Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 43(1): 26-38, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33712720

RESUMEN

Mitochondria are extraordinarily dynamic organelles that have a variety of morphologies, the status of which are controlled by the opposing processes of fission and fusion. Our recent study shows that inhibition of excessive mitochondrial fission by Drp1 inhibitor (Mdivi-1) leads to a reduction in infarct size and left ventricular (LV) dysfunction following cardiac ischemia-reperfusion (I/R) injury in high fat-fed induced pre-diabetic rats. In the present study, we investigated the cardioprotective effects of a mitochondrial fusion promoter (M1) and a combined treatment (M1 and Mdivi-1) in pre-diabetic rats. Wistar rats were given a high-fat diet for 12 weeks to induce prediabetes. The rats then subjected to 30 min-coronary occlusions followed by reperfusion for 120 min. These rats were intravenously administered M1 (2 mg/kg) or M1 (2 mg/kg) combined with Mdivi-1 (1.2 mg/kg) prior to ischemia, during ischemia or at the onset of reperfusion. We showed that administration of M1 alone or in combination with Mdivi-1 prior to ischemia, during ischemia or at the onset of reperfusion all significantly attenuated cardiac mitochondrial ROS production, membrane depolarization, swelling and dynamic imbalance, leading to reduced arrhythmias and infarct size, resulting in improved LV function in pre-diabetic rats. In conclusion, the promotion of mitochondrial fusion at any time-points during cardiac I/R injury attenuated cardiac mitochondrial dysfunction and dynamic imbalance, leading to decreased infarct size and improved LV function in pre-diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Estado Prediabético/metabolismo , Animales , Diabetes Mellitus Experimental/inducido químicamente , Dieta Alta en Grasa/efectos adversos , Relación Dosis-Respuesta a Droga , Masculino , Dinámicas Mitocondriales/efectos de los fármacos , Estructura Molecular , Daño por Reperfusión Miocárdica/inducido químicamente , Estado Prediabético/inducido químicamente , Quinazolinonas/farmacología , Ratas , Ratas Wistar , Relación Estructura-Actividad
2.
Mol Cell Biochem ; 476(2): 1233-1243, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33247805

RESUMEN

Vinblastine (VBL) has been considered as a first-line anti-tumor drug for many years. However, vinblastine-caused myocardial damage has been continually reported. The underlying molecular mechanism of the myocardial damage remains unknown. Here, we show that vinblastine induces myocardial damage and necroptosis is involved in the vinblastine-induced myocardial damage both in vitro and in vivo. The results of WST-8 and flow cytometry analysis show that vinblastine causes damage to H9c2 cells, and the results of animal experiments show that vinblastine causes myocardial cell damage. The necrosome components, receptor-interacting protein 1 (RIP1) receptor-interacting protein 3 (RIP3), are significantly increased in vinblastine-treated H9c2 cells, primary neonatal rat ventricular myocytes and rat heart tissues. And the downstream substrate of RIP3, mixed lineage kinase domain like protein (MLKL) was also increased. Pre-treatment with necroptosis inhibitors partially inhibits the necrosome components and MLKL levels and alleviates vinblastine-induced myocardial injury both in vitro and in vivo. This study indicates that necroptosis participated in vinblastine-evoked myocardial cell death partially, which would be a potential target for relieving the chemotherapy-related myocardial damage.


Asunto(s)
Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Necroptosis , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Vinblastina/toxicidad , Animales , Antineoplásicos Fitogénicos/toxicidad , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
3.
Arterioscler Thromb Vasc Biol ; 40(6): 1454-1463, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32295420

RESUMEN

Prostanoids are a group of bioactive lipids that are synthesized de novo from membrane phospholipid-released arachidonic acid and have diverse functions in normal physiology and disease. NSAIDs (non-steroidal anti-inflammatory drugs), which are among the most commonly used medications, ameliorate pain, fever, and inflammation by inhibiting COX (cyclooxygenase), which is the rate-limiting enzyme in the biosynthetic cascade of prostanoids. The use of NSAIDs selective for COX-2 inhibition increases the risk of a thrombotic event (eg, myocardial infarction and stroke). All NSAIDs are associated with an increased risk of heart failure. Substantial variation in clinical responses to aspirin exists and is associated with cardiovascular risk. Limited clinical studies suggest the involvement of prostanoids in vascular restenosis in patients who received angioplasty intervention. mPGES (microsomal PG [prostaglandin] E synthase)-1, an alternative target downstream of COX, has the potential to be therapeutically targeted for inflammatory disease, with diminished thrombotic risk relative to selective COX-2 inhibitors. mPGES-1-derived PGE2 critically regulates microcirculation via its receptor EP (receptor for prostanoid E) 4. This review summarizes the actions and associated mechanisms for modulating the biosynthesis of prostanoids in thrombosis, vascular remodeling, and ischemic heart disease as well as their therapeutic relevance.


Asunto(s)
Antiinflamatorios , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/fisiopatología , Descubrimiento de Drogas , Prostaglandinas/fisiología , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Inhibidores de la Ciclooxigenasa/efectos adversos , Insuficiencia Cardíaca/inducido químicamente , Humanos , Infarto del Miocardio/inducido químicamente , Daño por Reperfusión Miocárdica/inducido químicamente , Prostaglandina-E Sintasas/efectos de los fármacos , Prostaglandinas/biosíntesis , Factores de Riesgo , Accidente Cerebrovascular/inducido químicamente , Remodelación Vascular
4.
Biochemistry (Mosc) ; 86(4): 496-505, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33941070

RESUMEN

Antioxidant properties of rat galanin GWTLNSAGYLLGPHAIDNHRSFSDKHGLT-NH2 (Gal), N-terminal fragment of galanin (2-15 aa) WTLNSAGYLLGPHA (G1), and its modified analogue WTLNSAGYLLGPßAH (G2) were studied in vivo in the rat model of regional myocardial ischemia and reperfusion and in vitro in the process of Cu2+-induced free radical oxidation of human blood plasma low-density lipoproteins. Intravenous administration of G1, G2, and Gal to rats after ischemia induction reduced the infarction size and activities of the necrosis markers, creatine kinase-MB and lactate dehydrogenase, in blood plasma at the end of reperfusion. G1, G2, and Gal reduced formation of the spin adducts of hydroxyl radicals in the interstitium of the area at risk during reperfusion, moreover, G2 and Gal also reduced formation of the secondary products of lipid peroxidation in the reperfused myocardium. It was shown in the in vivo experiments and in the in vitro model system that the ability of galanin peptides to reduce formation of ROS and attenuate lipid peroxidation during myocardial reperfusion injury was not associated directly with their effects on activities of the antioxidant enzymes of the heart: Cu,Zn-superoxide dismutase, catalase, and glutathione peroxidase. The peptides G1, G2, and Gal at concentrations of 0.01 and 0.1 mM inhibited Cu2+-induced free radical oxidation of human low-density lipoproteins in vitro. The results of oxidative stress modeling demonstrated that the natural and synthetic agonists of galanin receptors reduced formation of the short-lived ROS in the reperfused myocardium, as well as of lipid radicals in blood plasma. Thus, galanin receptors could be a promising therapeutic target for cardiovascular diseases.


Asunto(s)
Galanina/farmacología , Peroxidación de Lípido , Daño por Reperfusión Miocárdica/metabolismo , Estrés Oxidativo , Administración Intravenosa , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Catalasa , Cobre/química , Cobre/toxicidad , Radicales Libres/toxicidad , Galanina/administración & dosificación , Galanina/uso terapéutico , Glutatión Peroxidasa , Corazón/efectos de los fármacos , Humanos , Masculino , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio/metabolismo , Ratas , Ratas Wistar , Superóxido Dismutasa
5.
J Cell Mol Med ; 24(2): 1332-1344, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31811750

RESUMEN

Fuziline, an aminoalcohol-diterpenoid alkaloid derived from Aconiti lateralis radix preparata, has been reported to have a cardioprotective activity in vitro. However, the potential mechanism of fuziline on myocardial protection remains unknown. In this study, we aimed to explore the efficacy and mechanism of fuziline on isoproterenol (ISO)-induced myocardial injury in vitro and in vivo. As a result, fuziline effectively increased cell viability and alleviated ISO-induced apoptosis. Meanwhile, fuziline significantly decreased the production of ROS, maintained mitochondrial membrane potential (MMP) and blocked the release of cytochrome C, suggesting that fuziline could play the cardioprotective role through restoring the mitochondrial function. Fuziline also could suppress ISO-induced endoplasmic reticulum (ER) stress via the PERK/eIF2α/ATF4/Chop pathway. In addition, using ROS scavenger NAC could decrease ISO-induced apoptosis and block ISO-induced ER stress, while PERK inhibitor GSK2606414 did not reduce the production of ROS, indicating that excess production of ROS induced by ISO triggered ER stress. And fuziline protected against ISO-induced myocardial injury by inhibiting ROS-triggered ER stress. Furthermore, fuziline effectively improved cardiac function on ISO-induced myocardial injury in rats. Western blot analysis also showed that fuziline reduced ER stress-induced apoptosis in vivo. Above these results demonstrated that fuziline could reduce ISO-induced myocardial injury in vitro and in vivo by inhibiting ROS-triggered ER stress via the PERK/eIF2α/ATF4/Chop pathway.


Asunto(s)
Alcaloides/farmacología , Diterpenos/farmacología , Estrés del Retículo Endoplásmico , Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/toxicidad , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Aconitum/química , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Agonistas Adrenérgicos beta/toxicidad , Animales , Apoptosis , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Extractos Vegetales/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
6.
Mol Cell Biochem ; 474(1-2): 1-14, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32681290

RESUMEN

Chronic ethanol abuse can lead to harmful consequences for the heart, resulting in systolic dysfunction, variability in the heart rate, arrhythmia, and cardiac remodelling. However, the precise molecular mechanism responsible for ethanol-induced cardiomyopathy is poorly understood. In this regard, the present study aimed to describe the RIP1/RIP3/MLKL-mediated necroptotic cell death that may be involved in ethanol-induced cardiomyopathy and characterize CBR-mediated effects on the signalling pathway and myocardial injury. We performed an ethanol vapour administration experiment to analyse the effects of ethanol on cardiac structure and function in male C57BL/6J mice. Ethanol induced a significant decline in the cardiac structure and function, as evidenced by a decline in ejection fraction and fractional shortening, and an increase in serum Creatine Kinase levels, myocardial collagen content, and inflammatory reaction. Furthermore, ethanol also upregulated the expression levels of necroptosis-related markers such as p-RIP1, p-RIP3, and p-MLKL in the myocardium. Nec-1 treatment exerted significant cardioprotective effects by salvaging the heart tissue, improving the cardiac function, and mitigating inflammation and necroptosis. In addition, ethanol abuse caused an imbalance in the endocannabinoid system and regulated two cannabinoid receptors (CB1R and CB2R) in the myocardium. Treatment with selective CB2R agonists, JWH-133 or AM1241, markedly improved the cardiac dysfunction and reduced the ethanol-induced necroptosis in the myocardium. Altogether, our data provide evidence that ethanol abuse-induced cardiotoxicity can possibly be attributed to the RIP1/RIP3/MLKL-mediated necroptosis. Moreover, pharmacological activation of CB2R may represent a new cardioprotective strategy against ethanol-induced cardiotoxicity.


Asunto(s)
Apoptosis , Etanol/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Necrosis , Sustancias Protectoras/farmacología , Receptor Cannabinoide CB2/agonistas , Animales , Cannabinoides/farmacología , Depresores del Sistema Nervioso Central/toxicidad , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal
7.
J Cell Biochem ; 120(3): 3709-3715, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30368869

RESUMEN

Acute statin treatment has been reported to be critical in protecting the cardiac cells against ischemia/reperfusion injury by activating PI3K/Akt signal pathway. In vitro rat myocardial ischemia/reperfusion model, chronic statin treatment led to upregulation of phosphatase and tensin homolog (PTEN). This has been potentially indicated the correlation in PTEN and protective effect of statin on myocardium. In this current study, we evaluated the role of sodium orthovanadate a nonspecific inhibitor to PTEN and its correlation with atorvastatin on protecting myocardium against ischemia/reperfusion injury. We found a long-term statin treatment could increase the PTEN level, and this process was counteracted in the presence of sodium orthovanadate. However, the phosphotyrosine level was not affected by this statin. Besides, this process was mediated by Akt signaling since phosphorylated Akt level was altered by statin and sodium orthovanadate treatment. In a conclusion, this study showed a potential mechanism underlying PTEN-induced attenuation in long-term statin's therapeutic effect, which provided the new insight into the synergic role of PTEN and atorvastatin in protecting cardiac cells against ischemia/reperfusion injury.


Asunto(s)
Atorvastatina/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Fosfohidrolasa PTEN/biosíntesis , Vanadatos/farmacología , Animales , Atorvastatina/farmacología , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/enzimología , Miocardio/patología , Ratas , Ratas Sprague-Dawley
8.
Am J Physiol Heart Circ Physiol ; 316(5): H1053-H1064, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30822117

RESUMEN

Arsenic is a common contaminant in drinking water throughout the world, and recent studies support a link between inorganic arsenic (iAS) exposure and ischemic heart disease in men and women. Female hearts exhibit an estrogen-dependent reduction in susceptibility to myocardial ischemic injury compared with males, and as such, female hearts may be more susceptible to the endocrine-disrupting effects of iAS exposure. However, iAS exposure and susceptibility to ischemic heart injury have not been examined in mechanistic studies. Male and female mice (8 wk) were exposed to environmentally relevant concentrations of sodium arsenite (0, 10, 100, and 1,000 parts/billion) via drinking water for 4 wk. Pre- and postexposure echocardiography was performed, and postexposure plasma was collected for 17ß-estradiol measurement. Hearts were excised and subjected to ischemia-reperfusion (I/R) injury via Langendorff perfusion. Exposure to 1,000 parts/billion iAS led to sex-disparate effects, such that I/R injury was exacerbated in female hearts but unexpectedly attenuated in males. Assessment of echocardiographic parameters revealed statistically significant structural remodeling in iAS-treated female hearts with no change in function; males showed no change. Plasma 17ß-estradiol levels were not significantly altered by iAS in male or female mice versus nontreated controls. Although total eNOS protein levels did not change in whole heart homogenates from iAS-treated male or female mice, eNOS phosphorylation (Ser1177) was significantly elevated in iAS-treated male hearts. These results suggest that iAS exposure can induce sex-disparate effects and modulate susceptibility to ischemic heart injury by targeting distinct sex-dependent pathways. NEW & NOTEWORTHY This is the first mechanistic study examining iAS exposure on myocardial ischemia-reperfusion injury in male and female mice. Following iAS exposure, ischemia-reperfusion injury was exacerbated in female hearts but attenuated in males. iAS treatment induced statistically significant cardiac remodeling in females, with no change in males. iAS treatment also enhanced phosphorylated eNOS levels at Ser1177, but only in male hearts. These results suggest that iAS alters susceptibility to myocardial I/R injury through distinct sex-dependent pathways.


Asunto(s)
Arsenitos/toxicidad , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Compuestos de Sodio/toxicidad , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiotoxicidad , Modelos Animales de Enfermedad , Estradiol/sangre , Femenino , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Factores Sexuales
9.
Biol Pharm Bull ; 42(5): 721-727, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30867343

RESUMEN

Astragaloside IV (AS-IV) is one of the main pharmacologically active compounds found in Astragalus membranaceus. AS-IV has protective effects against ischemia-reperfusion injury (IRI), but its mechanism of action has not yet been determined. This study aims to investigate the effect of AS-IV on IRI and its effect on the phosphadylinositol 3-kinase (PI3K)/Akt/heme oxygenase (HO-1) signaling pathway through in vitro experiments. Firstly, a cell culture model of myocardiocyte hypoxia-reoxygenation (H/R) injury was replicated. After AS-IV treatment, cell viability, reactive oxygen species (ROS) levels, as well as the content or activity of the cellular factors lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), were measured to evaluate the effect of treatment with AS-IV. The effect of AS-IV on HO-1 protein expression and nuclear factor E2-related factor 2 (Nrf2) and Bach1 protein expression was determined by Western blotting. Finally, a reversal of the effect of AS-IV treatment was observed following co-incubation with a PI3K inhibitor. Our results show that AS-IV has good protective effect on H/R injury and has anti-oxidative stress and anti-inflammatory effects. It can regulate the expression of Nrf2 and Bach1 proteins in the nucleus and promote the expression of HO-1 protein, while a PI3K inhibitor can partially reverse the above effects. This study suggests that the PI3K/Akt/HO-1 signaling pathway may be a key signaling pathway for the anti-IRI effect of AS-IV.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Saponinas/farmacología , Triterpenos/farmacología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inflamación , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Int Heart J ; 59(1): 190-196, 2018 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-29279520

RESUMEN

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


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calmodulina/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Taurina/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calmodulina/biosíntesis , Células Cultivadas , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/toxicidad , Microscopía Confocal , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Reacción en Cadena de la Polimerasa , ARN/genética , Ratas , Ratas Wistar
11.
J Cell Mol Med ; 21(1): 58-71, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27491814

RESUMEN

Mitochondrial dynamics-fission and fusion-are associated with ischaemic heart disease (IHD). This study explored the protective effect of vagal nerve stimulation (VNS) against isoproterenol (ISO)-induced myocardial ischaemia in a rat model and tested whether VNS plays a role in preventing disorders of mitochondrial dynamics and function. Isoproterenol not only caused cardiac injury but also increased the expression of mitochondrial fission proteins [dynamin-related peptide1 (Drp1) and mitochondrial fission protein1 (Fis-1)) and decreased the expression of fusion proteins (optic atrophy-1 (OPA1) and mitofusins1/2 (Mfn1/2)], thereby disrupting mitochondrial dynamics and leading to increase in mitochondrial fragments. Interestingly, VNS restored mitochondrial dynamics through regulation of Drp1, Fis-1, OPA1 and Mfn1/2; enhanced ATP content and mitochondrial membrane potential; reduced mitochondrial permeability transition pore (MPTP) opening; and improved mitochondrial ultrastructure and size. Furthermore, VNS reduced the size of the myocardial infarction and ameliorated cardiomyocyte apoptosis and cardiac dysfunction induced by ISO. Moreover, VNS activated AMP-activated protein kinase (AMPK), which was accompanied by phosphorylation of Ca2+ /calmodulin-dependent protein kinase kinase ß (CaMKKß) during myocardial ischaemia. Treatment with subtype-3 of muscarinic acetylcholine receptor (M3 R) antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide or AMPK inhibitor Compound C abolished the protective effects of VNS on mitochondrial dynamics and function, suggesting that M3 R/CaMKKß/AMPK signalling are involved in mediating beneficial effects of VNS. This study demonstrates that VNS modulates mitochondrial dynamics and improves mitochondrial function, possibly through the M3 R/CaMKKß/AMPK pathway, to attenuate ISO-induced cardiac damage in rats. Targeting mitochondrial dynamics may provide a novel therapeutic strategy in IHD.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Isoproterenol/farmacología , Dinámicas Mitocondriales/fisiología , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/metabolismo , Receptor Muscarínico M3/metabolismo , Animales , Apoptosis/fisiología , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Estimulación del Nervio Vago/métodos
12.
J Cell Mol Med ; 21(12): 3178-3189, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28905500

RESUMEN

The purpose of the present study was to investigate the effect of salidroside (Sal) on myocardial injury in lipopolysaccharide (LPS)-induced endotoxemic in vitro and in vivo. SD rats were randomly divided into five groups: control group, LPS group (15 mg/kg), LPS plus dexamethasone (2 mg/kg), LPS plus Sal groups with different Sal doses (20, 40 mg/kg). Hemodynamic measurement and haematoxylin and eosin staining were performed. Serum levels of creatine kinase (CK), lactate dehydrogenase, the activities of the antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-px), glutathione, tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were measured after the rats were killed. iNOS, COX-2, NF-κB and PI3K/Akt/mTOR pathway proteins were detected by Western blot. In vitro, we evaluated the protective effect of Sal on rat embryonic heart-derived myogenic cell line H9c2 induced by LPS. Reactive oxygen species (ROS) in H9c2 cells was measured by flow cytometry, and the activities of the antioxidant enzymes CAT, SOD, GSH-px, glutathione-S-transferase, TNF-α, IL-6 and IL-1ß in cellular supernatant were measured. PI3K/Akt/mTOR signalling was examined by Western blot. As a result, Sal significantly attenuated the above indices. In addition, Sal exerts pronounced cardioprotective effect in rats subjected to LPS possibly through inhibiting the iNOS, COX-2, NF-κB and PI3K/Akt/mTOR pathway in vivo. Furthermore, the pharmacological effect of Sal associated with the ROS-mediated PI3K/Akt/mTOR pathway was proved by the use of ROS scavenger, N-acetyl-l-cysteine, in LPS-stimulated H9C2 cells. Our results indicated that Sal could be a potential therapeutic agent for the treatment of cardiovascular disease.


Asunto(s)
Cardiotónicos/farmacología , Endotoxemia/tratamiento farmacológico , Glucósidos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fenoles/farmacología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Animales , Catalasa/genética , Catalasa/metabolismo , Línea Celular , Dexametasona/farmacología , Endotoxemia/inducido químicamente , Endotoxemia/genética , Endotoxemia/patología , Regulación de la Expresión Génica , Glutatión/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Hemodinámica/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
Cell Physiol Biochem ; 44(3): 1011-1023, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29179205

RESUMEN

BACKGROUND/AIMS: Myocardial ischemia/reperfusion (I/R) injury remains a great challenge in clinical therapy. Tissue inhibitor of metalloproteinases 3 (TIMP3) plays a crucial role in heart physiological and pathophysiological processes. However, the effects of TIMP3 on I/R injury remain unknown. METHODS: C57BL/6 mice were infected with TIMP3 adenovirus by local delivery in myocardium followed by I/R operation or doxorubicin treatment. Neonatal rat cardiomyocytes were pretreated with TIMP3 adenovirus prior to anoxia/reoxygenation (A/R) treatment in vitro. Histology, echocardiography, in vivo phenotypical analysis, flow cytometry and western blotting were used to investigate the altered cardiac function and underlying mechanisms. RESULTS: The results showed that upregulation of TIMP3 in myocardium markedly inhibited myocardial infarct areas and the cardiac dysfunction induced by I/R or by doxorubicin treatment. TUNEL staining revealed that TIMP3 overexpression attenuated I/R-induced myocardial apoptosis, accompanied by decreased Bax/Bcl-2 ratio, Cleaved Caspase-3 and Cleaved Caspase-9 expression. In vitro, A/R-induced cardiomyocyte apoptosis was abrogated by pharmacological inhibition of reactive oxygen species (ROS) production or MAPKs signaling. Attenuation of ROS production reversed A/R-induced MAPKs activation, whereas MAPKs inhibitors showed on effect on ROS production. Furthermore, in vivo or in vitro overexpression of TIMP3 significantly inhibited I/R- or A/R-induced ROS production and MAPKs activation. CONCLUSION: Our findings demonstrate that TIMP3 upregulation protects against cardiac I/R injury through inhibiting myocardial apoptosis. The mechanism may be related to inhibition of ROS-initiated MAPKs pathway. This study suggests that TIMP3 may be a potential therapeutic target for the treatment of I/R injury.


Asunto(s)
Miocardio/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Acetilcisteína/farmacología , Animales , Antracenos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Óxidos N-Cíclicos/farmacología , Doxorrubicina/toxicidad , Ecocardiografía , Corazón/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Marcadores de Spin , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína X Asociada a bcl-2/metabolismo
14.
Int J Med Sci ; 14(11): 1163-1172, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104471

RESUMEN

Background: Adverse stress exposure during the early neonatal period has been shown to cause aberrant development, resulting in an increased risk of adult disease. We tested the hypothesis that neonatal exposure to lipopolysaccharide (LPS) does not alter heart function at rest condition but causes heart dysfunction under stress stimulation later in life. Methods: Saline control or LPS were administered to neonatal rats via intraperitoneal injection. Experiments were conducted in 6 week-old male and female rats. Isolated hearts were perfused in a Langendorff preparation. Results: Neonatal LPS exposure exhibited no effects on the body weight of the developing rats, but induced decreases in the left ventricle (LV) to the body weight ratio in male rats. Neonatal LPS exposure showed no effects on the baseline heart function determined by in vivo and ex vivo experiments, but caused decreases in the post-ischemic recovery of the LV function in male but not female rats. Neonatal LPS-mediated LV dysfunction was associated with an increase in myocardial infarct size and the LDH release in the male rats. Conclusion: The present study provides novel evidence that neonatal immune challenges could induce gender-dependent long-term effects on cardiac development and heart function, which reinforces the notion that adverse stress exposure during the early neonatal period can aggravate heart functions and the development of a heart ischemia-sensitive phenotype later in life.


Asunto(s)
Corazón/fisiopatología , Lipopolisacáridos/toxicidad , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión/fisiopatología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Humanos , Masculino , Infarto del Miocardio/inducido químicamente , Daño por Reperfusión Miocárdica/inducido químicamente , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/inducido químicamente , Caracteres Sexuales
15.
Cardiovasc Diabetol ; 15(1): 146, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27733157

RESUMEN

BACKGROUND: Patients with diabetes are prone to develop cardiac hypertrophy and more susceptible to myocardial ischemia-reperfusion (I/R) injury, which are concomitant with hyperglycemia-induced oxidative stress and impaired endothelial nitric oxide (NO) synthase (eNOS)/NO signaling. Caveolae are critical in the transduction of eNOS/NO signaling in cardiovascular system. Caveolin (Cav)-3, the cardiomyocytes-specific caveolae structural protein, is decreased in the diabetic heart in which production of reactive oxygen species are increased. We hypothesized that treatment with antioxidant N-acetylcysteine (NAC) could enhance cardiac Cav-3 expression and attenuate caveolae dysfunction and the accompanying eNOS/NO signaling abnormalities in diabetes. METHODS: Control or streptozotocin-induced diabetic rats were either untreated or treated with NAC (1.5 g/kg/day, NAC) by oral gavage for 4 weeks. Rats in subgroup were randomly assigned to receive 30 min of left anterior descending artery ligation followed by 2 h of reperfusion. Isolated rat cardiomyocytes or H9C2 cells were exposed to low glucose (LG, 5.5 mmol/L) or high glucose (HG, 25 mmol/L) for 36 h before being subjected to 4 h of hypoxia followed by 4 h of reoxygenation (H/R). RESULTS: NAC treatment ameliorated myocardial dysfunction and cardiac hypertrophy, and attenuated myocardial I/R injury and post-ischemic cardiac dysfunction in diabetic rats. NAC attenuated the reductions of NO, Cav-3 and phosphorylated eNOS and mitigated the augmentation of O2-, nitrotyrosine and 15-F2t-isoprostane in diabetic myocardium. Immunofluorescence analysis demonstrated the colocalization of Cav-3 and eNOS in isolated cardiomyocytes. Immunoprecipitation analysis revealed that diabetic conditions decreased the association of Cav-3 and eNOS in isolated cardiomyocytes, which was enhanced by treatment with NAC. Disruption of caveolae by methyl-ß-cyclodextrin or Cav-3 siRNA transfection reduced eNOS phosphorylation. NAC treatment attenuated the reductions of Cav-3 expression and eNOS phosphorylation in HG-treated cardiomyocytes or H9C2 cells. NAC treatment attenuated HG and H/R induced cell injury, which was abolished during concomitant treatment with Cav-3 siRNA or eNOS siRNA. CONCLUSIONS: Hyperglycemia-induced inhibition of eNOS activity might be consequences of caveolae dysfunction and reduced Cav-3 expression. Antioxidant NAC attenuated myocardial dysfunction and myocardial I/R injury by improving Cav-3/eNOS signaling.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Cardiomegalia/prevención & control , Caveolina 3/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Cardiomiopatías Diabéticas/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/enzimología , Cardiomegalia/fisiopatología , Caveolas/efectos de los fármacos , Caveolas/enzimología , Caveolas/patología , Caveolina 3/genética , Hipoxia de la Célula , Línea Celular , Citoprotección , Diabetes Mellitus Experimental/inducido químicamente , Cardiomiopatías Diabéticas/inducido químicamente , Cardiomiopatías Diabéticas/enzimología , Cardiomiopatías Diabéticas/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Interferencia de ARN , Ratas Sprague-Dawley , Estreptozocina , Transfección , Función Ventricular Izquierda/efectos de los fármacos
16.
Inflamm Res ; 65(8): 613-22, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27071824

RESUMEN

OBJECTIVE: The aim of the present study was to investigate the cardioprotective effects of baicalein, main bioactive constituent from roots of Scutellaria baicalensis and Scutellaria lateriflora, on isoproterenol (ISO) induced acute myocardial infarction model in rats and to explore the underlying mechanisms. METHOD: Rats were treated with baicalein (50 mg/kg and 100 mg/kg) orally for 14 days and on 13th and 14th day, myocardial injury was induced by ISO injection (100 mg/kg, subcutaneous) at an interval of 24 h. RESULT: Our study showed that ISO administration resulted in significant elevations in the levels of cardiac injury biomarkers such as cardiac troponin I, creatine kinase-MB, AST and ALT. Concentrations of reactive nitrogen species and reactive oxygen species in the heart tissue increased significantly while antioxidant enzymes level declined. The levels of tissue pro-inflammatory cytokines tumor necrosis factor-α and interleukin-6 were significantly increased after ISO administration. Pretreatment with baicalein significantly reversed these alterations induced by ISO administration. Exploration of the underlying mechanisms of protective effect of baicalein pretreatment revealed that it repressed the expression of nuclear factor kappa B and restored the ISO induced elevation of pro-inflammatory cytokines, oxidative and nitrosative stress. We found that baicalein pretreatment enhanced the level of antioxidant defense enzymes like SOD, catalase and GSH. Furthermore, the present study also demonstrated cardioprotective effects of baicalein by the histopathological findings. CONCLUSION: Taken together, our findings demonstrated that baicalein pretreatment might have a potential benefit in prevention and terminating ischemic heart diseases like myocardial infarction.


Asunto(s)
Antiinflamatorios/uso terapéutico , Cardiotónicos/uso terapéutico , Flavanonas/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Biomarcadores/sangre , Cardiotónicos/farmacología , Catalasa/metabolismo , Forma MB de la Creatina-Quinasa/sangre , Flavanonas/farmacología , Glutatión/metabolismo , Interleucina-6/metabolismo , Isoproterenol , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Superóxido Dismutasa/metabolismo , Troponina I/sangre , Factor de Necrosis Tumoral alfa/metabolismo
17.
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
18.
Am J Physiol Heart Circ Physiol ; 308(9): H998-H1006, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25681431

RESUMEN

Increases in airborne particulate matter (PM) are linked to increased mortality from myocardial ischemia. PM contains environmentally persistent free radicals (EPFRs) that form as halogenated hydrocarbons chemisorb to transition metal oxide-coated particles, and are capable of sustained redox cycling. We hypothesized that exposure to the EPFR DCB230 would increase cardiac vulnerability to subsequent myocardial ischemia-reperfusion (MI/R) injury. Rats were exposed to DCB230 or vehicle via nose-only inhalation (230 µg max/day) over 30 min/day for 7 days. MI/R or sham MI/R (sham) was initiated 24 h after the final exposure. Following 1 or 7 days of reperfusion, left ventricular (LV) function was assessed and infarct size measured. In vehicle-exposed rats, MI/R injury did not significantly reduce cardiac output (CO), stroke volume (SV), stroke work (SW), end-diastolic volume (EDV), or end-systolic volume (ESV) after 1 day of reperfusion, despite significant reductions in end-systolic pressure (ESP). Preload-recruitable SW (PRSW; contractility) was elevated, presumably to maintain LV function. MI/R 1-day rats exposed to DCB230 also had similarly reduced ESP. Compared with vehicle controls, CO, SV, and SW were significantly reduced in DCB230-exposed MI/R 1-day rats; moreover, PRSW did not increase. DCB230's effects on LV function dissipated within 8 days of exposure. These data show that inhalation of EPFRs can exacerbate the deficits in LV function produced by subsequent MI/R injury. Infarct size was not different between the MI/R groups. We conclude that inhalation of EPFRs can compromise cardiac function during MI/R injury and may help to explain the link between PM and MI/R-related mortality.


Asunto(s)
Radicales Libres/toxicidad , Infarto del Miocardio/etiología , Daño por Reperfusión Miocárdica/etiología , Material Particulado/toxicidad , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Exposición por Inhalación , Masculino , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Ratas Sprague-Dawley , Medición de Riesgo , Factores de Riesgo , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular/efectos de los fármacos
19.
Am J Physiol Heart Circ Physiol ; 308(7): H759-67, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25595132

RESUMEN

We investigated the role of inducible nitric oxide (NO) synthase (iNOS) on ischemic myocardial damage in rats exposed to daily low nontoxic levels of carbon monoxide (CO). CO is a ubiquitous environmental pollutant that impacts on mortality and morbidity from cardiovascular diseases. We have previously shown that CO exposure aggravates myocardial ischemia-reperfusion (I/R) injury partly because of increased oxidative stress. Nevertheless, cellular mechanisms underlying cardiac CO toxicity remain hypothetical. Wistar rats were exposed to simulated urban CO pollution for 4 wk. First, the effects of CO exposure on NO production and NO synthase (NOS) expression were evaluated. Myocardial I/R was performed on isolated perfused hearts in the presence or absence of S-methyl-isothiourea (1 µM), a NOS inhibitor highly specific for iNOS. Finally, Ca(2+) handling was evaluated in isolated myocytes before and after an anoxia-reoxygenation performed with or without S-methyl-isothiourea or N-acetylcystein (20 µM), a nonspecific antioxidant. Our main results revealed that 1) CO exposure altered the pattern of NOS expression, which is characterized by increased neuronal NOS and iNOS expression; 2) cardiac NO production increased in CO rats because of its overexpression of iNOS; and 3) the use of a specific inhibitor of iNOS reduced myocardial hypersensitivity to I/R (infarct size, 29 vs. 51% of risk zone) in CO rat hearts. These last results are explained by the deleterious effects of NO and reactive oxygen species overproduction by iNOS on diastolic Ca(2+) overload and myofilaments Ca(2+) sensitivity. In conclusion, this study highlights the involvement of iNOS overexpression in the pathogenesis of simulated urban CO air pollution exposure.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Monóxido de Carbono/toxicidad , Infarto del Miocardio/inducido químicamente , Daño por Reperfusión Miocárdica/inducido químicamente , Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Acoplamiento Excitación-Contracción/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Masculino , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miofibrillas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Regulación hacia Arriba
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA