Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Braz. J. Anesth. (Impr.) ; 72(6): 768-773, Nov.-Dec. 2022. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1420611

RESUMO

Abstract Background Dexmedetomidine (Dex) is widely used, and its most common side effect is bradycardia. The complete mechanism through which Dex induces bradycardia has not been elucidated. This research investigates the expression of gap junction proteins Connexin30.2 (Cx30.2) and Connexin40 (Cx40) within the sinoatrial node of rats with Dex-induced sinus bradycardia. Methods Eighty rats were randomly assigned to five groups. Saline was administered to rats in Group C. In the other four groups, the rats were administered Dex to induce bradycardia. In groups D1and D2, the rats were administered Dex at a loading dose of 30 μg.kg−1 and 100 μg.kg−1 for 10 min, then at 15 μg.kg−1.h−1 and 50 μg.kg−1.h−1 for 120 min separately. The rats in group D1A and D2A were administered Dex in the same way as in group D1and D2; however, immediately after the administration of the loading dose, 0.5 mg atropine was administered intravenously, and then at 0.5 mg.kg−1.h−1 for 120 min. The sinoatrial node was acquired after intravenous infusion was completed. Quantitative real-time polymerase chain reaction and western blot analyses were performed to measure mRNA and protein expression of Cx30.2 and Cx40, respectively. Results The expression of Cx30.2 increased, whereas the expression of Cx40 decreased within the sinoatrial node of rats with Dex-induced sinus bradycardia. Atropine reversed the effects of Dex on the expression of gap junction proteins. Conclusion Dex possibly altered the expression of gap junction proteins to slow down cardiac conduction velocity in the sinoatrial node.


Assuntos
Animais , Ratos , Nó Sinoatrial/metabolismo , Dexmedetomidina , Arritmias Cardíacas , Derivados da Atropina/metabolismo , Bradicardia/induzido quimicamente , Conexinas/genética , Conexinas/metabolismo
2.
Braz J Anesthesiol ; 72(6): 768-773, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35618083

RESUMO

BACKGROUND: Dexmedetomidine (Dex) is widely used, and its most common side effect is bradycardia. The complete mechanism through which Dex induces bradycardia has not been elucidated. This research investigates the expression of gap junction proteins Connexin30.2 (Cx30.2) and Connexin40 (Cx40) within the sinoatrial node of rats with Dex-induced sinus bradycardia. METHODS: Eighty rats were randomly assigned to five groups. Saline was administered to rats in Group C. In the other four groups, the rats were administered Dex to induce bradycardia. In groups D1 and D2, the rats were administered Dex at a loading dose of 30 µg.kg-1 and 100 µg.kg-1 for 10 min, then at 15 µg.kg-1.h-1 and 50 µg.kg-1.h-1 for 120 min separately. The rats in group D1A and D2A were administered Dex in the same way as in group D1 and D2; however, immediately after the administration of the loading dose, 0.5 mg atropine was administered intravenously, and then at 0.5 mg.kg-1.h-1 for 120 min. The sinoatrial node was acquired after intravenous infusion was completed. Quantitative real-time polymerase chain reaction and western blot analyses were performed to measure mRNA and protein expression of Cx30.2 and Cx40, respectively. RESULTS: The expression of Cx30.2 increased, whereas the expression of Cx40 decreased within the sinoatrial node of rats with Dex-induced sinus bradycardia. Atropine reversed the effects of Dex on the expression of gap junction proteins. CONCLUSION: Dex possibly altered the expression of gap junction proteins to slow down cardiac conduction velocity in the sinoatrial node.


Assuntos
Dexmedetomidina , Nó Sinoatrial , Ratos , Animais , Nó Sinoatrial/metabolismo , Bradicardia/induzido quimicamente , Conexinas/genética , Conexinas/metabolismo , Arritmias Cardíacas , Derivados da Atropina/metabolismo
3.
Int Immunopharmacol ; 86: 106768, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32679539

RESUMO

OBJECTIVE: Dexmedetomidine (DEX) was reported to protect heart against ischemic-reperfusion (IR) but the mechanism herein remains elusive. This study aims to explore the mechanism of DEX on pyroptosis induced by myocardial ischemic reperfusion (MIR). METHODS: MIR rat models were established and injected DEX or miR-29b agomir/antagomir separately. The possible effect of DEX or miR-29b on myocardial cells was assessed according to measurement on creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), interleukin-1ß (IL-1ß) and interleukin-18 (IL-18), myocardial infarction size, myocardial injury and apoptosis. Western blot determined the expression levels of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC) and cleaved-caspase-1. Hypoxia/reoxygenation (H/R) cell model was established. The lactate dehydrogenase (LDH) content released by myocardial cells was examined. The relation between miR-29b and FoxO3a was confirmed by dual luciferase reporter gene assay. FoxO3a or ARC level was elevated in H/R myocardial cells to detect its effect on pyroptosis. RESULTS: MIR rat models were successfully established, in which cell pyroptosis was triggered as evidenced by increased expression levels of NLRP3, ASC and cleaved-caspase-1. Rats with DEX precondition had attenuated cell pyroptosis and ameliorated inflammatory response. FoxO3a was a target of miR-29b. MiR-29b agomir or miR-29b antagomir could inhibit or promote the protective effect of DEX on MIR. Overexpression of FoxO3a/ARC axis could suppress myocardial pyroptosis induced by H/R. CONCLUSION: DEX could ameliorate MIR injury (MIRI) and H/R injury in rats and inhibit H/R induced pyroptosis in myocardial cells via down-regulating miR-29b to activate FoxO3a/ARC axis.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Dexmedetomidina/uso terapêutico , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miocárdio/patologia , Substâncias Protetoras/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Linhagem Celular , Modelos Animais de Doenças , Regulação para Baixo , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Masculino , Miocárdio/metabolismo , Piroptose/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
4.
Med Sci Monit ; 25: 1769-1779, 2019 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-30848248

RESUMO

BACKGROUND Cardiac remote ischemic conditioning (RIC) is a noninvasive cardioprotective method in ischemia-reperfusion injury and acute myocardial infarction (AMI). The aims of this study were to investigate the effects of RIC in a rat model of AMI. MATERIAL AND METHODS Adult male Sprague-Dawley rats included the AMI group that underwent ligation of the left anterior descending (LAD) coronary artery (n=24), the RIC group that consisted the AMI rat model treated with RIC once daily in the left hind limb until days 1, 7 and 14 (n=24), and the sham group (n=24). Myocardial infarct size was measured by routine histology with triphenyltetrazolium chloride (TTC) and Masson's trichrome histochemical staining for myocardial necrosis and fibrosis, respectively. Serum levels of Bcl-2, Bax, caspase-3, and inducible nitric oxide synthase (iNOS) were measured by enzyme-linked immunosorbent assay (ELISA). The apoptosis index was detected using the TUNEL assay. Spectrophotometry of the myocardium was used to identify mitochondrial complexes and myocardial ATP. RESULTS The RIC group showed improved cardiac hemodynamics, reduced the size of the myocardial infarction, upregulated expression of Bcl-2, and down-regulation of the levels of Bax, caspase-3, and iNOS, and reduced cardiac myocyte apoptosis and inhibited the opening of the mitochondrial permeability transition pore (MPTP). CONCLUSIONS In a rat model of AMI, RIC improved the hemodynamic index, reduce the levels of apoptosis and myocardial injury, and improved mitochondrial function.


Assuntos
Precondicionamento Isquêmico/métodos , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Cardiotônicos , Caspase 3/análise , Caspase 3/sangue , Modelos Animais de Doenças , Traumatismos Cardíacos/prevenção & controle , Hemodinâmica , Masculino , Mitocôndrias/metabolismo , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/sangue , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/terapia , Proteína X Associada a bcl-2/análise , Proteína X Associada a bcl-2/sangue
5.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 34(2): 106-110, 2018 Feb 08.
Artigo em Chinês | MEDLINE | ID: mdl-29926671

RESUMO

OBJECTIVES: To investigate the effect of taurine magnesium coordination compound (TMCC) on torsades de pointes (TdP) in isolated guinea pig hearts. METHODS: Healthy male guinea pigs weighting 250~300 g were randomly divided into 4 groups:①TdP model group (n=7):Isolated hearts were perfused by normal K-H solution 20 minutes, then perfused by slowly activated delayed rectifier potassium current(IKs) blocker 10µmol/L Chromanol 293B under hypokalemic solution(1.8 mmol/L) to establish TdP model;②~④ TdP model + TMCC group (n=6):Isolated hearts were perfused by normal K-H solution for 20 minutes, then perfused by IKs blocker 10µmol/L Chromanol 293B under hypokalemic solution(1.8 mmol/L) for 60 minutes, at the same time TMCC which concentration was 1, 2, 4 mmol/L was administered respectively by Langendorff retrograde aortic perfusion method. Cardiac surface electrocardiogram of guinea pigs in vitro was collected and recorded by Biopac electrophysiological recorder. Incidence of TdP, transmural dispersion of repolarization (TDR), instability of QT interval were acquired from Lead Ⅱ electrocardiograph (ECG) wave forms to describe the effect of TMCC on TdP model. Datas were acquired at the time of 20 min and pre-TdP, in case there was no TdP observed, a value of 60 min was entered for calculation purpose. RESULTS: Incidence of TdP in TdP model group was 6/7. TdP incidence could be decreased significantly by 1, 2, 4 mmol/L TMCC, and was 5/6, 1/6, 0/6 respectively. Compared with the pre-drug, Chromanol 293B under hypokalemic solution in TdP model group increased TDR(corrected) evidently(P<0.01). Compared with the pre-drug, 1, 2, 4 mmol/L TMCC in TdP model + TMCC group could decrease the increased TDR(corrected) induced by Chromanol 293B under hypokalemic solution(P>0.05). Compared with the TdP model group, 2, 4 mmol/L TMCC could evidently decrease the instability of QT interval induced by Chromanol 293B under hypokalemic solution(P<0.05). During the establishment of TdP model, P waves in more than one cardiac cycle continuously were disappeared in ECG. However, P wave could always be seen independent in ECG acquired from TdP model + TMCC group. CONCLUSIONS: TMCC can play the role against TdP through decreasing TDR and instability of QT interval, and inhibiting early after depolarization(EAD).


Assuntos
Antiarrítmicos/farmacologia , Magnésio/farmacologia , Taurina/farmacologia , Torsades de Pointes/tratamento farmacológico , Animais , Eletrocardiografia , Cobaias , Técnicas In Vitro , Síndrome do QT Longo , Masculino , Distribuição Aleatória
6.
Acta Pharmacol Sin ; 39(3): 382-392, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29072257

RESUMO

Short QT syndrome (SQTS) is a genetic arrhythmogenic disease that can cause malignant arrhythmia and sudden cardiac death. The current therapies for SQTS have application restrictions. We previously found that Mg· (NH2CH2CH2SO3)2· H2O, a taurine-magnesium coordination compound (TMCC) exerted anti-arrhythmic effects with low toxicity. In this study we established 3 different models to assess the potential anti-arrhythmic effects of TMCC on type 2 short QT syndrome (SQT2). In Langendorff guinea pig-perfused hearts, perfusion of pinacidil (20 µmol/L) significantly shortened the QT interval and QTpeak and increased rTp-Te (P<0.05 vs control). Subsequently, perfusion of TMCC (1-4 mmol/L) dose-dependently increased the QT interval and QTpeak (P<0.01 vs pinacidil). TMCC perfusion also reversed the rTp-Te value to the normal range. In guinea pig ventricular myocytes, perfusion of trapidil (1 mmol/L) significantly shortened the action potential duration at 50% (APD50) and 90% repolarization (APD90), which was significantly reversed by TMCC (0.01-1 mmol/L, P<0.05 vs trapidil). In HEK293 cells that stably expressed the outward delayed rectifier potassium channels (IKs), perfusion of TMCC (0.01-1 mmol/L) dose-dependently inhibited the IKs current with an IC50 value of 201.1 µmol/L. The present study provides evidence that TMCC can extend the repolarization period and inhibit the repolarizing current, IKs, thereby representing a therapeutic candidate for ventricular arrhythmia in SQT2.


Assuntos
Arritmias Cardíacas/prevenção & controle , Complexos de Coordenação/farmacologia , Sistema de Condução Cardíaco/anormalidades , Cardiopatias Congênitas/prevenção & controle , Magnésio/farmacologia , Taurina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/induzido quimicamente , Células Cultivadas , Cobaias , Cardiopatias Congênitas/induzido quimicamente , Humanos , Magnésio/química , Modelos Teóricos , Miócitos Cardíacos/fisiologia , Pinacidil/antagonistas & inibidores , Pinacidil/farmacologia , Taurina/química , Trapidil/antagonistas & inibidores , Trapidil/farmacologia
7.
Mol Med Rep ; 16(4): 4259-4264, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28765943

RESUMO

It has been previously demonstrated that taurine magnesium coordination compound (TMCC) produces antiarrhythmic effects in vivo. The present study investigated the acute and chronic effect of TMCC on sodium channels in HEK cells stably expressing human cardiac Nav1.5 sodium channels. The current amplitude, activation and inactivation kinetics, recovery time from inactivation, and use­dependent block of sodium channels were analyzed using the whole­cell patch clamp technique. Western blotting was used to analyze Nav1.5 expression following chronic TMCC treatment. In HEK cells expressing Nav1.5 channels, TMCC acutely inhibited Na+ currents in a dose­dependent manner. In addition, acute application of TMCC shifted the activation and inactivation curves, and prolonged the recovery time from inactivation, but did not exhibit a use­dependent block of Nav1.5. By contrast, chronic TMCC treatment only produced a use­dependent block of Nav1.5 and downregulated Nav1.5 expression. The results of the present study suggested that TMCC may produce antiarrhythmic actions via acute inhibition of sodium channel currents and chronic downregulation of Nav1.5 expression.


Assuntos
Complexos de Coordenação/farmacologia , Magnésio/farmacologia , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Taurina/farmacologia , Células HEK293 , Humanos
8.
Biochem Biophys Res Commun ; 488(2): 278-284, 2017 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-28479248

RESUMO

Recent studies have demonstrated that remote ischemic conditioning (RIC) creates cardioprotection against ischemia/reperfusion injury and myocardial infarction (MI); however, the effects of non-invasive remote ischemic conditioning (nRIC) on prognosis and rehabilitation after MI (post-MI) remain unknown. We successfully established MI models involving healthy adult male Sprague-Dawley rats. The nRIC group repeatedly underwent 5 min of ischemia and 5 min of reperfusion in the left hind limb for three cycles every other day until weeks 4, 6, and 8 after MI. nRIC improved cardiac hemodynamic function and mitochondrial respiratory function through increasing myocardial levels of mitochondrial respiratory chain complexes I, II, III, IV, and adenosine triphosphate (ATP) and decreasing the activity of nitric oxide synthase (NOS). nRIC could inhibit cardiomyocytes apoptosis and reduce myocardium injury through raising the expression of Bcl-2 and reduced the content of creatine kinase-MB, cardiac troponin I and Bax. The results indicated that long-term nRIC could accelerate recovery and improve prognosis and rehabilitation in post-MI rats.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/reabilitação , Infarto do Miocárdio/terapia , Animais , Masculino , Infarto do Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 32(3): 209-213, 2016 Mar 08.
Artigo em Chinês | MEDLINE | ID: mdl-29931878

RESUMO

OBJECTIVE: To investigate the inotropic effects of dioscin (Dio)in rat isolated-heart and intracellular free calcium concentration in isolated rat ventricular myocytes and to explore its mechanism preliminarily. METHODS: Left ventricle contractile function was measured using the Langendorff non-recirculating mode of isolated rat heart perfusion. Effects of dioscin and dioscin+SEA0400, sodium-calcium exchanger (NCX) inhibitor, were investigated by measuring left ventricular systolic pressure (LVSP) and left ventricular end diastolic pressure (LVEDP). Also, heart rate (HR), peak rate of rise/fall of left ventricular pressure (±dp/dtmax) of isolated rat heart were calculated; Effects of dioscin and SEA0400 on intracellular free calcium concentration in rat H9c2 cells were measured by Fluo3-AM and then detected the fluorescence intensity with confocal microscopy. RESULTS: With 1 µmol/L dioscin, LVSP was significantly increased by 19.7% (P<0.01) and dp/dtmax was increased by 9.6%; With 1 µmol/L dioscin, the relative fluorescence intensity of intracellular free calcium concentrations were strong significantly(P<0.01). While in presence of SEA0400, the relative fluorescence intensity was changed to 17.09±0.63 (P<0.01) by 1 µmol/L dioscin. With 1 µmol/L dioscin, the relative fluorescence intensity was weak(P<0.01) without calcium or sodium in the extracellular fluid. CONCLUSIONS: Dioscin shows positive inotropic effect on isolated rat heart, enhancing the LVSP and +dp/dtmax; Dioscin increases the intracellular concentration of Ca2+ in the cardiac myocytes by increasing Na+ influx and facilitating the reverse mode of the sodium-calcium exchanger.


Assuntos
Diosgenina/análogos & derivados , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo , Compostos de Anilina/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Diosgenina/farmacologia , Miócitos Cardíacos/metabolismo , Éteres Fenílicos/farmacologia , Ratos , Sódio/metabolismo
10.
J Surg Res ; 164(1): 162-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726056

RESUMO

BACKGROUND: Transient limb ischemia induces remote early preconditioning that protects the myocardium from ischemia/reperfusion (I/R). However, it is unknown whether limb ischemia induces remote late preconditioning and whether it induces the same magnitude of cardioprotection compared with cardial ischemic preconditioning (CIP). We tested the hypothesis that late remote preconditioning of noninvasive limb ischemia (NLIP) offers the same magnitude of cardioprotection against myocardium I/R injury. METHODS: Thirty Wistar rats weighing 240-260 g each were randomly divided into three groups: I/R, CIP, and NLIP. The mean arterial pressure (MAP), heart rate (HR), ST-segment, ventricular arrhythmia, and CK-MB, cTnI, and superoxide dismutase (SOD) activity were measured after 0 and 30 min of ischemia and after 120 min of reperfusion. Myocardial infarct size, histologic examination, MMP-2, MMP-9, and TIMP-1 protein expression were determined at the end of the experiment. RESULTS: Compared with I/R groups, CIP and NLIP reduced ST-segment elevation (P<0.01), decreased incidence and duration of ventricular arrhythmia (P<0.01) during ischemia, decreased CK-MB (P<0.05), and cTnI (P<0.01) activity, and increased SOD (P<0.05) activity after reperfusion. The myocardial infarct size (P<0.01) was significantly reduced, and cell injury was attenuated in the CIP and NLIP groups compared with the I/R group. MMP-2 and MMP-9 protein expression was significantly decreased in the CIP and NLIP groups (P<0.01), while TIMP-1 expression was significantly increased in the CIP and NLIP groups compared with the I/R group (P<0.01). CONCLUSION: Remote preconditioning via NLIP has late cardioprotection against myocardium I/R injury and has a similar magnitude of cardioprotection compared with CIP in rats.


Assuntos
Membro Posterior/irrigação sanguínea , Precondicionamento Isquêmico Miocárdico/métodos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Pressão Sanguínea , Creatina Quinase Forma MB/sangue , Eletrocardiografia , Frequência Cardíaca , Masculino , Metaloproteinase 2 da Matriz/sangue , Metaloproteinase 9 da Matriz/sangue , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Ratos , Ratos Wistar , Superóxido Dismutase/sangue , Inibidor Tecidual de Metaloproteinase-1/sangue , Troponina I/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...