificidad por Sustrato , Taquicardia Ventricular/metabolismo
16.
Cell Physiol Biochem ; 26(4-5): 503-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21063088

RESUMEN

RATIONALE: The plateau phase of the ventricular action potential is the result of balanced Ca(2+) influx and K(+) efflux. The action potential is terminated by repolarizing K(+) currents. Under ß-adrenergic stimulation, both the Ca(2+)-influx and the delayed rectifier K(+) currents I(K) are stimulated to adjust the cardiac action potential duration to the enhanced heart rate and to ascertain adequate increase in net Ca(2+) influx. Intracellularly, a Calsequestrin2 (CASQ2)-ryanodine receptor complex serves as the most effective Ca(2+) reservoir/release system to aid the control of intracellular Ca(2+) levels. Currently, it is unclear if disease-associated CASQ2 gene variants alter intracellular free Ca(2+) concentrations and if cardiac ion channels are affected by it. OBJECTIVE: The goal of this study is to test if CASQ2 determines intracellular free Ca(2+) concentrations and to identify cardiac ion channels that are affected by it. Further, we aim to study disease-associated CASQ2 gene variants in this context. METHODS AND RESULTS: Here, we study the effects of the CASQ2 mutations R33Q, F189L, and D307H, located in highly conserved regions, on the functions of cardiac potassium channels in Xenopus oocytes using two electrode voltage clamp. As a result, CASQ2 wild type and CASQ2-mutants modulated hERG functions differently. Free Ca(2+) measurements and molecular dynamics simulations imply alterations in Ca(2+) buffer capacity paralled by changes in the dynamic behavior of the CASQ2-mutants compared to CASQ2 wild type. CONCLUSIONS: These in vitro and in silico data suggest a regulatory role of CASQ2 on cytosolic Ca(2+) and hERG channels which may contribute to the etiology of CPVT.


Asunto(s)
Calsecuestrina/fisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Potenciales de Acción/fisiología , Sustitución de Aminoácidos , Animales , Calcio/metabolismo , Calsecuestrina/genética , Calsecuestrina/metabolismo , Humanos , Simulación de Dinámica Molecular , Mutación , Oocitos/metabolismo , Potasio/metabolismo , Estructura Terciaria de Proteína , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Taquicardia Ventricular/etiología , Xenopus/crecimiento & desarrollo
17.
J Mol Cell Cardiol ; 49(1): 95-105, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20302875

RESUMEN

Mutations in the human cardiac calsequestrin gene (CASQ2) are linked to catecholaminergic polymorphic ventricular tachycardia (CPVT-2). This inherited disorder is characterized by life-threatening arrhythmias induced by physical and emotional stress in young patients. Here we identified a novel heterozygous missense mutation (K206N) in the CASQ2 gene in a symptomatic family in which one member died of cardiac arrest. The functional properties of CSQ(K206N) were investigated in comparison to the wild-type form of CASQ2 (CSQ(WT)) by expression in eukaryotic cell lines and neonatal mouse myocytes. The mutation created an additional N-glycosylation site resulting in a higher molecular weight form of the recombinant protein on immunoblots. The mutation reduced the Ca(2+) binding capacity of the protein and exhibited an altered aggregation state. Consistently, CSQ(K206N)-expressing myocytes exhibited an impaired response to caffeine administration, suggesting a lower Ca(2+) load of the sarcoplasmic reticulum (SR). The interaction of the mutated CSQ with triadin and the protein levels of the ryanodine receptor were unchanged but the maximal specific [(3)H]ryanodine binding was increased in CSQ(K206N)-expressing myocytes, suggesting a higher opening state of the SR Ca(2+) release channel. Myocytes with expression of CSQ(K206N) showed a higher rate of spontaneous SR Ca(2+) releases under basal conditions and after beta-adrenergic stimulation. We conclude that CSQ(K206N) caused a reduced Ca(2+) binding leading to an abnormal regulation of intracellular Ca(2+) in myocytes. This may then contribute to the increased propensity to trigger spontaneous Ca(2+) transients in CSQ(K206N)-expressing myocytes.


Asunto(s)
Calcio/metabolismo , Calsecuestrina , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcio de la Dieta/metabolismo , Calsecuestrina/genética , Calsecuestrina/metabolismo , Calsecuestrina/fisiología , Proteínas Portadoras , Células/metabolismo , Humanos , Células Musculares/metabolismo , Proteínas Musculares , Mutación , Mutación Missense , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología
18.
J Biol Chem ; 285(5): 3076-83, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19920148

RESUMEN

Mutations in cardiac ryanodine receptor (RYR2) and cardiac calsequestrin (CASQ2) genes are linked to catecholaminergic polymorphic ventricular tachycardia, a life-threatening genetic disease. They predispose young individuals to cardiac arrhythmia in the absence of structural abnormalities. One such mutation that changes an aspartic residue to histidine at position 307 in CASQ2 has been linked to catecholaminergic polymorphic ventricular tachycardia. In this study we made a transgenic mouse model expressing the mutant CASQ2(D307H) protein in a CASQ2 null background and investigated if the disease is caused by accelerated degradation of the mutant protein. Our data suggest that the mutant protein can be expressed, is relatively stable, and targets appropriately to the junctional sarcoplasmic reticulum. Moreover, it partially normalizes the ultrastructure of the sarcoplasmic reticulum, which was altered in the CASQ2 null background. In addition, overexpression of the mutant protein does not cause any pathology and/or structural changes in the myocardium. We further demonstrate, using purified protein, that the mutant protein is very stable under chemical and thermal denaturation but shows abnormal Ca(2+) buffering characteristics at high calcium concentrations. In addition, trypsin digestion studies reveal that the mutant protein is more susceptible to protease activity only in the presence of high Ca(2+). These studies collectively suggest that the D307H mutation can compromise the dynamic behavior of CASQ2 including supramolecular rearrangement upon Ca(2+) activation.


Asunto(s)
Calcio/química , Calsecuestrina/genética , Mutación , Retículo Sarcoplasmático/metabolismo , Animales , Tampones (Química) , Calsecuestrina/fisiología , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos , Miocardio/patología , Regiones Promotoras Genéticas , Pliegue de Proteína , Proteínas/metabolismo , Taquicardia Ventricular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...