Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800907

RESUMEN

BACKGROUND: In the present study, we examined superoxide-mediated excitatory nociceptive transmission on at-level neuropathic pain following spinal thoracic 10 contusion injury (SCI) in male Sprague Dawley rats. METHODS: Mechanical sensitivity at body trunk, neuronal firing activity, and expression of superoxide marker/ionotropic glutamate receptors (iGluRs)/CamKII were measured in the T7/8 dorsal horn, respectively. RESULTS: Topical treatment of superoxide donor t-BOOH (0.4 mg/kg) increased neuronal firing rates and pCamKII expression in the naïve group, whereas superoxide scavenger Tempol (1 mg/kg) and non-specific ROS scavenger PBN (3 mg/kg) decreased firing rates in the SCI group (* p < 0.05). SCI showed increases of iGluRs-mediated neuronal firing rates and pCamKII expression (* p < 0.05); however, t-BOOH treatment did not show significant changes in the naïve group. The mechanical sensitivity at the body trunk in the SCI group (6.2 ± 0.5) was attenuated by CamKII inhibitor KN-93 (50 µg, 3.9 ± 0.4) or Tempol (1 mg, 4 ± 0.4) treatment (* p < 0.05). In addition, the level of superoxide marker Dhet showed significant increase in SCI rats compared to the sham group (11.7 ± 1.7 vs. 6.6 ± 1.5, * p < 0.05). CONCLUSIONS: Superoxide and the pCamKII pathway contribute to chronic at-level neuropathic pain without involvement of iGluRs following SCI.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Hiperalgesia/tratamiento farmacológico , Proteínas del Tejido Nervioso/fisiología , Neuralgia/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Superóxidos/metabolismo , Animales , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Contusiones/fisiopatología , Óxidos N-Cíclicos/farmacología , Depuradores de Radicales Libres/uso terapéutico , Hiperalgesia/etiología , Masculino , Modelos Animales , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuralgia/etiología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores Ionotrópicos de Glutamato/efectos de los fármacos , Marcadores de Spin , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Sulfonamidas/farmacología , Transmisión Sináptica
2.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L810-L821, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30758990

RESUMEN

We have shown that calcium (Ca2+) oscillations in human pulmonary fibroblasts (HPFs) contribute to profibrotic effects of transforming growth factor-ß (TGF-ß) and that disruption of these oscillations blunts features of pulmonary fibrosis. Prostaglandin E2 (PGE2) exerts antifibrotic effects in the lung, but the mechanisms for this action are not well defined. We thus sought to explore interactions between PGE2 and the profibrotic agent TGF-ß in pulmonary fibroblasts (PFs) isolated from patients with or without idiopathic pulmonary fibrosis (IPF). PGE2 inhibited TGF-ß-promoted [Ca2+] oscillations and prevented the activation of Akt and Ca2+/calmodulin-dependent protein kinase-II (CaMK-II) but did not prevent activation of Smad-2 or ERK. PGE2 also eliminated TGF-ß-stimulated expression of collagen A1, fibronectin, and α-smooth muscle actin and reduced stress fiber formation in the HPFs. RNA sequencing revealed that HPFs preferentially express EP2 receptors relative to other prostanoid receptor subtypes: EP2 expression is ~10-fold higher than that of EP4 receptors; EP1 and EP3 receptors are barely detectable; and EP2-receptor expression is ~3.5-fold lower in PFs from IPF patients than in normal HPFs. The inhibitory effects of PGE2 on synthetic function and stress fiber formation were blocked by selective EP2 or EP4 antagonists and mimicked by selective EP2 or EP4 agonists, the phosphodiesterase inhibitor isobutylmethylxanthine and forskolin, all of which elevate cellular cAMP concentrations. We conclude that PGE2, likely predominantly via EP2 receptors, interferes with Ca2+ signaling, CaMK-II activation, and Akt activation in IPF-HPFs and HPFs treated with TGF-ß. Moreover, a decreased expression of EP2 receptors in pulmonary fibroblasts from IPF patients may contribute to the pathophysiology of this disease.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Dinoprostona/farmacología , Fibroblastos/metabolismo , Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Anciano , Anciano de 80 o más Años , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Femenino , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/patología , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Fibrosis Pulmonar/patología , Subtipo EP2 de Receptores de Prostaglandina E/biosíntesis , Proteína Smad2/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis
3.
Hum Mol Genet ; 25(11): 2194-2207, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27005420

RESUMEN

Limb girdle muscular dystrophy 2A is due to loss-of-function mutations in the Calpain 3 (CAPN3) gene. Our previous data suggest that CAPN3 helps to maintain the integrity of the triad complex in skeletal muscle. In Capn3 knock-out mice (C3KO), Ca2+ release and Ca2+/calmodulin kinase II (CaMKII) signaling are attenuated. We hypothesized that calpainopathy may result from a failure to transmit loading-induced Ca2+-mediated signals, necessary to up-regulate expression of muscle adaptation genes. To test this hypothesis, we compared transcriptomes of muscles from wild type (WT) and C3KO mice subjected to endurance exercise. In WT mice, exercise induces a gene signature that includes myofibrillar, mitochondrial and oxidative lipid metabolism genes, necessary for muscle adaptation. C3KO muscles fail to activate the same gene signature. Furthermore, in agreement with the aberrant transcriptional profile, we observe a commensurate functional defect in lipid metabolism whereby C3KO muscles fail to release fatty acids from stored triacylglycerol. In conjunction with the defects in oxidative metabolism, C3KO mice demonstrate reduced exercise endurance. Failure to up-regulate genes in C3KO muscles is due, in part, to decreased levels of PGC1α, a transcriptional co-regulator that orchestrates the muscle adaptation response. Destabilization of PGC1α is attributable to decreased p38 MAPK activation via diminished CaMKII signaling. Thus, we elucidate a pathway downstream of Ca2+-mediated CaMKII activation that is dysfunctional in C3KO mice, leading to reduced transcription of genes involved in muscle adaptation. These studies identify a novel mechanism of muscular dystrophy: a blunted transcriptional response to muscle loading resulting in chronic failure to adapt and remodel.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calpaína/genética , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Animales , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calpaína/biosíntesis , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Mutación , Estrés Oxidativo/genética , Activación Transcripcional/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
4.
Neurochem Res ; 43(4): 948-958, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29478199

RESUMEN

Epidemiological investigations and experimental studies indicate that chronic arsenic exposure can reduce learning and memory function. However, the underlying mechanism of this effect remains largely unknown. Emerging evidence suggests that microRNA (miRNA) play an important role in toxicant exposure and a regulatory role in cognitive function. In this study, we observed that subchronic arsenic exposure induced impairment of learning and memory and significantly up-regulated miRNA-219 (miR-219) expression in the mouse hippocampus. Furthermore, the expression of CaMKII, an experimentally validated target of miR-219, was decreased in the mice exposed to arsenic. Suppression of miR-219 by adeno-associated viral (AAV)-delivered anti-miR-219 prevented the arsenic-induced impairment of learning and memory and relieved the pathological changes in the synaptic structure of the hippocampus. Furthermore, we observed that the NMDA receptor subunit 2 (NR2) and the memory-related proteins c-Fos and c-Jun were up-regulated by inhibition of miR-219 in the mouse hippocampus. Taken together, the results of this study indicate that inhibition of miR-219 regulates arsenic-induced damage in the structure of the hippocampus and impairment of learning and memory, possibly by targeting CaMKII. Suppression of miR-219 may be a potential strategy to ameliorate arsenic-induced neurotoxicity.


Asunto(s)
Arsénico/toxicidad , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/metabolismo , MicroARNs/metabolismo , Sinapsis/metabolismo , Animales , Dependovirus/metabolismo , Hipocampo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Ratones , MicroARNs/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
5.
Cephalalgia ; 38(6): 1057-1070, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28738691

RESUMEN

Background A common characteristic of migraine-inducing substances is that they cause headache and no pain in other areas of the body. Few studies have compared pain mechanisms in the trigeminal and spinal systems and, so far, no major differences have been noted. We compared signalling molecules in the trigeminal and spinothalamic system after infusion of the migraine-provoking substance glyceryltrinitrate. Method A catheter was placed in the femoral vein of rats and one week later glyceryltrinitrate 4 µg/kg/min was infused for 20 min. Protein expression in the dura mater, trigeminal ganglion, nucleus caudalis, dorsal root ganglion and the dorsal horn of the thoracic spinal cord was analysed at different time points using western blotting and immunohistochemistry. Results Glyceryltrinitrate caused a threefold increase in expression of phosphorylated extracellular signal-regulated kinases at 30 min in the dura mater and nucleus caudalis ( P < 0.05) and at 2 h in the trigeminal ganglion with very few expressions in the dorsal root ganglion. In the nucleus caudalis, expression of phosphorylated extracellular signal-regulated kinases and Cam KII increased 2.6-fold and 3.2-fold, respectively, at 2 h after glycerytrinitrate infusion ( P < 0.01). p-CREB/ATF-1 upregulation was observed only at 30 min ( P < 0.05) in the nucleus caudalis. None of these markers showed increased expression in the regions of thoracic spinal cord dorsal horn. Conclusion The dura, trigeminal ganglion and nucleus caudalis are activated shortly after glycerytrinitrate infusion with long-lasting expression of phosphorylated extracellular signal-regulated kinases observed in the nucleus caudalis. These activations were not observed at the spinal level.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Núcleo Caudal del Trigémino/efectos de los fármacos , Ganglio del Trigémino/efectos de los fármacos , Animales , Duramadre/efectos de los fármacos , Masculino , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/fisiopatología , Nitroglicerina/toxicidad , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Núcleo Caudal del Trigémino/metabolismo , Ganglio del Trigémino/metabolismo , Regulación hacia Arriba , Vasodilatadores/toxicidad
6.
Int Heart J ; 59(1): 190-196, 2018 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-29279520

RESUMEN

The aim of this study was to investigate the effects of taurine (Tau) on primary cultured neonatal myocardial cells treated with hydrogen peroxide (H2O2) and the underlying mechanism. Primary cardiac myocytes from neonatal Wistar rats were pre-incubated with Tau, and its effects on cell viability and expression of CaM, CaMKII, p53, Bcl-2, and Bax were examined. Tau enhanced the viability of myocardial cells, decreased apoptosis, and alleviated the intracellular calcium overload, especially at dosages of 40 or 80 mM (P < 0.01 or P < 0.001, respectively). Moreover, Tau could inhibit the H2O2-induced decrease in CamKII and CaM expression at both the mRNA and protein levels. The pattern of CaMKII expression was consistent with that of the anti-apoptotic protein Bcl-2, but contrasted the pattern of the pro-apoptotic proteins p53 and Bax. Thus, our results show that Tau protects myocardial cells against damage caused by H2O2 exposure, suggesting that it might play a role in the mitochondrial apoptotic pathway by upregulating the expression of CaMKII to rescue myocardial cells. However, the underlying mechanism still needs to be investigated. In addition, we tested the protective effect of taurine on cardiac myocytes, and the effect of taurine on another model, specifically an animal model.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calmodulina/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Taurina/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calmodulina/biosíntesis , Células Cultivadas , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/toxicidad , Microscopía Confocal , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Reacción en Cadena de la Polimerasa , ARN/genética , Ratas , Ratas Wistar
7.
Neural Plast ; 2016: 4273280, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27957346

RESUMEN

Vagus nerve stimulation (VNS) enhances the consolidation of extinction of conditioned fear. High frequency stimulation of the infralimbic cortex (IL) produces long-term potentiation in the basolateral amygdala (BLA) in rats given VNS-paired extinction training, whereas the same stimulation produces long-term depression in sham-treated rats. The present study investigated the state of synaptic plasticity-associated proteins in the BLA that could be responsible for this shift. Male Sprague-Dawley rats were separated into 4 groups: auditory fear conditioning only (fear-conditioned); fear conditioning + 20 extinction trials (extended-extinction); fear conditioning + 4 extinction trials paired with sham stimulation (sham-extinction); fear conditioning + 4 extinction trials paired with VNS (VNS-extinction). Freezing was significantly reduced in extended-extinction and VNS-extinction rats. Western blots were used to quantify expression and phosphorylation state of synaptic plasticity-associated proteins such as Arc, CaMKII, ERK, PKA, and AMPA and NMDA receptors. Results show significant increases in GluN2B expression and phosphorylated CaMKII in BLA samples from VNS- and extended-extinction rats. Arc expression was significantly reduced in VNS-extinction rats compared to all groups. Administration of the GluN2B antagonist ifenprodil immediately after fear extinction training blocked consolidation of extinction learning. Results indicate a role for BLA CaMKII-induced GluN2B expression and reduced Arc protein in VNS-enhanced extinction.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteínas del Citoesqueleto/biosíntesis , Miedo/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Estimulación del Nervio Vago/métodos , Amígdala del Cerebelo/efectos de los fármacos , Animales , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Proteínas del Citoesqueleto/antagonistas & inhibidores , Antagonistas de Aminoácidos Excitadores/farmacología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/efectos de los fármacos , Miedo/psicología , Masculino , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Estimulación del Nervio Vago/psicología
8.
Molecules ; 21(4): 418, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-27043504

RESUMEN

Calcium overload is considered to be one of the mechanisms of cerebral ischemia. Ca(2+) influx and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and cAMP response element-binding protein (CREB) phosphorylation are considered to be involved in N-Methyl-d-aspartate (NMDA)-induced apoptosis process. This study investigated the neuroprotective effects of bryonolic acid (BA) in an NMDA-induced rat adrenal pheochromocytoma cell line (PC12) cells and the potential mechanism. PC12 was treated by NMDA to establish an excitotoxicity model. BA (110,100 and 1000 µM final concentration) was added to the medium 24 h prior to the addition of NMDA. Subsequently, a methyl thiazolyl tetrazolium (MTT) assay and a lactate dehydrogenase (LDH) release were performed. Ca(2+) concentration was demonstrated using a scanning-dual wavelength fluorimetric method. In addition, protein and mRNA levels were determined via Western blot and real-time PCR. In the presence of BA, MTT assay and LDH assay showed that more cells were viable in comparison with the NMDA group. Moreover, the concentration of Ca(2+) decreased with the addition of BA in culture. Furthermore, BA could upregulate protein expressions of Bcl-2, p-CREB, and p-CaMKII and downregulate protein expression of Bax. The mRNA results showed that the pattern of mRNA expression were similar to their respective protein levels. All these results indicate that BA protected PC12 cells against NMDA-induced apoptosis by inhibiting Ca(2+) influx and regulating gene expression in the Ca(2+)-CaMKII-CREB signal pathway. Therefore, the present study supports the notion that BA may be a promising neuroprotective agent for the treatment of cerebral ischemia disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Triterpenos/administración & dosificación , Animales , Isquemia Encefálica/inducido químicamente , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Supervivencia Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , N-Metilaspartato/toxicidad , Fármacos Neuroprotectores/química , Células PC12 , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Transducción de Señal/efectos de los fármacos , Triterpenos/química , Proteína X Asociada a bcl-2/biosíntesis
9.
J Mol Cell Cardiol ; 87: 38-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26241845

RESUMEN

Stromal interaction molecule 1 (STIM1) is a Ca(2+) sensor that partners with Orai1 to elicit Ca(2+) entry in response to endoplasmic reticulum (ER) Ca(2+) store depletion. While store-operated Ca(2+) entry (SOCE) is important for maintaining ER Ca(2+) homeostasis in non-excitable cells, it is unclear what role it plays in the heart, although STIM1 is expressed in the heart and upregulated during disease. Here we analyzed transgenic mice with STIM1 overexpression in the heart to model the known increase of this protein in response to disease. As expected, STIM1 transgenic myocytes showed enhanced Ca(2+) entry following store depletion and partial co-localization with the type 2 ryanodine receptor (RyR2) within the sarcoplasmic reticulum (SR), as well as enrichment around the sarcolemma. STIM1 transgenic mice exhibited sudden cardiac death as early as 6weeks of age, while mice surviving past 12weeks of age developed heart failure with hypertrophy, induction of the fetal gene program, histopathology and mitochondrial structural alterations, loss of ventricular functional performance and pulmonary edema. Younger, pre-symptomatic STIM1 transgenic mice exhibited enhanced pathology following pressure overload stimulation or neurohumoral agonist infusion, compared to controls. Mechanistically, cardiac myocytes isolated from STIM1 transgenic mice displayed spontaneous Ca(2+) transients that were prevented by the SOCE blocker SKF-96365, increased L-type Ca(2+) channel (LTCC) current, and enhanced Ca(2+) spark frequency. Moreover, adult cardiac myocytes from STIM1 transgenic mice showed both increased diastolic Ca(2+) and maximal transient amplitude but no increase in total SR Ca(2+) load. Associated with this enhanced Ca(2+) profile was an increase in cardiac nuclear factor of activated T-cells (NFAT) and Ca(2+)/calmodulin-dependent kinase II (CaMKII) activity. We conclude that STIM1 has an unexpected function in the heart where it alters communication between the sarcolemma and SR resulting in greater Ca(2+) flux and a leaky SR compartment.


Asunto(s)
Canales de Calcio/biosíntesis , Calcio/metabolismo , Cardiomiopatías/genética , Retículo Sarcoplasmático/metabolismo , Animales , Canales de Calcio/genética , Señalización del Calcio/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Ratones , Ratones Transgénicos , Células Musculares/metabolismo , Células Musculares/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/patología , Molécula de Interacción Estromal 1
10.
J Cell Biochem ; 116(10): 2258-69, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25820554

RESUMEN

Isoproterenol (ISO) induced nuclear translocation of calpain-2 which further increased susceptibility of cardiomyocyte apoptosis in tail-suspended rats. The underlying mechanisms remain elusive. In the present study, the results showed that ISO (10 nM) significantly elevated NADPH oxidases (NOXs) activity and NOXs-derived ROS productions which induced nuclear translocation of calpain-2 in cardiomyocytes of tail-suspended rats. In contrast, the inhibition of NADPH oxidase or cleavage of ROS not only reduced ROS productions, but also resisted nuclear translocation of calpain-2 and decreased ISO-induced apoptosis of cardiomyocyte in tail-suspended rats. ISO also increased the constitutive binding between calpain-2 and Ca(2+)/calmodulin-dependent protein kinase II δB (CaMK II δB) in nuclei, concomitant with the promotion of CaMK II δB degradation and subsequent down-regulation of Bcl-2 mRNA expression and the ratio of Bcl-2 to Bax protein in tail-suspended rat cardiomyocytes. These effects of ISO on cardiomyocytes were abolished by a calpain inhibitor PD150606. Inhibition of calpain significantly reduced ISO-induced loss of the mitochondrial membrane potential, cytochrome c release into the cytoplasm, as well as the activation of caspase-3 and caspase-9 in mitochondrial apoptotic pathway. In summary, the above results suggest that ISO increased NOXs-derived ROS which activated nuclear translocation of calpain-2, subsequently nuclear calpain-2 degraded CaMK II δB which reduced the ratio of Bcl-2 to Bax, and finally the mitochondria apoptosis pathway was triggered in tail-suspended rat cardiomyocytes. Therefore, calpain-2 may represent a potentially therapeutic target for prevention of oxidative stress-associated cardiomyocyte apoptosis.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calpaína/biosíntesis , NADH NADPH Oxidorreductasas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteína X Asociada a bcl-2/biosíntesis , Acrilatos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calpaína/metabolismo , Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/administración & dosificación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
11.
J Card Fail ; 21(9): 751-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26067684

RESUMEN

BACKGROUND: Alternative splicing factor (ASF)-regulated alternative splicing of calcium/calmodulin-dependent protein kinase IIδ (CaMKIIδ) plays an important role in pathologic cardiac remodeling. ASF can be phosphorylated by dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A). This study aimed to investigate the possible involvement of the Dyrk1A-ASF-CaMKIIδ signaling pathway in the progression of myocardial infarction (MI)-induced heart failure (HF). METHODS AND RESULTS: MI in rats was induced by means of left anterior descending coronary artery ligation. Seven weeks after MI, the increase in left ventricular internal diameter at end-diastole (LVIDd), and the decrease in both ejection fraction (EF) and fractional shortening (FS) indicated that MI rats had developed HF. Quantitative real time reverse-transcription polymerase chain reaction indicated the dysregulation of CaMKIIδ alternative splicing, ie, up-regulation of CaMKIIδA and CaMKIIδC and down-regulation of CaMKIIδB in the hearts of HF rats. Electrophoresis and immunostaining revealed that HF activated the phosphorylation of ASF and affected its subcellular localization. Western blot analysis demonstrated a significant elevation in the activity and expression of Dyrk1A in HF rats. Inversely, treatment of MI-induced HF rats with Dyrk1A inhibitor, either harmine or EGCG, improved the symptoms of HF, reversed the molecular changes of Dyrk1A and ASF, and regulated alternative splicing of CaMKIIδ in HF rats. CONCLUSIONS: Enhanced activation of Dyrk1A-ASF-CaMKIIδ signaling pathway may underlie the mechanisms of HF after MI, and Dyrk1A inhibition may contribute to inactivation of this pathway and thereby retard the progression of MI-induced HF.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Infarto del Miocardio/complicaciones , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , ARN/genética , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Inmunohistoquímica , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Fosforilación , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Quinasas DyrK
12.
Biochim Biophys Acta ; 1832(8): 1129-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23545117

RESUMEN

Mutations in the RNA binding protein FUS (fused in sarcoma) have been linked to a subset of familial amyotrophic lateral sclerosis (ALS) cases. The mutations are clustered in the C-terminal nuclear localization sequence (NLS). Various FUS mutants accumulate in the cytoplasm whereas wild-type (WT) FUS is mainly nuclear. Here we investigate the effect of one ALS causing mutant (FUS-ΔNLS, also known as R495X) on pre-mRNA splicing and RNA expression using genome wide exon-junction arrays. Using a non-neuronal stable cell line with inducible FUS expression, we detected early changes in RNA composition. In particular, mutant FUS-ΔNLS increased calcium/calmodulin-dependent protein kinase II inhibitor 2 (CAMK2N2) at both mRNA and protein levels, whereas WT-FUS had no effect. Chromatin immunoprecipitation experiments showed that FUS-ΔNLS accumulated at the CAMK2N2 promoter region, whereas promoter occupation by WT-FUS remained constant. Given the loss of FUS-ΔNLS in the nucleus through the mutation-induced translocation, this increase of promoter occupancy is surprising. It indicates that, despite the obvious cytoplasmic accumulation, FUS-ΔNLS can act through a nuclear gain of function mechanism.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Mutación , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Línea Celular , Núcleo Celular/genética , Cromatina/genética , Cromatina/metabolismo , Citoplasma/genética , Exones , Estudio de Asociación del Genoma Completo/métodos , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Precursores del ARN/genética , Empalme del ARN/genética , Proteína FUS de Unión a ARN/biosíntesis
13.
Pflugers Arch ; 466(2): 319-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23881186

RESUMEN

Increased adult cardiac fibroblast proliferation results in an increased collagen deposition responsible for the fibrosis accompanying pathological remodelling of the heart. The mechanisms regulating cardiac fibroblast proliferation remain poorly understood. Using a minimally invasive transverse aortic banding (MTAB) mouse model of cardiac hypertrophy, we have assessed fibrosis and cardiac fibroblast proliferation. We have investigated whether calcium/calmodulin-dependent protein kinase IIδ (CaMKIIδ) regulates proliferation in fibroblasts isolated from normal and hypertrophied hearts. It is known that CaMKIIδ plays a central role in cardiac myocyte contractility, but nothing is known of its role in adult cardiac fibroblast function. The MTAB model used here produces extensive hypertrophy and fibrosis. CaMKIIδ protein expression and activity is upregulated in MTAB hearts and, specifically, in cardiac fibroblasts isolated from hypertrophied hearts. In response to angiotensin II, cardiac fibroblasts isolated from MTAB hearts show increased proliferation rates. Inhibition of CaMKII with autocamtide inhibitory peptide inhibits proliferation in cells isolated from both sham and MTAB hearts, with a significantly greater effect evident in MTAB cells. These results are the first to show selective upregulation of CaMKIIδ in adult cardiac fibroblasts following cardiac hypertrophy and to assign a previously unrecognised role to CaMKII in regulating adult cardiac fibroblast function in normal and diseased hearts.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cardiomegalia/fisiopatología , Fibroblastos/fisiología , Angiotensina II/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proliferación Celular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo
14.
Genet Mol Res ; 13(2): 2914-21, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24634301

RESUMEN

Aerosol oligonucleotide therapy has vast potential in pulmonary system diseases. Reactive oxygen species (ROS) play an important role in complex physiological processes such as cell signaling, apoptosis, etc. Therefore, to determine the mechanism of ROS involvement in polyethylenimine/oligonucleotide (PEI/ON) endocytosis in cells, we measured the fluorescence intensities of fluorescein isothiocyanate-labeled ON complexes with PEI and the changes in cytosolic Ca(2+) concentration ([Ca(2+)]c) in A549 cells after hydrogen peroxide (H2O2) stimulation. Results showed that improved uptake of PEI/ON complexes was independent of the rise of [Ca(2+)]c in A549 cells, including the Ca(2+) inflow and the release of Ca(2+) from intracellular stores induced by 500 µM H2O2. However, the enhanced uptake efficiency was almost completely abolished by the calcium/calmodulin-dependent protein kinase II (CaMKII) inhibitor and the microtube depolymerized drug. CaMKII-dependent microtube polymerization may be responsible for the enhanced uptake of PEI/ON complexes in A549 cells under oxidative stress conditions. This study is useful for research aimed at improving aerosol oligonucleotide therapy in pulmonary system diseases.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Enfermedades Pulmonares/tratamiento farmacológico , Oligonucleótidos/metabolismo , Polietileneimina/metabolismo , Administración por Inhalación , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Enfermedades Pulmonares/patología , Oligonucleótidos/química , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Polietileneimina/química , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
15.
J Neurochem ; 126(3): 349-59, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23651084

RESUMEN

The pre-Bötzinger complex (pre-BötC) in the ventrolateral medulla oblongata is a presumed kernel of respiratory rhythmogenesis. Ca(2+) -activated non-selective cationic current is an essential cellular mechanism for shaping inspiratory drive potentials. Ca(2+) /calmodulin-dependent protein kinase II (CaMKII), an ideal 'interpreter' of diverse Ca(2+) signals, is highly expressed in neurons in mediating various physiological processes. Yet, less is known about CaMKII activity in the pre-BötC. Using neurokinin-1 receptor as a marker of the pre-BötC, we examined phospho (P)-CaMKII subcellular distribution, and found that P-CaMKII was extensively expressed in the region. P-CaMKII-ir neurons were usually oval, fusiform, or pyramidal in shape. P-CaMKII immunoreactivity was distributed within somas and dendrites, and specifically in association with the post-synaptic density. In dendrites, most synapses (93.1%) examined with P-CaMKII expression were of asymmetric type, occasionally with symmetric type (6.9%), whereas in somas, 38.1% were of symmetric type. P-CaMKII asymmetric synaptic identification implicates that CaMKII may sense and monitor Ca(2+) activity, and phosphorylate post-synaptic proteins to modulate excitatory synaptic transmission, which may contribute to respiratory modulation and plasticity. In somas, CaMKII acts on both symmetric and asymmetric synapses, mediating excitatory and inhibitory synaptic transmission. P-CaMKII was also localized to the perisynaptic and extrasynaptic regions in the pre-BötC.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Bulbo Raquídeo/enzimología , Sinapsis/enzimología , Transmisión Sináptica/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/análisis , Inmunohistoquímica , Bulbo Raquídeo/ultraestructura , Microscopía Electrónica de Transmisión , Neuronas/enzimología , Neuronas/ultraestructura , Ratas , Ratas Sprague-Dawley
17.
Anesth Analg ; 116(3): 712-21, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23400989

RESUMEN

BACKGROUND: Abnormalities in peripheral nerves and dorsal root ganglia are noticed in the early stage of experimentally provoked diabetic neuropathy. Enzyme calcium/calmodulin-dependent protein kinase II (CaMKII) may have a modulating role in diabetic neuropathy because of its role in calcium homeostasis. METHODS: A model of type 1 diabetes mellitus (DM1) was induced with 55 mg/kg of the streptozotocin and for DM2 induction a combination of high-fat diet and low-dose streptozotocin (35 mg/kg) was used. Pain-related behavior was analyzed using thermal and mechanical stimuli. Two weeks and 2 months after induction of diabetes rats were euthanized, and the expression of CaMKII and its isoforms in the dorsal root ganglia were analyzed using immunofluorescence. RESULTS: Both types of diabetes were successfully induced, as confirmed by hyperglycemia. Increased pain-related behavior became evident in DM1 rats in 2 weeks after diabetes induction, but not in DM2 rats. The expression of total CaMKII and the phosphorylated α isoform of CaMKII increased in DM1 animals concurrently with pain-related behavior. Expression of α, ß, γ, and δ isoforms in DM1 animals and expression of total CaMKII and all of its analyzed isoforms in DM2 animals remained unchanged. CONCLUSIONS: Our findings may indicate involvement of CaMKII in transmission of nociceptive input early in DM1, but not in DM2. CaMKII may be a suitable pharmacological target for diabetic neuropathy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 2/enzimología , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Dolor/enzimología , Animales , Conducta Animal/fisiología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Masculino , Dolor/fisiopatología , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
18.
J Neurosci ; 31(2): 725-34, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21228181

RESUMEN

Group I metabotropic glutamate receptors (mGluRs) stimulation activates translation-dependent epileptogenesis in the hippocampus. This translation is regulated by repressors, including BC1 RNA and fragile X mental retardation protein (FMRP). Recent data indicate that group I mGluR stimulation exerts bidirectional control over FMRP level by activating translation and ubiquitin-proteasome system (UPS)-dependent proteolysis for the up- and downregulation of the protein, respectively. At present, the temporal relationship of translation and proteolysis on FMRP and their interplay for group I mGluR-mediated translation and epileptogenesis are unknown. We addressed these issues by using mouse hippocampal slices. Agonist [(S)-3,5-dihydroxyphenylglycine (DHPG)] stimulation of group I mGluRs caused a biphasic change in FMRP level. An initial decrease (within 10 min) was followed by an increase at 30 min. When slices were pretreated with translation inhibitor (anisomycin or cycloheximide), group I mGluRs elicited a sustained decrease in FMRP. This decrease was prevented by a proteasome inhibitor [Z-Leu-Leu-Leu-CHO (MG-132)]. When slices were pretreated with MG-132 alone, DHPG no longer elicited any change in FMRP. MG-132 also suppressed increase in other proteins, including postsynaptic density-95 and α-calcium/calmodulin-dependent protein kinase II, normally elicited by group I mGluR stimulation. Physiological experiments showed that proteasome inhibitor suppressed group I mGluR-induced prolonged synchronized discharges. However, proteasome inhibitor did not affect group I mGluR-induced prolonged synchronized discharges in Fmr1(-/-) preparations, where functional FMRP is absent. The results suggest that constitutive FMRP in hippocampal cells acts as a brake on group I mGluR-mediated translation and epileptogenesis. FMRP downregulation via UPS removes this brake enabling group I mGluR-mediated translation and epileptogenesis.


Asunto(s)
Epilepsia/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/biosíntesis , Hipocampo/metabolismo , Receptores de Glutamato Metabotrópico/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Homólogo 4 de la Proteína Discs Large , Epilepsia/fisiopatología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Guanilato-Quinasas , Hipocampo/fisiopatología , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Biosíntesis de Proteínas , Receptores de Glutamato Metabotrópico/agonistas
19.
J Mol Cell Cardiol ; 53(4): 459-68, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22884543

RESUMEN

Failure of molecular chaperones to direct the correct folding of newly synthesized proteins leads to the accumulation of misfolded proteins in cells. HSPA4 is a member of the heat shock protein 110 family (HSP110) that acts as a nucleotide exchange factor of HSP70 chaperones. We found that the expression of HSPA4 is upregulated in murine hearts subjected to pressure overload and in failing human hearts. To investigate the cardiac function of HSPA4, Hspa4 knockout (KO) mice were generated and exhibited cardiac hypertrophy and fibrosis. Hspa4 KO hearts were characterized by a significant increase in heart weight/body weight ratio, elevated expression of hypertrophic and fibrotic gene markers, and concentric hypertrophy with preserved contractile function. In response to pressure overload, cardiac hypertrophy and remodeling were further aggravated in the Hspa4 KO compared to wild type (WT) mice. Cardiac hypertrophy in Hspa4 KO hearts was associated with enhanced activation of gp130-STAT3, CaMKII, and calcineurin-NFAT signaling. Protein blot and immunofluorescent analyses showed a significant accumulation of polyubiquitinated proteins in cardiac cells of Hspa4 KO mice. These results suggest that the myocardial remodeling of Hspa4 KO mice is due to accumulation of misfolded proteins resulting from impaired chaperone activity. Further analyses revealed a significant increase in cross sectional area of cardiomyocytes, and in expression levels of hypertrophic markers in cultured neonatal Hspa4 KO cardiomyocytes suggesting that the hypertrophy of mutant mice was a result of primary defects in cardiomyocytes. Gene expression profile in hearts of 3.5-week-old mice revealed a differentially expressed gene sets related to ion channels, muscle-specific contractile proteins and stress response. Taken together, our in vivo data demonstrate that Hspa4 gene ablation results in cardiac hypertrophy and fibrosis, possibly, through its role in protein quality control mechanism.


Asunto(s)
Cardiomegalia/genética , Proteínas del Choque Térmico HSP110/fisiología , Miocardio/patología , Animales , Animales Recién Nacidos , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Calcineurina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , Proteínas Contráctiles/genética , Receptor gp130 de Citocinas/biosíntesis , Fibrosis/genética , Proteínas del Choque Térmico HSP110/genética , Homeostasis , Humanos , Canales Iónicos/genética , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Transcripción NFATC/metabolismo , Pliegue de Proteína , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal , Estrés Fisiológico/genética , Proteínas Ubiquitinadas/metabolismo , Remodelación Ventricular
20.
J Cell Mol Med ; 16(12): 3052-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22970977

RESUMEN

There are controversies concerning the capacity of Rosuvastatin to attenuate heart failure in end-stage hypertension. The aim of the study was to show whether the Rosuvastatin might be effective or not for the heart failure treatment. Twenty-one spontaneously hypertensive rats (SHRs) aged 52 weeks with heart failure were randomly divided into three groups: two receiving Rosuvastatin at 20 and 40 mg/kg/day, respectively, and the third, placebo for comparison with seven Wistar-Kyoto rats (WKYs) as controls. After an 8-week treatment, the systolic blood pressure (SBP) and echocardiographic features were evaluated; mRNA level of B-type natriuretic peptide (BNP) and plasma NT-proBNP concentration were measured; the heart tissues were observed under electron microscope (EM); myocardial sarcoplasmic reticulum Ca(2+) pump (SERCA-2) activity and mitochondria cytochrome C oxidase (CCO) activity were measured; the expressions of SERCA-2a, phospholamban (PLB), ryanodine receptor2 (RyR2), sodium-calcium exchanger 1 (NCX1), Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and protein phosphatase inhibitor-1 (PPI-1) were detected by Western blot and RT-qPCR; and the total and phosphorylation of protein kinase Cα/ß (PKCα/ß) were measured. Aged SHRs with heart failure was characterized by significantly decreased left ventricular ejection fraction and left ventricular fraction shortening, enhanced left ventricular end-diastolic diameter and LV Volume, accompanied by increased plasma NT-proBNP and elevated BNP gene expression. Damaged myofibrils, vacuolated mitochondria and swollen sarcoplasmic reticulum were observed by EM. Myocardium mitochondria CCO and SERCA-2 activity decreased. The expressions of PLB and NCX1 increased significantly with up-regulation of PPI-1 and down-regulation of CaMKII, whereas that of RyR2 decreased. Rosuvastatin was found to ameliorate the heart failure in aged SHRs and to improve changes in SERCA-2a, PLB, RyR2, NCX1, CaMKII and PPI-1; PKCα/ß2 signal pathway to be suppressed; the protective effect of Rosuvastatin to be dose dependent. In conclusion, the heart failure of aged SHRs that was developed during the end stage of hypertension could be ameliorated by Rosuvastatin.


Asunto(s)
Fluorobencenos/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-alfa/metabolismo , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Envejecimiento , Animales , Presión Sanguínea/efectos de los fármacos , Proteínas de Unión al Calcio/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Regulación hacia Abajo , Complejo IV de Transporte de Electrones/metabolismo , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Miofibrillas/efectos de los fármacos , Miofibrillas/patología , Péptido Natriurético Encefálico/sangre , Péptido Natriurético Encefálico/genética , Fragmentos de Péptidos/sangre , Fosforilación , Proteínas/análisis , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Rosuvastatina Cálcica , Canal Liberador de Calcio Receptor de Rianodina/biosíntesis , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal , Intercambiador de Sodio-Calcio/biosíntesis , Volumen Sistólico/efectos de los fármacos , Regulación hacia Arriba , Función Ventricular Izquierda/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda