Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Clinics (Sao Paulo) ; 79: 100410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38901133

RESUMEN

BACKGROUND: Cuproptosis is known to regulate diverse physiological functions in many diseases, but its role in regulating Myocardial Ischemia-Reperfusion Injury (MI/RI) remains unclear. METHODS: For this purpose, the MI/RI microarray datasets GSE61592 were downloaded from the Gene Expression Omnibus (GEO) database, and the Differently Expressed Genes (DEGs) in MI/RI were identified using R software. Moreover, the MI/RI mice model was established to confirm further the diagnostic value of Pyruvate Dehydrogenase B (Pdhb), Dihydrolipoamide S-acetyltransferase (Dlat), and Pyruvate dehydrogenase E1 subunit alpha 1 (Pdhα1). RESULTS: The analysis of microarray datasets GSE61592 revealed that 798 genes were upregulated and 768 were downregulated in the myocardial tissue of the ischemia-reperfusion injury mice. Furthermore, Dlat, Pdhb, Pdhα1, and cuproptosis-related genes belonged to the downregulated genes. The receiver operating characteristics curve analysis results indicated that the Dlat, Pdhb, and Pdhα1 levels were downregulated in MI/RI and were found to be potential biomarkers for MI/RI diagnosis and prognosis. Similarly, analysis of Dlat, Pdhb, and Pdhα1 levels in the MI/RI mice revealed Pdhb being the key diagnostic marker. CONCLUSIONS: This study demonstrated the prognostic value of cuproptosis-related genes (Dlat, Pdhb, and Pdhα1), especially Pdhb, MI/RI, providing new insight into the MI/RI treatment.


Asunto(s)
Biología Computacional , Daño por Reperfusión Miocárdica , Animales , Daño por Reperfusión Miocárdica/genética , Ratones , Regulación hacia Abajo/genética , Masculino , Modelos Animales de Enfermedad , Regulación hacia Arriba , Ratones Endogámicos C57BL , Perfilación de la Expresión Génica/métodos , Piruvato Deshidrogenasa (Lipoamida)/genética , Biomarcadores/análisis , Acetiltransferasas/genética
2.
Clinics ; Clinics;79: 100410, 2024. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1569135

RESUMEN

Abstract Background: Cuproptosis is known to regulate diverse physiological functions in many diseases, but its role in regulating Myocardial Ischemia-Reperfusion Injury (MI/RI) remains unclear. Methods: For this purpose, the MI/RI microarray datasets GSE61592 were downloaded from the Gene Expression Omnibus (GEO) database, and the Differently Expressed Genes (DEGs) in MI/RI were identified using R software. Moreover, the MI/RI mice model was established to confirm further the diagnostic value of Pyruvate Dehydrogenase B (Pdhb), Dihydrolipoamide S-acetyltransferase (Dlat), and Pyruvate dehydrogenase E1 subunit alpha 1 (Pdhα1). Results: The analysis of microarray datasets GSE61592 revealed that 798 genes were upregulated and 768 were downregulated in the myocardial tissue of the ischemia-reperfusion injury mice. Furthermore, Dlat, Pdhb, Pdhα1, and cuproptosis-related genes belonged to the downregulated genes. The receiver operating characteristics curve analysis results indicated that the Dlat, Pdhb, and Pdhα1 levels were downregulated in MI/RI and were found to be potential biomarkers for MI/RI diagnosis and prognosis. Similarly, analysis of Dlat, Pdhb, and Pdhα1 levels in the MI/RI mice revealed Pdhb being the key diagnostic marker. Conclusions: This study demonstrated the prognostic value of cuproptosis-related genes (Dlat, Pdhb, and Pdhα1), especially Pdhb, MI/RI, providing new insight into the MI/RI treatment.

3.
Braz. J. Pharm. Sci. (Online) ; 59: e23002, 2023. graf
Artículo en Inglés | LILACS | ID: biblio-1520312

RESUMEN

Abstract This study aimed to investigate the role and signaling pathways of β3-AR in myocardial ischemia/reperfusion (I/R) injury, which is one of the leading causes of death worldwide. 47 male rats were randomly divided into two main groups to evaluate infarct size and molecular parameters. Rats in both groups were randomly divided into 4 groups. Control (sham), I/R (30 min ischemia/120 min reperfusion), BRL37344 (BRL) (A) (5 µg/kg single-dose pre-treatment (preT) before I/R) and BRL (B) (5 µg/kg/day preT for 10 days before I/R). Infarct size was determined with triphenyltetrazolium chloride staining and analyzed with ImageJ program. The levels of AMPK, SIRT1, mTOR, and p70SK6 responsible for cellular energy and autophagy were evaluated by western blot. Infarct size increased in the I/R group (44.84 ± 1.47%) and reduced in the single-dose and 10-day BRL-treated groups (32.22 ± 1.57%, 29.65 ± 0.55%; respectively). AMPK and SIRT1 levels were decreased by I/R but improved in the treatment groups. While mTOR and p70S6K levels increased in the I/R group, they decreased with BRL preT. BRL preT protects the heart against I/R injury. These beneficial effects are mediated in part by activation of AMPK and SIRT1, inhibition of mTOR and p70S6K, and consequently protected autophagy.


Asunto(s)
Animales , Masculino , Ratas , Autofagia , Daño por Reperfusión Miocárdica/patología , Agonistas Adrenérgicos , Isquemia/patología , Western Blotting/métodos , Isquemia Miocárdica/patología , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Sirtuina 1/clasificación , Corazón/fisiopatología , Infarto
4.
Rev. bras. cir. cardiovasc ; Rev. bras. cir. cardiovasc;37(3): 370-379, May-June 2022. tab, graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1376533

RESUMEN

ABSTRACT Introduction: The objective of this study is to investigate the protective mechanism of dexmedetomidine (Dex) in myocardial ischemia/reperfusion (MIR)-induced acute lung injury (ALI) of diabetic rats by inhibiting hypoxia-inducible factor-1α (HIF-1α). Methods: Initially, healthy male Sprague Dawley rats were treated with streptozocin to induce diabetes. Then, three weeks after the induction, Dex or lentiviral vector (LV)-HIF-1α was injected into the rats 30 minutes prior to the MIR modeling. After four weeks, lung tissues were harvested for pathological changes observation and the wet/dry weight (W/D) ratio determination. Afterwards, oxidative stress indicators and pro-inflammatory factors were measured. In addition, HIF-1α expression was assessed by immunohistochemistry and western blot analysis. Results: Dex could suppress inflammatory cell infiltration, improve lung tissue structure, reduce pathological score and the W/D ratio, and block oxidative stress and inflammatory response in MIR-induced ALI of diabetic rats. Besides, Dex could also inhibit HIF-1α expression. Moreover, Dex + LV-HIF-1α reversed the protective role of Dex on diabetic MIR-induced ALI. Conclusion: Our study has made it clear that Dex inhibited the upregulation of HIF-1α in diabetic MIR-induced ALI, and thus protect lung functions by quenching the accumulation of oxygen radical and reducing lung inflammatory response.

5.
Braz J Cardiovasc Surg ; 37(3): 370-379, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35605218

RESUMEN

INTRODUCTION: The objective of this study is to investigate the protective mechanism of dexmedetomidine (Dex) in myocardial ischemia/reperfusion (MIR)-induced acute lung injury (ALI) of diabetic rats by inhibiting hypoxia-inducible factor-1α (HIF-1α). METHODS: Initially, healthy male Sprague Dawley rats were treated with streptozocin to induce diabetes. Then, three weeks after the induction, Dex or lentiviral vector (LV)-HIF-1α was injected into the rats 30 minutes prior to the MIR modeling. After four weeks, lung tissues were harvested for pathological changes observation and the wet/dry weight (W/D) ratio determination. Afterwards, oxidative stress indicators and pro-inflammatory factors were measured. In addition, HIF-1α expression was assessed by immunohistochemistry and western blot analysis. RESULTS: Dex could suppress inflammatory cell infiltration, improve lung tissue structure, reduce pathological score and the W/D ratio, and block oxidative stress and inflammatory response in MIR-induced ALI of diabetic rats. Besides, Dex could also inhibit HIF-1α expression. Moreover, Dex + LV-HIF-1α reversed the protective role of Dex on diabetic MIR-induced ALI. CONCLUSION: Our study has made it clear that Dex inhibited the upregulation of HIF-1α in diabetic MIR-induced ALI, and thus protect lung functions by quenching the accumulation of oxygen radical and reducing lung inflammatory response.


Asunto(s)
Lesión Pulmonar Aguda , Dexmedetomidina , Diabetes Mellitus Experimental , Daño por Reperfusión Miocárdica , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/prevención & control , Animales , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/patología , Masculino , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Ratas , Ratas Sprague-Dawley , Transducción de Señal
6.
Biomedicines ; 11(1)2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36672525

RESUMEN

Ischemia/reperfusion injury is a process associated with cardiologic interventions, such as percutaneous coronary angioplasty after an acute myocardial infarction. Blood flow restoration causes a quick burst of reactive oxygen species (ROS), which generates multiple organelle damage, leading to the activation of cell death pathways. Therefore, the intervention contributes to a greater necrotic zone, thus increasing the risk of cardiovascular complications. A major cardiovascular ROS source in this setting is the activation of multiple NADPH oxidases, which could result via the occupancy of type 1 angiotensin II receptors (AT1R); hence, the renin angiotensin system (RAS) is associated with the generation of ROS during reperfusion. In addition, ROS can promote the expression of NF-κΒ, a proinflammatory transcription factor. Recent studies have described an intracellular RAS pathway that is associated with increased intramitochondrial ROS through the action of isoform NOX4 of NADPH oxidase, thereby contributing to mitochondrial dysfunction. On the other hand, the angiotensin II/ angiotensin type 2 receptor (Ang II/AT2R) axis exerts its effects by counter-modulating the action of AT1R, by activating endothelial nitric oxide synthase (eNOS) and stimulating cardioprotective pathways such as akt. The aim of this review is to discuss the possible use of AT1R blockers to hamper both the Ang II/AT1R axis and the associated ROS burst. Moreover; we suggest that AT1R antagonist drugs should act synergistically with other cardioprotective agents, such as ascorbic acid, N-acetylcysteine and deferoxamine, leading to an enhanced reduction in the reperfusion injury. This therapy is currently being tested in our laboratory and has shown promising outcomes in experimental studies.

7.
Eur J Pharmacol ; 914: 174665, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34861208

RESUMEN

INTRODUCTION: The function of endoplasmic reticulum (ER), a Ca2+ storage compartment and site of protein folding, is altered by disruption of intracellular homeostasis. Misfolded proteins accumulated in the ER lead to ER stress (ERS), unfolded protein response (UPR) activation and ER Ca2+ loss. Myocardial stunning is a temporary contractile dysfunction, which occurs after brief ischemic periods with minimal or no cell death, being oxidative stress and Ca2+ overload potential underlying mechanisms. Myocardial stunning induces ERS response with negatively impact on the post-ischemic mechanical performance through an unknown mechanism. AIMS: In this study, we explored whether ER Ca2+ efflux through the translocon, a major Ca2+ leak channel, contributes to Ca2+ mishandling and the consequent contractile abnormalities of the stunned myocardium. METHODS: Mechanical performance, cytosolic Ca2+, UPR markers and oxidative state were evaluated in perfused rat/mouse hearts subjected to a brief ischemia followed by reperfusion (I/R) in absence or presence of the translocon inhibitor, emetine (1 µM), comparing its effects with those of the chaperones TUDCA (30 µM) and 4-PBA (3 mM). RESULTS: Emetine treatment precluded the I/R-induced increase in UPR signaling markers and improved the contractile recovery together with a remarkable attenuation in myocardial stiffness when compared to I/R hearts with no drug. This alleviation of I/R-induced mechanical abnormalities was more effective than that obtained with the chemical chaperones, TUDCA and 4-PBA. Moreover, emetine treatment produced a striking improvement in diastolic Ca2+ handling with a partial recovery of the I/R-induced oxidative stress. CONCLUSION: Blocking ER Ca2+ store depletion via translocon suppressed ER stress and improved mechanical performance and diastolic Ca2+ handling of stunned myocardium. Modulation of translocon permeability emerges as a therapeutic approach to face dysfunctional consequences of the I/R injury.


Asunto(s)
Calcio/metabolismo , Emetina/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Contracción Miocárdica , Aturdimiento Miocárdico , Canales de Translocación SEC/antagonistas & inhibidores , Respuesta de Proteína Desplegada , Animales , Señalización del Calcio , Ratones , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Aturdimiento Miocárdico/tratamiento farmacológico , Aturdimiento Miocárdico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/fisiología
8.
Ann Transl Med ; 9(4): 312, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33708939

RESUMEN

BACKGROUND: Brazilin, a major ingredient of Caesalpinia sappan L., possesses multiple pharmaceutical activities, although whether or not brazilin exerts any protective effect on myocardial ischemia-reperfusion injury (MIRI) has not yet been reported. The present study determined the cardioprotective effects of brazilin, and elucidated the role of nuclear factor E2-associated factor 2 (Nrf2) in this process. METHODS: Following treatment with brazilin, H9c2 cells were subjected to 6 h of hypoxia/3 h of reoxygenation. CCK-8 assay and flow cytometry were employed to detect cell viability and apoptosis, respectively. Furthermore, after brazilin treatment, isolated rat hearts underwent 30 min of ischemia, followed by 90 min of reperfusion. Triphenyltetrazolium chloride (TTC) and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining were performed to measure myocardial infarct size and apoptosis, respectively. The changes in the levels of proteins were detected by western blotting. RESULTS: Brazilin treatment dose-dependently led to a significant enhancement in cell viability, a reduction in myocardial infarct size, and a decrease in release of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH). Moreover, brazilin also remarkably inhibited apoptosis and led to various improvements in cardiac function. Additionally, brazilin treatment caused a marked alleviation of oxidative stress, as evidenced by the fact that brazilin reduced the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA), while enhancing the activities of superoxide dismutase (SOD) and glutathione peroxidase (GXH-Px). Mechanistically, it was found that brazilin induced Nrf2 nuclear translocation, with a concomitant upregulation of both heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase (NQO1) expression. Furthermore, the phosphorylation level and transcriptional activity of Nrf2 were enhanced by brazilin, although these enhancements were abrogated by treatment with a protein kinase C (PKC) inhibitor. Finally, it was observed that the protective effects of brazilin could be negated through inhibition of Nrf2, which suggested that the cardioprotection afforded by brazilin was Nrf2-dependent. CONCLUSIONS: Taken together, our results have demonstrated that brazilin may afford protection against MIRI through the activation of Nrf2 via the PKC signaling pathway. These results may lay the foundation for the further use of brazilin in the prevention of MIRI in clinical practice.

9.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;53(9): 0-0, 2020. tab, graf
Artículo en Inglés | LILACS, Coleciona SUS | ID: biblio-1132553

RESUMEN

Myocardial ischemia/reperfusion (MI/R) injury is a complex phenomenon that causes severe damage to the myocardium. However, the potential molecular mechanisms of MI/R injury have not been fully clarified. We identified potential molecular mechanisms and therapeutic targets in MI/R injury through analysis of Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were found between MI/R injury and normal samples, and overlapping DEGs were found between GSE61592 and GSE67308. Gene Ontology (GO) and pathway analysis were performed for overlapping DEGs by Database for Annotation, Visualization and Integration Discovery (DAVID). Then, a network of protein-protein interaction (PPI) was constructed through the Search Tool for the Retrieval of Interacting Genes (STRING) database. Potential microRNAs (miRNAs) and therapeutic small molecules were screened out using microRNA.org database and the Comparative Toxicogenomics database (CTD), respectively. Finally, we identified 21 overlapping DEGs related to MI/R injury. These DEGs were significantly enriched in IL-17 signaling pathway, cytosolic DNA-sensing pathway, chemokine signaling, and cytokine-cytokine receptor interaction pathway. According to the degree in the PPI network, CCL2, LCN2, HP, CCL7, HMOX1, CCL4, and S100A8 were found to be hub genes. Furthermore, we identified potential miRNAs (miR-24-3p, miR-26b-5p, miR-2861, miR-217, miR-4251, and miR-124-3p) and therapeutic small molecules like ozone, troglitazone, rosiglitazone, and n-3 polyunsaturated fatty acids for MI/R injury. These results identified hub genes and potential small molecule drugs, which could contribute to the understanding of molecular mechanisms and treatment for MI/R injury.


Asunto(s)
Daño por Reperfusión Miocárdica , MicroARNs , Biología Computacional , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Mapas de Interacción de Proteínas , Ontología de Genes
10.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;53(2): e9106, 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1055491

RESUMEN

Reperfusion strategies in acute myocardial infarction (AMI) can cause a series of additional clinical damage, defined as myocardial ischemia/reperfusion (I/R) injury, and thus there is a need for effective therapeutic methods to attenuate I/R injury. miR-26a-5p has been proven to be an essential regulator for biological processes in different cell types. Nevertheless, the role of miR-26a-5p in myocardial I/R injury has not yet been reported. We established an I/R injury model in vitro and in vivo. In vitro, we used cardiomyocytes to simulate I/R injury using hypoxia/reoxygenation (H/R) assay. In vivo, we used C57BL/6 mice to construct I/R injury model. The infarct area was examined by TTC staining. The level of miR-26a-5p and PTEN was determined by bioinformatics methods, qRT-PCR, and western blot. In addition, the viability and apoptosis of cardiomyocytes were separately detected by MTT and flow cytometry. The targeting relationship between miR-26a-5p and PTEN was analyzed by the TargetScan website and luciferase reporter assay. I/R and H/R treatment induced myocardial tissue injury and cardiomyocyte apoptosis, respectively. The results showed that miR-26a-5p was down-regulated in myocardial I/R injury. PTEN was found to be a direct target of miR-26a-5p. Furthermore, miR-26a-5p effectively improved viability and inhibited apoptosis in cardiomyocytes upon I/R injury by inhibiting PTEN expression to activate the PI3K/AKT signaling pathway. miR-26a-5p could protect cardiomyocytes against I/R injury by regulating the PTEN/PI3K/AKT pathway, which offers a potential approach for myocardial I/R injury treatment.


Asunto(s)
Animales , Conejos , Daño por Reperfusión Miocárdica/metabolismo , Isquemia Miocárdica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Miocitos Cardíacos/patología , MicroARNs/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Western Blotting , Modelos Animales de Enfermedad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Citometría de Flujo , Ratones Endogámicos C57BL
11.
Braz. arch. biol. technol ; Braz. arch. biol. technol;62: e19190055, 2019. graf
Artículo en Inglés | LILACS | ID: biblio-1055417

RESUMEN

Abstract This study aimed to investigate the cardioprotection of rosuvastatin pre-conditioning (R-Pre) in a rat model of myocardial ischemia / reperfusion (I/R). Male SD rats were assigned into three groups: sham group, I/R group and R-Pre group. Rats in I/R group and R-Pre group received ischemia for 30 min and reperfusion for 2 h. In R-Pre group, rats received intragastrical administration with rosuvastatin at 5 mg/kg once daily for 1 week. After 2-h reperfusion, the cardiac function was detected by ultrasonography; the blood was collected for biochemical analysis; the heart was collected for the TUNEL staining and immunohistochemistry for Bcl-2 and Bax. Our results showed rosuvastatin pre-conditioning for 1 week could significantly reduce the infarct ratio and improve the cardiac function after myocardial I/R injury, in which attenuation of oxidative stress and cell apoptosis played an important role. Our study provides evidence on the cardioprotection of rosuvastatin pre-conditioning and highlight the use of rosuvastatin before cardiopulmonary bypass.


Asunto(s)
Animales , Ratas , Reperfusión Miocárdica , Isquemia/terapia , Cardiotónicos/administración & dosificación , Apoptosis , Estrés Oxidativo , Modelos Animales , Rosuvastatina Cálcica/administración & dosificación
12.
Am J Physiol Heart Circ Physiol ; 314(6): H1289-H1297, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29631370

RESUMEN

Vagal stimulation (VS) during myocardial ischemia and reperfusion has beneficial effects. However, it is not known whether short-term VS applied before ischemia or at the onset of reperfusion protects the ischemic myocardium. This study was designed to determine whether short-term VS applied before ischemia or at the onset of reperfusion reduces myocardial infarct size (IS), mimicking classic preconditioning and postconditioning. A second objective was to study the participation of muscarinic and nicotinic receptors in the protection of both preischemic and reperfusion stimulation. FVB mice were subjected to 30 min of regional myocardial ischemia followed by 2 h of reperfusion without VS, with 10-min preischemic VS (pVS), or with VS during the first 10 min of reperfusion (rVS). pVS reduced IS, and this effect was abolished by atropine and wortmannin. rVS also reduced IS in a similar manner, and this effect was abolished by the α7-nicotinic acetylcholine receptor blocker methyllycaconitine. pVS increased Akt and glycogen synthase kinase (GSK)-3ß phosphorylation. No changes in Akt and GSK-3ß phosphorylation were observed in rVS. Stimulation-mediated IS protection was abolished with the JAK2 blocker AG490. rVS did not modify IL-6 and IL-10 levels in the plasma or myocardium. Splenic denervation and splenectomy did not abolish the protective effect of rVS. In conclusion, pVS and rVS reduced IS by different mechanisms: pVS activated the Akt/GSK-3ß muscarinic pathway, whereas rVS activated α7-nicotinic acetylcholine receptors and JAK2, independently of the cholinergic anti-inflammatory pathway. NEW & NOTEWORTHY Our data suggest, for the first time, that vagal stimulation applied briefly either before ischemia or at the beginning of reperfusion mimics classic preconditioning and postconditioning and reduces myocardial infarction, activating different mechanisms. We also infer an important role of α7-nicotinic receptors for myocardial protection independent of the cholinergic anti-inflammatory pathway.


Asunto(s)
Corazón/inervación , Poscondicionamiento Isquémico , Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Estimulación del Nervio Vago , Nervio Vago/fisiopatología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Janus Quinasa 2/metabolismo , Masculino , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Muscarínicos/metabolismo , Transducción de Señal , Factores de Tiempo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
13.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;44(11): 1148-1155, Nov. 2011. ilus, tab
Artículo en Inglés | LILACS | ID: lil-604273

RESUMEN

The efficacy of endothelin receptor antagonists in protecting against myocardial ischemia/reperfusion (I/R) injury is controversial, and the mechanisms remain unclear. The aim of this study was to investigate the effects of CPU0123, a novel endothelin type A and type B receptor antagonist, on myocardial I/R injury and to explore the mechanisms involved. Male Sprague-Dawley rats weighing 200-250 g were randomized to three groups (6-7 per group): group 1, Sham; group 2, I/R + vehicle. Rats were subjected to in vivo myocardial I/R injury by ligation of the left anterior descending coronary artery and 0.5 percent sodium carboxymethyl cellulose (1 mL/kg) was injected intraperitoneally immediately prior to coronary occlusion. Group 3, I/R + CPU0213. Rats were subjected to identical surgical procedures and CPU0213 (30 mg/kg) was injected intraperitoneally immediately prior to coronary occlusion. Infarct size, cardiac function and biochemical changes were measured. CPU0213 pretreatment reduced infarct size as a percentage of the ischemic area by 44.5 percent (I/R + vehicle: 61.3 ± 3.2 vs I/R + CPU0213: 34.0 ± 5.5 percent, P < 0.05) and improved ejection fraction by 17.2 percent (I/R + vehicle: 58.4 ± 2.8 vs I/R + CPU0213: 68.5 ± 2.2 percent, P < 0.05) compared to vehicle-treated animals. This protection was associated with inhibition of myocardial inflammation and oxidative stress. Moreover, reduction in Akt (protein kinase B) and endothelial nitric oxide synthase (eNOS) phosphorylation induced by myocardial I/R injury was limited by CPU0213 (P < 0.05). These data suggest that CPU0123, a non-selective antagonist, has protective effects against myocardial I/R injury in rats, which may be related to the Akt/eNOS pathway.


Asunto(s)
Animales , Masculino , Ratas , Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Pirazoles/farmacología , Receptor de Endotelina A/antagonistas & inhibidores , Receptor de Endotelina B/antagonistas & inhibidores , Análisis de Varianza , Modelos Animales de Enfermedad , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA