Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 491(3): 687-692, 2017 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-28754591

RESUMEN

Ca2+/calmodulin-dependent protein kinase II (CaMKII) has been implicated in myocardial ischemia/reperfusion (IR) injury. The aim of this study was to determine the effect of CaMKII on the damage to membrane skeleton proteins, which is an important cause of IR injury. Isolated rat hearts were subjected to 45-min global ischemia/2-h reperfusion. Both KN-62 and KN-93 were used to inhibit CaMKII. Compared with controls, the hearts in the IR group exhibited remarkable myocardial injury area, LDH release, cell apoptosis and contractile dysfunction, along with an increase in the phosphorylation of CaMKII and its substrate phospholamban. Treatment with either KN-62 or KN-93 mitigated both the heart injury and the phosphorylation of CaMKII and phospholamban. The analysis of cell skeleton proteins revealed that IR injury resulted in an increase in the 150-kDa fragments resulting from the degradation of α-fodrin and dystrophin translocating from the sarcolemmal membrane to the cytosol and a decrease in the 220-kDa isoform of ankyrin-B. As expected, Evans blue dye staining showed an increase in membrane permeability or membrane rupture in the IR group. All of these alterations were alleviated by treatment with either KN-62 or KN-93. In addition, both KN-62 and KN-93 blocked the activity and membrane recruitment of calpain, a key protease responsible for destroying cell skeleton proteins during IR injury. In conclusion, our data provide evidence that damage to membrane skeleton proteins via calpain is a destructive downstream event of CaMKII activation in the setting of myocardial IR injury.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Animales , Bencilaminas/administración & dosificación , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calpaína/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/administración & dosificación , Corazón/efectos de los fármacos , Técnicas In Vitro , Masculino , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Sulfonamidas/administración & dosificación , Resultado del Tratamiento
2.
Clin Exp Pharmacol Physiol ; 43(10): 951-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27279457

RESUMEN

This study determined the effects of glutamate on the Ca(2+) paradoxical heart, which is a model for Ca(2+) overload-induced injury during myocardial ischaemia and reperfusion, and evaluated its effect on a known mediator of injury, calpain. An isolated rat heart was retrogradely perfused in a Langendorff apparatus. Ca(2+) paradox was elicited via perfusion with a Ca(2+) -free Krebs-Henseleit (KH) solution for 3 minutes followed by Ca(2+) -containing normal KH solution for 30 minutes. The Ca(2+) paradoxical heart exhibited almost no viable tissue on triphenyltetrazolium chloride staining and markedly increased LDH release, caspase-3 activity, cytosolic cytochrome c content, and apoptotic index. These hearts also displayed significantly increased LVEDP and a disappearance of LVDP. Glutamate (5 and 20 mmol/L) significantly alleviated Ca(2+) paradox-induced injury. In contrast, 20 mmol/L mannitol had no effect on Ca(2+) paradox. Ca(2+) paradox significantly increased the extent of the translocation of µ-calpain to the sarcolemmal membrane and the proteolysis of α-fodrin, which suggests calpain activation. Glutamate also blocked these effects. A non-selective inhibitor of glutamate transporters, dl-TBOA (10 µmol/L), had no effect on control hearts, but it reversed glutamate-induced cardioprotection and reduction in calpain activity. Glutamate treatment significantly increased intracellular glutamate content in the Ca(2+) paradoxical heart, which was also blocked by dl-TBOA. We conclude that glutamate protects the heart against Ca(2+) overload-induced injury via glutamate transporters, and the inhibition of calpain activity is involved in this process.


Asunto(s)
Calcio/toxicidad , Calpaína/antagonistas & inhibidores , Calpaína/metabolismo , Cardiotónicos/uso terapéutico , Ácido Glutámico/uso terapéutico , Daño por Reperfusión Miocárdica/metabolismo , Animales , Calcio/metabolismo , Cardiotónicos/farmacología , Ácido Glutámico/farmacología , Corazón/efectos de los fármacos , Corazón/fisiología , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/prevención & control , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
3.
Cell Physiol Biochem ; 35(6): 2121-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25896056

RESUMEN

AIMS: The aim of this study was to determine whether calpain is involved in Cl(-)-induced myocardial ischemia/reperfusion (I/R) injury. METHODS: Isolated rat hearts were subjected to either 45 min of global no-flow ischemia followed by reperfusion or successive perfusion with Ca(2+)-free KH solution for 3 min and normal KH solution for 30 min, also known as Ca(2+) paradox. RESULTS: The hearts in the I/R group exhibited increases in myocardial injury area, LDH release, caspase 3 activity and apoptotic indices and a marked decline in cardiac performance. As was the case regarding the effects of MDL 28170, an inhibitor of calpain, treatment with 5 µM NPPB, 5 µM DIDS and low Cl(-) significantly attenuated cardiac injury. Moreover, each of the treatments significantly protected against Ca(2+) overload-induced injury in the setting of Ca(2+) paradox. The Western blot and immunofluorescence data revealed that there was an increase in the percentages of calpain membrane-positive cells and the numbers of fragments resulting from the calpain-mediated proteolysis of α-fodrin in both the I/R and the Ca(2+) paradox, indicating that the activation of calpain occurred. More importantly, these effects were mitigated by the blockade of transmembrane Cl(-) flux, as was accomplished via MDL 28170. CONCLUSION: Our results provide evidence that the blockade of transmembrane Cl(-) flux mitigates I/R-induced cardiac injury via the inhibition of calpain activity. They also indicate that intracellular Ca(2+) overload regulates calpain activation in the setting of Cl(-)-induced injury.


Asunto(s)
Calpaína/antagonistas & inhibidores , Cloruros/efectos adversos , Cloruros/antagonistas & inhibidores , Lesiones Cardíacas/inducido químicamente , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 3/metabolismo , Dipéptidos/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Lesiones Cardíacas/tratamiento farmacológico , Masculino , Proteínas de Microfilamentos/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Proteolisis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Shock ; 60(2): 255-261, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37278996

RESUMEN

ABSTRACT: Mitochondrial damage is an important cause of heart dysfunction after severe burn injury. However, the pathophysiological process remains unclear. This study aims to examine the mitochondrial dynamics in the heart and the role of µ-calpain, a cysteine protease, in this scenario. Rats were subjected to severe burn injury treatment, and the calpain inhibitor MDL28170 was administered intravenously 1 h before or after burn injury. Rats in the burn group displayed weakened heart performance and decreased mean arterial pressure, which was accompanied by a diminishment of mitochondrial function. The animals also exhibited higher levels of calpain in mitochondria, as reflected by immunofluorescence staining and activity tests. In contrast, treatment with MDL28170 before any severe burn diminished these responses to a severe burn. Burn injury decreased the abundance of mitochondria and resulted in a lower percentage of small mitochondria and a higher percentage of large mitochondria. Furthermore, burn injury caused an increase in the fission protein DRP1 in the mitochondria and a decrease in the inner membrane fusion protein OPA1. Similarly, these alterations were also blocked by MDL28170. Of note, inhibition of calpain yielded the emergence of more elongated mitochondria along with membrane invagination in the middle of the longitude, which is an indicator of the fission process. Finally, MDL28170, administered 1 h after burn injury, preserved mitochondrial function and heart performance, and increased the survival rate. Overall, these results provided the first evidence that mitochondrial recruitment of calpain confers heart dysfunction after severe burn injury, which involves aberrant mitochondrial dynamics.


Asunto(s)
Quemaduras , Calpaína , Ratas , Animales , Dinámicas Mitocondriales , Mitocondrias/metabolismo , Quemaduras/complicaciones , Quemaduras/tratamiento farmacológico , Quemaduras/metabolismo
5.
Clin Exp Pharmacol Physiol ; 39(4): 385-92, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22356295

RESUMEN

The calcium paradox represents an important model in which to study myocardial injuries due to intracellular Ca(2+) overload. In a previous study, calpain was transiently activated in Ca(2+) -paradoxic hearts. The aim of the present study was to determine the role of calpain in myocardial dysfunction in hearts subjected to the Ca(2+) paradox and to elucidate the underlying mechanisms. Rat hearts were isolated, Langendorff perfused and subjected to the Ca(2+) paradox, which was induced by 3 min Ca(2+) depletion followed by 30 min Ca(2+) repletion, in the presence or absence of the calpain inhibitor 10 umol/L MDL 28170. Cardiac function was evaluated. Furthermore, cell death and the degradation of troponin I (TnI) were assessed and calpain activity was determined by measurement of the α-fodrin fragment and confocal image analysis. Upon Ca(2+) repletion, the hearts immediately deteriorated, exhibiting a marked depression in cardiac function and an enlarged myocardial injury area. This was accompanied by significant increases in lactate dehydrogenase, mitochondrial release of cytochrome c, the apoptotic index and degraded TnI. These changes were significantly inhibited by MDL 28170, with the exception of TnI degradation. Compared with the control group, Ca(2+) -paradoxic hearts showed a marked increase in cleaved 150 kDa fragments resulting from specific calpain-mediated proteolysis of α-fodrin. This effect was attenuated by MDL 28170. Confocal image analysis revealed the translocation of both µ- and m-calpain to the sarcolemmal membrane in Ca(2+) -paradoxic hearts, indicating increased activity of both isoforms. The results suggest that the Ca(2+) paradox promotes calpain activity, leading to necrosis, apoptosis and myocardial dysfunction.


Asunto(s)
Calcio/deficiencia , Calpaína/antagonistas & inhibidores , Cardiotónicos/farmacología , Dipéptidos/farmacología , Glicoproteínas/farmacología , Miocardio/metabolismo , Animales , Calcio/farmacología , Calpaína/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Masculino , Miocardio/patología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
6.
Cell Death Discov ; 8(1): 10, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013173

RESUMEN

Stress cardiomyopathy is a major clinical complication after severe burn. Multiple upstream initiators have been identified; however, the downstream targets are not fully understood. This study assessed the role of the plasma membrane in this process and its relationship with the protease µ-calpain and tumor necrosis factor-alpha (TNF-α). Here, third-degree burn injury of approximately 40% of the total body surface area was established in rats. Plasma levels of LDH and cTnI and cardiac cell apoptosis increased at 0.5 h post burn, reached a peak at 6 h, and gradually declined at 24 h. This effect correlated well with not only the disruption of cytoskeletal proteins, including dystrophin and ankyrin-B, but also with the activation of µ-calpain, as indicated by the cleaved fragments of α-spectrin and membrane recruitment of the catalytic subunit CAPN1. More importantly, these alterations were diminished by blocking calpain activity with MDL28170. Burn injury markedly increased the cellular uptake of Evans blue, indicating membrane integrity disruption, and this effect was also reversed by MDL28170. Compared with those in the control group, cardiac cells in the burn plasma-treated group were more prone to damage, as indicated by a marked decrease in cell viability and increases in LDH release and apoptosis. Of note, these alterations were mitigated by CAPN1 siRNA. Moreover, after neutralizing TNF-α with rhTNFR:Fc, calpain activity was blocked, and heart function was improved. In conclusion, we identified µ-calpain as a trigger for severe burn-induced membrane disruption in the heart and provided evidence for the application of rhTNFR:Fc to inhibit calpain for cardioprotection.

7.
Crit Care Med ; 38(12): 2365-76, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20890194

RESUMEN

OBJECTIVE: Acute myocardial ischemia induces electrical and chemical uncoupling of gap junctions, which contributes to conduction abnormalities and re-entrant arrhythmias. We tested the hypothesis that structure and function of Connexin43 may vibrate during acute myocardial ischemia and reperfusion and κ-opioid receptor stimulation may stabilize the alteration of Connexin43. DESIGN: An animal intervention study was conducted with comparison to a control group. SETTING: University preclinical research laboratory. SUBJECTS: Age-, weight-, and sex-matched Sprague-Dawley rats. INTERVENTIONS: Adult rat hearts were subjected to ischemia or ischemia/reperfusion, which was induced by temporary occlusion of the left main coronary artery. U50488H was given 10 mins before tissue specimens were taken or before ischemia (1.5 mg/kg, intravenous) and nor-BNI was given 15 mins before tissue specimens were taken or before ischemia (2 mg/kg, intravenous). Tissue samples came from left ventricular myocardium of the rat hearts. MEASUREMENTS AND MAIN RESULTS: Electrocardiogram, immunohistochemistry, immunoblotting, and reverse transcription-polymerase chain reaction were used to measure changes of arrhythmias, protein, and gene expression of Connexin43, respectively. κ-opioid receptor activation with U50 decreased arrhythmia in a model of myocardial ischemia and reperfusion. In normal hearts, immunohistochemical data showed reduced amount and lateralization of Connexin43 induced by κ-opioid receptor activation, whereas immunoblotting data demonstrated no significant changes between control and U50 group. During ischemia, however, Connexin43 protein underwent dephosphorylation and degradation, and Connexin43 mRNA was upregulated. These alterations were significantly attenuated on κ-opioid receptor stimulation. During ischemia and reperfusion, Connexin43 protein underwent dephosphorylation and degradation and recovered slowly during reperfusion. Activation of κ-opioid receptor accelerated recovery of phosphorylated and total Connexin43. CONCLUSIONS: In normal rat hearts, Connexin43 translocates from intercellular junctions to intracellular locations on κ-opioid receptor activation. In rat hearts experiencing acute myocardial ischemia and reperfusion, protein and gene expression of Connexin43 undergo vibration. This phenomenon is stabilized when κ-opioid receptor is activated and by the fact that κ-opioid receptor produces antiarrhythmic effects.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Conexina 43/metabolismo , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Receptores Opioides kappa/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/metabolismo , Animales , Arritmias Cardíacas/fisiopatología , Western Blotting , Conexina 43/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Uniones Comunicantes/efectos de los fármacos , Inmunohistoquímica , Masculino , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Cell Death Dis ; 11(5): 388, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32439852

RESUMEN

Previous studies indicated that Ca2+/calmodulin-dependent kinase II (CaMKII), a kinase involved in the modulation of ryanodine receptor activity, activates Ca2+-regulated protease µ-calpain to promote myocardial ischemia/reperfusion injury. This study was performed to explore the underlying mechanisms in CaMKII-induced calpain activation to better understand heart injury. To examine the Ca2+ paradox and ischemia/reperfusion injury, isolated rat hearts were subjected to a Ca2+-free solution for 3 min, or left coronary artery occlusion for 40 min, prior to restoration of normal perfusion. Blockade of trans-sarcoplasmic reticulum Ca2+ flux using ryanodine and thapsigargin failed to prevent Ca2+ paradox-induced heart injury. In contrast, the Ca2+ paradox increased CaMKII auto-phosphorylation at Thr287, while the CaMKII inhibitor KN-62 and the Na+/Ca2+ exchanger inhibitor KB-R7943 alleviated heart injury and calpain activity. Intriguingly, the binding of µ-calpain large subunit calpain-1 (CAPN1) to phospho-CaMKII was blunted by both inhibitors. Thus, a Ca2+ leak via the ryanodine receptor is not an essential element in CaMKII-elicited calpain activation. In hearts receiving vector injection, ischemia/reperfusion caused elevated calpain activity and α-fodrin degradation, along with membrane integrity damage, similar to the effects noted in control hearts. Importantly, all these alterations were diminished with delivery of adeno-associated virus expressing mutant CaMKIIδC T287A. Ischemia/reperfusion increased CaMKII auto-phosphorylation and binding of CAPN1 to phospho-CaMKII, and facilitated the translocation of phospho-CaMKII and CAPN1 to the plasma membrane, all of which were reversed by injecting CaMKII mutant. Furthermore, the relocation capacity and the interaction of CaMKII with CAPN1 appeared to be dependent upon CaMKII autophosphorylation, as its mutant delivery increased the level of CaMKII, but did not increase membrane content of CaMKII and CAPN1, or their interactions. Together, CaMKII/calpain interaction represents a new avenue for mediating myocardial ischemia/reperfusion injury, and CaMKII likely serves as both a kinase and a carrier, thereby promoting calpain membrane translocation and activation.


Asunto(s)
Calcio/metabolismo , Calpaína/metabolismo , Isquemia/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Proteínas de Unión al Calcio/metabolismo , Isquemia/metabolismo , Masculino , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Tapsigargina/farmacología
9.
Mol Med Rep ; 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31432114

RESUMEN

Contracture or diastolic dysfunction is a primary cause of injury following ischemia/reperfusion (IR). The present study examined whether Ca2+/calmodulin­dependent kinase II (CaMKII) is involved in contracture. Isolated rat hearts were subjected to either global IR or Ca2+ paradox (CaP), which is characterized by contracture. Left ventricular end­diastolic pressure, electron microscopy and troponin I  TnI) in coronary effluent were examined to indicate the extent of contracture. Compared with control hearts, both the IR and CaP groups exhibited an increase in necrosis and apoptosis, and a marked depression in contractile function. Western blot analysis showed that IR stimulated the phosphorylation (Thr287) and oxidation (Met281/282) of CaMKII, and the phosphorylation of phospholamban (PLN), a substrate of CaMKII. By contrast, only the phosphorylation of CaMKII was increased in the CaP group. Treatment with either 3 µM KN­62, an inhibitor of CaMKII, or 5 µM KB­R7943, an inhibitor of the Na+/Ca2+ exchanger, mitigated the damage and the post­translational modification of both CaMKII and PLN. Similar to the effect of the negative inotropic agent 2,3­butanedione­monoxime, the increased cell survival after treatment with KN­62 was associated with improved diastolic function. Examination using electron microscopy and a biochemical test showed the development of contraction bands, disruption of the sarcolemmal membrane and an increase in the release of TnI in both IR and CaP hearts; these results indicated the occurrence of contracture. Furthermore, these changes were inhibited by either KN­62 or KB­R7943. Taken together, these data provided evidence that CaMKII mediates reperfusion­elicited contracture, and that the activation of CaMKII via phosphorylation is involved in this process.

11.
Burns Trauma ; 6: 28, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30338266

RESUMEN

BACKGROUND: The molecular pattern of severe burn-induced acute lung injury, characterized by cell structure damage and leukocyte infiltration, remains unknown. This study aimed to determine whether calpain, a protease involved in both processes, mediates severe burn-induced acute lung injury. METHODS: Rats received full-thickness scald burns covering 30% of the total body surface area, followed by instant fluid resuscitation. MDL28170 (Tocris Bioscience), an inhibitor of calpain, was given intravenously 1 h before or after the scald burn. The histological score, wet/dry weight ratio, and caspase-3 activity were examined to evaluate the degree of lung damage. Calpain activity and its source were detected by an assay kit and immunofluorescence staining. The proteolysis of membrane skeleton proteins α-fodrin and ankyrin-B, which are substrates of calpain, was measured by Western blot. RESULTS: Time-course studies showed that tissue damage reached a peak between 1 and 6 h post-scald burn and gradually diminished at 24 h. More importantly, calpain activity reached peak levels at 1 h and was maintained until 24 h, paralleled by lung damage to some extent. Western blot showed that the levels of the proteolyzed forms of α-fodrin and ankyrin-B correlated well with the degree of damage. MDL28170 at a dose of 3 mg/kg b. w. given 1 h before burn injury not only antagonized the increase in calpain activity but also ameliorated scald burn-induced lung injury, including the degradation of α-fodrin and ankyrin-B. Immunofluorescence images revealed calpain 1 and CD45 double-positive cells in the lung tissue of rats exposed to scald burn injury, suggesting that leukocytes were a dominant source of calpain. Furthermore, this change was blocked by MDL28170. Finally, MDL28170 given at 1 h post-scald burn injury significantly ameliorated the wet/dry weight ratio compared with burn injury alone. CONCLUSIONS: Calpain, a product of infiltrating leukocytes, is a mediator of scald burn-induced acute lung injury that involves enhancement of inflammation and proteolysis of membrane skeleton proteins. Its late effects warrant further study.

12.
Eur J Pharmacol ; 563(1-3): 88-91, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17397825

RESUMEN

Arachidonic acid inhibits the activity of a number of different Cl- channels, however its molecular mechanism of action is not known. Here we show that inhibition of cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channels by arachidonic acid is weakened following mutagenesis of two positively charged pore-lining amino acids. Charge-neutralizing mutants K95Q and R303Q both increased the Kd for inhibition from approximately 3.5 microM in wild type to approximately 17 microM. At both sites, the effects of mutagenesis were dependent of the charge of the substituted side chain. We suggest that arachidonic acid interacts electrostatically with positively charged amino acid side chains in the cytoplasmic vestibule of the CFTR channel pore to block Cl- permeation.


Asunto(s)
Ácido Araquidónico/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Activación del Canal Iónico , Animales , Ácido Araquidónico/farmacología , Arginina , Línea Celular , Cloruros/metabolismo , Cricetinae , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Relación Dosis-Respuesta a Droga , Glutamina , Activación del Canal Iónico/efectos de los fármacos , Lisina , Potenciales de la Membrana , Mutación , Técnicas de Placa-Clamp , Transfección
13.
Eur J Pharmacol ; 561(1-3): 14-22, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-17306252

RESUMEN

This study determined the role of the reverse mode Na(+)/Ca(2+) exchanger (NCX) in cardioprotection of metabolic inhibition preconditioning in isolated ventricular myocyctes. Activity of the reverse mode NCX was assessed by changes of [Ca(2+)](i) upon withdrawal of extracellular Na(+). [Ca(2+)](i) was measured by spectrofluorometry, using Fura-2 as Ca(2+) indicator. The amplitude of contraction and exclusion of trypan blue by myocytes served as indices of contractile function and viability, respectively. Firstly, NCX activity significantly decreased during simulated reperfusion after severe metabolic inhibition (index ischaemia) in myocytes subjected to metabolic inhibition preconditioning. This inhibitory effect on NCX activity correlated with the enhancing effect of metabolic inhibition preconditioning on cell viability following ischaemic insult. Treatment myocytes with E4031, an activator of reverse mode NCX, during index ischaemia and reperfusion attenuated the enhancing effects of metabolic inhibition preconditioning on cell contraction and viability. Secondly, NCX activity was significantly higher at the end of metabolic inhibition preconditioning. More importantly, E4031 pretreatment mimicked the beneficial effects of metabolic inhibition preconditioning in myocytes and ischaemic preconditioning in the isolated perfused heart, respectively, and these effects were abolished by KB-R7943, an inhibitor of reverse mode NCX. The results indicate that increased reverse mode NCX activity during preconditioning triggered cardioprotection, and reduced reverse mode NCX activity during reperfusion after index ischaemia conferred cardioprotection.


Asunto(s)
Calcio/metabolismo , Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/fisiología , Animales , Antiarrítmicos/farmacología , Supervivencia Celular , Fura-2/análisis , Masculino , Contracción Miocárdica/efectos de los fármacos , Isquemia Miocárdica/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Espectrometría de Fluorescencia , Tiourea/análogos & derivados , Tiourea/farmacología , Función Ventricular/efectos de los fármacos , Función Ventricular/fisiología
14.
Eur J Pharmacol ; 572(1): 1-11, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17822695

RESUMEN

The pathogenesis of myocardial stunning caused by brief ischemia and reperfusion remains unclear. The aim of the present study was to investigate the underlying mechanism of myocardial stunning. An isolated cell model of myocardial stunning was firstly established in isolated rat ventricular myocytes exposed to 8 min of simulated ischemia and 30 min of reperfusion, the cardiomyocyte contractile function was used to evaluate myocardial stunning. A diastolic Ca(2+) overload without significant changes in systolic Ca(2+) and the amplitude of Ca(2+) transient during the first 10 min of reperfusion played an important role in the occurrence of myocardial stunning. Decreasing Ca(2+) entry into myocardial cells with low Ca(2+) reperfusion was a very efficient way to prevent myocardial stunning. Diastolic Ca(2+) overload was closely related to the reverse mode of Na(+)/Ca(2+) exchanger (NCX) rather than L-type Ca(2+) channel. The activity of the reverse mode of NCX was found significantly higher at the initial time of reperfusion, and KB-R7943, a selective inhibitor of the reverse mode of NCX, administered at first 10 min of reperfusion rather than at the time of ischemia significantly attenuated myocardial stunning. In addition, NCX inhibition also attenuated the Ca(2+) oscillation and cardiac dysfunction when field stimulus was stopped at first 10 min of reperfusion. These data suggest that one of the important mechanisms of triggering myocardial stunning is diastolic Ca(2+) overload caused by activation of the reverse mode of NCX of cardiomyocytes during the initial period of reperfusion following brief ischemia.


Asunto(s)
Calcio/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Aturdimiento Miocárdico/fisiopatología , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Canales de Calcio Tipo L/fisiología , Diástole , Técnicas In Vitro , Masculino , Contracción Miocárdica , Daño por Reperfusión Miocárdica/metabolismo , Aturdimiento Miocárdico/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Tiourea/administración & dosificación , Tiourea/análogos & derivados , Tiourea/farmacología
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(5): 633-8, 2016 May.
Artículo en Zh | MEDLINE | ID: mdl-27222176

RESUMEN

OBJECTIVE: To investigate the Effect of 2,3-butanedione monoxime (BDM) on calcium paradox-induced heart injury and its underlying mechanisms. METHODS: Thirty-two adult male SD rats were randomized into 4 groups, namely the control group, BDM treatment control group, calcium paradox group, and BDM treatment group. Isolated Sprague Dawley male rat hearts underwent Langendorff perfusion and the left ventricular pressure (LVP) and left ventricular end-diastolic pressure (LVEDP) were monitored. Left ventricular developed pressure (LVDP) was calculated to evaluate the myocardial performance. Lactate dehydrogenase (LDH) content in the coronary flow was determined. Triphenyltetrazolium chloride staining was used to measure the infarct size, and myocardial cell apoptosis was tested with TUNEL method. Western blotting was used to determine the expression of cleaved caspase-3 and cytochrome c. RESULTS: Compared with the control group, BDM at 20 mmol/L had no effect on cardiac performance, cell death, apoptotic index or the content of LDH, cleaved caspase-3 and cytochrome c at the end of perfusion under control conditions (P>0.05). Calcium paradox treatment significantly decreased the cardiac function and the level of LVDP and induced a larger infarct size (P<0.01), an increased myocardial apoptosis index (P<0.01), and up-regulated expressions of cleaved caspase-3 and cytochrome c (P<0.01). BDM (20 mmol/L) significantly attenuated these effects induced by calcium paradox, and markedly down-regulated the levels of LVEDP and LDH (P<0.01), lowered myocardial apoptosis index, decreased the content of cleaved caspase-3 and cytochrome c (P<0.01), increased LVDP, and reduced the infarct size (P<0.01). CONCLUSION: BDM suppresses cell apoptosis and contracture and improves heart function and cell survival in rat hearts exposed to calcium paradox, suggesting the value of BDM as an potential drug for myocardial ischemia reperfusion injur.


Asunto(s)
Calcio/efectos adversos , Diacetil/análogos & derivados , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Apoptosis , Caspasa 3/metabolismo , Citocromos c/metabolismo , Diacetil/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda
16.
Di Yi Jun Yi Da Xue Xue Bao ; 25(1): 30-2, 36, 2005 Jan.
Artículo en Zh | MEDLINE | ID: mdl-15683992

RESUMEN

OBJECTIVE: To develop a method for acquisition of efficient and stable retroviral packaging cells on the basis of fluorescence-activated cell sorting (FACS) technique. METHODS: PA317 cells were transfected by the recombinant retroviral vector pLEGFP via liposome, and the result of transfection was examined using fluorescent microscope. Polyclones and monoclone of the packaging cells were obtained with FACS and identified by PCR and reverse transcriptional PCR (RT-PCR). NIH3T3 cells were used to determine the virus titer in the supernatant. RESULTS: Examination under fluorescence microscope confirmed the success of cell transfection with the retrovirus, and polyclonal and monoclonal cells with efficient and stable expression were obtained after FACS. The inserted EGFP gene fragment could be amplified by PCR from the genomic DNA of the polyclonal and monoclonal cells and by RT-PCR from the retrovirus RNA in the supernatant of monoclonal cell culture and some of the monoclonal cell cultures (6/8). Determination of the virus titer in the cell culture supernatant showed efficient viral production by the cells. CONCLUSION: FACS is an efficient method for obtaining stable retroviral packaging cells.


Asunto(s)
Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Retroviridae/genética , Regulación Viral de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Retroviridae/fisiología , Ensayo de Placa Viral/métodos
17.
Int J Clin Exp Pathol ; 8(9): 10239-49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26617732

RESUMEN

BACKGROUND: Reverse-mode of the Na(+)/Ca(2+) exchanger (NCX) stimulation provides cardioprotective effects for the ischemic/reperfused heart during ischemic preconditioning (IP). This study was designed to test the hypothesis that pretreatment with an inhibitor of cardiac delayed-rectifying K(+) channel (IKr), E4031, increases reverse-mode of NCX activity, and triggers preconditioning against infarct size (IS) and arrhythmias caused by ischemia/reperfusion injury through mitoKCa channels. MATERIALS AND METHODS: In the isolated perfused rat heart, myocardial ischemia/reperfusion injury was created by occlusion of the left anterior descending coronary artery for 30 min followed by 120 min reperfusion. Two cycles of coronary occlusion for 5 min and reperfusion were performed, or pretreatment with E4031 or sevoflurane (Sevo) before the 30 min occlusion with the reversed-mode of NCX inhibitor (KB-R7943) or not. RESULTS: E4031 or Sevo preconditioning not only markedly decreased IS but also reduced arrhythmias, which was significantly blunted by KB-R7943. Furthermore, these effects of E4031 preconditioning on IS and arrhythmias were abolished by inhibition of the mitoKCa channels. Similarly, pretreatment with NS1619, an opener of the mitoKCa channels, for 10 min before occlusion reduced both the infarct size and arrhythmias caused by ischemia/reperfusion. However, these effects weren't affected by blockade of the NCX with KB-R7943. CONCLUSION: Taken together, these preliminary results conclude that pretreatment with E4031 reduces infarct size and produces anti-arrhythmic effect via stimulating the reverse-mode NCX, and that the mitoKCa channels mediate the protective effects.


Asunto(s)
Precondicionamiento Isquémico , Mitocondrias/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Canales de Potasio Calcio-Activados/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Antiarrítmicos/farmacología , Arritmias Cardíacas/metabolismo , Masculino , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Piperidinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley
18.
Br J Pharmacol ; 135(7): 1675-84, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11934807

RESUMEN

The role of protein kinase C-epsilon (PKC-epsilon) in the development of kappa-opioid receptor (kappa-OR) tolerance to the effects of trans-(+/-)-3,4-dichloro-N-methyl-N-(2-[1-pyrrolidinyl]cyclohexyl) (U50,488H), the selective agonist of kappa-OR, was determined in rat ventricular myocytes. Incubation of ventricular myocytes with 1 microM U50,488H for 24 h significantly attenuated the inhibitory effects of 30 microM U50,488H on the electrically-induced [Ca(2+)](i) transient and forskolin-stimulated cyclic AMP accumulation, indicating the development of tolerance to the kappa-OR agonist. Chronic treatment of ventricular myocytes with U50,488H also induced translocation of PKC-epsilon to the particulate fraction. On the other hand, administration of 30 microM U50,488H for 10 min induced translocation of PKC-alpha to the particulate fraction in naïve ventricular myocytes, but not in cells pretreated with 1 microM U50,488H for 24 h. In ventricular myocytes incubated for 24 h with 1 microM U50,488H together with 1 microM chelerythrine or 1 microM GF109203X, PKC inhibitors, or 0.1 microM epsilonV1-2 peptide, a selective inhibitor of PKC-epsilon, 30 microM U50,488H still produced the inhibitory effect on the electrically-induced [Ca(2+)](i) transient as it did in naïve ventricular myocytes. Chronic treatment of ventricular myocytes with U50,488H and chelerythrine also attenuated the development of tolerance to acute U50,488H on cyclic AMP accumulation. Cells exposed to chelerythrine, GF109203X, or epsilonV1-2 peptide alone did not show an altered [Ca(2+)](i) response to U50,488H. These results indicate that activation of PKC-epsilon is a critical step in the development of tolerance in the kappa-OR.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Isoenzimas/metabolismo , Miocardio/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Receptores Opioides kappa/metabolismo , Alcaloides , Animales , Benzofenantridinas , Transporte Biológico , Calcio/metabolismo , Colforsina/farmacología , AMP Cíclico/metabolismo , Interacciones Farmacológicas , Tolerancia a Medicamentos , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Immunoblotting , Técnicas In Vitro , Indoles/farmacología , Masculino , Maleimidas/farmacología , Fragmentos de Péptidos/farmacología , Fenantridinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
19.
Br J Pharmacol ; 140(4): 750-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14534156

RESUMEN

The effect of preconditioning with U50488 H (UP), a selective kappa-opioid receptor (kappa-OR) agonist, on infarct size and intracellular Ca2+ ([Ca2+]i) in the heart subjected to ischaemic insults were studied and evaluated. U50488 H administered intravenously reduced the infarct size 18-48 h after administration in isolated hearts subjected to regional ischaemia/reperfusion (I/R). The effect was dose dependent. A peak effect was reached at 10 mg x kg-1 U50488 H and at 24 h after administration. The effect of 10 mg x kg-1 U50488 H at 24 h after administration was abolished by nor-binaltorphimine (nor-BNI), a selective kappa-OR antagonist, indicating the effect was kappa-OR mediated. The infarct reducing effect of U50488 H was attenuated when a selective blocker of mitochondrial (5-hydroxydecanoic acid, 5-HD) or sarcolemmal (HRM-1098) ATP-sensitive potassium channel (KATP) was coadministered with U50488 H 24 h before ischaemia or when 5-HD was administered just before ischaemia. U50488 H also attenuated the elevation in [Ca2+]i and reduction in electrically induced [Ca2+]i transient in cardiomyocytes subjected to ischaemic insults. The effects were reversed by blockade of KATP channel, which abolished the protective effect of preconditioning with U50488 H. The results indicated that mitochondrial KATP channel serves as both a trigger and a mediator, while sarcolemmal KATP channel as a trigger only, of delayed cardioprotection of kappa-OR stimulation. The effects of these channels may result from prevention/attenuation of [Ca2+]i overload induced by ischaemic insults.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Calcio/metabolismo , Corazón/fisiología , Naltrexona/análogos & derivados , Canales de Potasio/fisiología , Receptores Opioides kappa/efectos de los fármacos , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/antagonistas & inhibidores , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/uso terapéutico , Animales , Calcio/antagonistas & inhibidores , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/lesiones , Relación Dosis-Respuesta a Droga , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inyecciones Intravenosas , Precondicionamiento Isquémico Miocárdico/métodos , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Naltrexona/farmacología , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/clasificación , Canales de Potasio/efectos de los fármacos , Ratas/fisiología , Ratas Sprague-Dawley , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/metabolismo , Factores de Tiempo
20.
Eur J Pharmacol ; 473(2-3): 97-103, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12892826

RESUMEN

The effects of trans-(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]-benzeneacetamide methanesulfonate salt (U50,488H), a selective kappa-opioid receptor agonist, on transient outward K+ current (Ito1) and ultra-rapid delayed rectifier K+ current (IKur) in young human atrial myocytes were evaluated with a whole-cell patch-clamp technique. At +10 mV, U50,488H decreased Ito1 in a concentration-dependent manner (IC50=12.4+/-3.5 microM), while at +50 mV, U50,488H produced biphasic effects on Ito1-increasing and decreasing the current at 1-3 and 10-30 microM, respectively. U50,488H at 10 microM shifted the midpoint (V0.5) of Ito1 activation in a depolarizing direction by approximately 5 mV, accelerated the inactivation, and slowed the recovery from inactivation of Ito1. In addition, U50,488H inhibited IKur in a concentration-dependent manner (IC50=3.3+/-0.6 microM). The effects of U50,488H on the two types of K+ currents were not antagonized by either 5 microM nor-binaltorphimine or 300 nM naloxone. These results indicate that U50,488H affects both Ito1 and IKur in young human atrial myocytes in an opioid receptor-independent manner.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Miocitos Cardíacos/efectos de los fármacos , Naltrexona/análogos & derivados , Canales de Potasio de Dominio Poro en Tándem , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/efectos de los fármacos , Adolescente , Niño , Preescolar , Canales de Potasio de Tipo Rectificador Tardío , Femenino , Atrios Cardíacos , Humanos , Lactante , Masculino , Miocitos Cardíacos/fisiología , Naloxona/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Técnicas de Placa-Clamp , Canales de Potasio/fisiología , Receptores Opioides kappa/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA