Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Cardiovasc Toxicol ; 24(11): 1236-1252, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39264521

RESUMEN

Uremic cardiomyopathy (UC) represents a complex syndrome characterized by different cardiac complications, including systolic and diastolic dysfunction, left ventricular hypertrophy, and diffuse fibrosis, potentially culminating in myocardial infarction (MI). Revascularization procedures are often necessary for MI management and can induce ischemia reperfusion injury (IR). Despite this clinical relevance, the role of fine particulate matter (PM2.5) in UC pathology and the underlying subcellular mechanisms governing this pathology remains poorly understood. Hence, we investigate the impact of PM2.5 exposure on UC susceptibility to IR injury. Using a rat model of adenine-induced chronic kidney disease (CKD), the animals were exposed to PM2.5 at 250 µg/m3 for 3 h daily over 21 days. Subsequently, hearts were isolated and subjected to 30 min of ischemia followed by 60 min of reperfusion to induce IR injury. UC hearts exposed to PM2.5 followed by IR induction (Adenine + PM_IR) exhibited significantly impaired cardiac function and increased cardiac injury (increased infarct size and apoptosis). Analysis at the subcellular level revealed reduced mitochondrial copy number, impaired mitochondrial bioenergetics, decreased expression of PGC1-α (a key regulator of mitochondrial biogenesis), and compromised mitochondrial quality control mechanisms. Additionally, increased mitochondrial oxidative stress and perturbation of the PI3K/AKT/AMPK signaling axis were evident. Our findings therefore collectively indicate that UC myocardium when exposed to PM2.5 is more vulnerable to IR-induced injury, primarily due to severe mitochondrial impairment.


Asunto(s)
Apoptosis , Cardiomiopatías , Modelos Animales de Enfermedad , Metabolismo Energético , Mitocondrias Cardíacas , Daño por Reperfusión Miocárdica , Material Particulado , Transducción de Señal , Uremia , Animales , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/inducido químicamente , Material Particulado/toxicidad , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Mitocondrias Cardíacas/efectos de los fármacos , Masculino , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/fisiopatología , Apoptosis/efectos de los fármacos , Uremia/metabolismo , Uremia/inducido químicamente , Uremia/patología , Uremia/complicaciones , Metabolismo Energético/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/metabolismo , Contaminantes Atmosféricos/toxicidad , Ratas Sprague-Dawley , Proteínas Proto-Oncogénicas c-akt/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Adenina/toxicidad , Adenina/farmacología , Estrés Oxidativo/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/fisiopatología , Fosfatidilinositol 3-Quinasa/metabolismo
2.
Cardiovasc Toxicol ; 24(9): 918-928, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39026038

RESUMEN

Cardiovascular disease remains the leading cause of death worldwide, with acute myocardial infarction and anticancer drug-induced cardiotoxicity being the significant factors. The most effective treatment for acute myocardial infarction is rapid restoration of coronary blood flow by thrombolytic therapy or percutaneous coronary intervention. However, myocardial ischemia-reperfusion injury (MI/RI) after reperfusion therapy is common in acute myocardial infarction, thus affecting the prognosis of patients with acute myocardial infarction. There is no effective treatment for MI/RI. Anthracyclines such as Doxorubicin (DOX) have limited clinical use due to their cardiotoxicity, and the mechanism of DOX-induced cardiac injury is complex and not yet fully understood. N6-methyladenosine (m6A) plays a crucial role in many biological processes. Emerging evidence suggests that m6A methylation plays a critical regulatory role in MI/RI and DOX-induced cardiotoxicity (DIC), suggesting that m6A may serve as a novel biomarker and therapeutic target for MI/RI and DIC. M6A methylation may mediate the pathophysiological processes of MI/RI and DIC by regulating cellular autophagy, apoptosis, oxidative stress, and inflammatory response. In this paper, we first focus on the relationship between m6A methylation and MI/RI, then further elucidate that m6A methylation may mediate the pathophysiological process of MI/RI through the regulation of cellular autophagy, apoptosis, oxidative stress, and inflammatory response. Finally, briefly outline the roles played by m6A in DIC, which will provide a new methodology and direction for the research and treatment of MI/RI and DIC.


Asunto(s)
Adenosina , Apoptosis , Cardiotoxicidad , Doxorrubicina , Daño por Reperfusión Miocárdica , Estrés Oxidativo , Doxorrubicina/efectos adversos , Animales , Humanos , Adenosina/análogos & derivados , Adenosina/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/genética , Metilación , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Transducción de Señal , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocardio/metabolismo , Miocardio/patología , Mediadores de Inflamación/metabolismo
3.
Adv Sci (Weinh) ; 9(15): e2105408, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35319828

RESUMEN

Acute myocardial infarction (MI) is the leading cause of death worldwide. Exogenous delivery of nitric oxide (NO) to the infarcted myocardium has proven to be an effective strategy for treating MI due to the multiple physiological functions of NO. However, reperfusion of blood flow to the ischemic tissues is accompanied by the overproduction of toxic reactive oxygen species (ROS), which can further exacerbate tissue damage and compromise the therapeutic efficacy. Here, an injectable hydrogel is synthesized from the chitosan modified by boronate-protected diazeniumdiolate (CS-B-NO) that can release NO in response to ROS stimulation and thereby modulate ROS/NO disequilibrium after ischemia/reperfusion (I/R) injury. Furthermore, administration of CS-B-NO efficiently attenuated cardiac damage and adverse cardiac remodeling, promoted repair of the heart, and ameliorated cardiac function, unlike a hydrogel that only released NO, in a mouse model of myocardial I/R injury. Mechanistically, regulation of the ROS/NO balance activated the antioxidant defense system and protected against oxidative stress induced by I/R injury via adaptive regulation of the Nrf2-Keap1 pathway. Inflammation is then reduced by inhibition of the activation of NF-κB signaling. Collectively, these results show that this dual-function hydrogel may be a promising candidate for the protection of tissues and organs after I/R injury.


Asunto(s)
Daño por Reperfusión Miocárdica , Animales , Hidrogeles , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico , Especies Reactivas de Oxígeno/metabolismo
4.
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
5.
Kaohsiung J Med Sci ; 38(1): 38-48, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34369657

RESUMEN

Proteins in Jumonji family function as histone demethylases and participate in cardiac development. Jumonji domain containing 5 (JMJD5) is responsible for the embryonic development through removing methyl moieties from H3K36me2 histone, and has pro-proliferative effect on heart and eye development. However, the protective role of JMJD5 against oxygen-glucose deprivation and reperfusion (OGD/R)-induced injury in cardiomyocytes has not been fully understood. Firstly, myocardial ischemia/reperfusion (I/R) rat model was established by ligation of left coronary artery. OGD/R was performed in non-transfected H9C2 or H9C2 transfected with pcDNA-JMJD5 plasmid to induce cell cytotoxicity. Data from qRT-PCR and western blot showed that JMJD5 was reduced in the heart tissues of myocardial I/R rat model and OGD/R-induced H9C2. Secondly, JMJD5 over-expression attenuated OGD/R-induced decrease in cell viability and increase in lactate dehydrogenase secretion and cell apoptosis in H9C2. Mitophagy was promoted by pcDNA-mediated over-expression of JMJD5 with enhanced protein expression of LC3-I, LC3-II, Atg5, and Beclin 1. Thirdly, knockdown of JMJD5 aggravated OGD/R-induced decrease in hypoxia-inducible factor-1α (HIF-1α), whereas JMJD5 over-expression enhanced BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein) through upregulation of HIF-1α. Lastly, BNIP3 silencing promoted cell apoptosis, suppressed mitophagy, and attenuated the protective effects of JMJD5 over-expression against OGD/R-induced injury in H9C2. In conclusion, JMJD5 exerted protective effects against OGD/R-induced injury in cardiomyocytes through upregulation of HIF-1α-BNIP3.


Asunto(s)
Glucosa , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Animales , Apoptosis , Supervivencia Celular , Modelos Animales de Enfermedad , Glucosa/deficiencia , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Histona Demetilasas con Dominio de Jumonji , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/citología , Sustancias Protectoras , Ratas
6.
Sci Rep ; 11(1): 13845, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226619

RESUMEN

Long-term nicotine intake is associated with an increased risk of myocardial damage and dysfunction. However, it remains unclear whether targeting mitochondrial reactive oxygen species (ROS) prevents nicotine-induced cardiac remodeling and dysfunction. This study investigated the effects of mitoTEMPO (a mitochondria-targeted antioxidant), and resveratrol (a sirtuin activator) , on nicotine-induced cardiac remodeling and dysfunction. Sprague-Dawley rats were administered 0.6 mg/kg nicotine daily with 0.7 mg/kg mitoTEMPO, 8 mg/kg resveratrol, or vehicle alone for 28 days. At the end of the study, rat hearts were collected to analyze the cardiac structure, mitochondrial ROS level, oxidative stress, and inflammation markers. A subset of rat hearts was perfused ex vivo to determine the cardiac function and myocardial susceptibility to ischemia-reperfusion injury. Nicotine administration significantly augmented mitochondrial ROS level, cardiomyocyte hypertrophy, fibrosis, and inflammation in rat hearts. Nicotine administration also induced left ventricular dysfunction, which was worsened by ischemia-reperfusion in isolated rat hearts. MitoTEMPO and resveratrol both significantly attenuated the adverse cardiac remodeling induced by nicotine, as well as the aggravation of postischemic ventricular dysfunction. Findings from this study show that targeting mitochondrial ROS with mitoTEMPO or resveratrol partially attenuates nicotine-induced cardiac remodeling and dysfunction.


Asunto(s)
Antioxidantes/farmacología , Mitocondrias Cardíacas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Animales , Remodelación Atrial/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Nicotina/toxicidad , Ratas , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular/efectos de los fármacos
7.
J Med Chem ; 64(13): 9166-9181, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34132541

RESUMEN

Timely myocardial reperfusion salvages ischemic myocardium from infarction, whereas reperfusion itself induces cardiomyocyte death, which is called myocardial ischemia/reperfusion (MI/R) injury. Herein, ß-carboline derivative 17c was designed and synthesized with obvious myocardial protective activity for the first time. Pretreatment of 17c effectively protected the cardiomyocyte H9c2 cells from H2O2-induced lactate dehydrogenase leakage and restored the endogenous antioxidants, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Besides, 17c effectively protected the mitochondria through decreasing the reactive oxygen species overproduction and enhancing the mitochondrial membrane potential. As a result, 17c significantly reduced the necrosis of cardiomyocytes in H2O2-induced oxidative stress, which was more potent than polydatin. In MI/R injury rats, 17c pretreatment obviously increased the levels of SOD and GSH-Px and inhibited the apoptosis of cardiomyocytes. Through this way, the size of myocardial infarction was significantly reduced after MI/R injury in vivo, better than that of polydatin, suggesting that 17c is a promising cardioprotectant for the prevention of MI/R injury.


Asunto(s)
Carbolinas/farmacología , Descubrimiento de Drogas , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Sustancias Protectoras/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Glutatión Peroxidasa/metabolismo , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Masculino , Estructura Molecular , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Superóxido Dismutasa/metabolismo
8.
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
9.
J Extracell Vesicles ; 10(4): e12072, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33664937

RESUMEN

Extracellular vesicles (EVs) curb important biological functions. We previously disclosed that ischemia-reperfusion (IR) induces increased release of EVs (IR-EVs) in the heart. However, the role of IR-EVs in IR pathological process remains poorly understood. Here we found that adoptive transfer of IR-EVs aggravated IR induced heart injury, and EV inhibition by GW4869 reduced the IR injury. Our in vivo and in vitro investigations substantiated that IR-EVs facilitated M1-like polarization of macrophages with increased expression of proinflammatory cytokines. Further, we disclosed the miRNA profile in cardiac EVs and confirmed the enrichment of miRNAs, such as miR-155-5p in IR-EVs compared to EVs from the sham heart (S-EVs). In particular, IR-EVs transferred miR-155-5p to macrophages and enhanced the inflammatory response through activating JAK2/STAT1 pathway. Interestingly, IR-EVs not only boosted the local inflammation in the heart, but even triggered systemic inflammation in distant organs. Taken together, we newly identify an IR-EVs-miR-155-5p-M1 polarization axis in the heart post IR. The EVs derived from IR-injured heart contribute to both local and systemic inflammation. Importantly, EV inhibition by GW4869 is supposed to be a promising therapeutic strategy for IR injury.


Asunto(s)
Vesículas Extracelulares/metabolismo , Lesiones Cardíacas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/efectos de los fármacos , Lesiones Cardíacas/inducido químicamente , Janus Quinasa 2 , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Daño por Reperfusión Miocárdica/inducido químicamente , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
10.
Eur Rev Med Pharmacol Sci ; 25(2): 968-975, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33577052

RESUMEN

OBJECTIVE: The aim of this study was to explore the effects of sevoflurane (SEV) pretreatment on Adriamycin (ADR)-induced myocardial injury through the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase-3ß (GSK-3ß) pathway. MATERIALS AND METHODS: A total of 24 rats weighing 200-250 g were divided into four groups, including: control group (C group), ADR injection group (ADR group), SEV pretreatment group (ADR + SEV group) and inhibitor group (ADR + SEV + LY group). H9c2 cells were cultured in vitro and were divided into control group (C group), ADR treatment group (ADR group), and SEV pretreatment group (ADR + SEV group) and inhibitor group (ADR + SEV + LY group) as well. Next, the content of aspartate aminotransferase (AST), lactate dehydrogenase (LDH) and creatine kinase (CK) in the serum was detected via Enzyme-Linked Immunosorbent Assay (ELISA). Hematoxylin-eosin (HE) staining assay was performed to determine the severity of myocardial injury. Meanwhile, the apoptosis rate of cells was detected through terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL) assay. Additionally, Western blotting (WB) was employed to measure the protein expression levels of phosphorylated (p)-GSK-3ß, p-PI3K, Akt and p-Akt. RESULTS: Compared with C group, ADR significantly increased the content of AST, LDH and CK in the serum (p<0.01), reduced protein expression levels of p-GSK-3ß, p-PI3K and p-Akt (p<0.01), increased apoptosis rate (p<0.01), and induced myocardial injury. SEV pretreatment significantly alleviated the effect of ADR, manifested as significantly lowered content of AST, LDH and CK in the serum (p<0.01), distinctly elevated protein expression levels of p-GSK-3ß, p-PI3K and p-Akt (p<0.01), notably reduced apoptosis rate (p<0.01), and relieved myocardial injury. LY294002 remarkably inhibited the protective effect of SEV against myocardial injury (p<0.01) CONCLUSIONS: SEV is able to prominently ameliorate ADR-induced myocardial injury by regulating the phosphorylation level of the PI3K/Akt/GSK-3ß signaling pathway.


Asunto(s)
Anestésicos por Inhalación/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Sevoflurano/farmacología , Anestésicos por Inhalación/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Doxorrubicina , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Sevoflurano/administración & dosificación
11.
Basic Clin Pharmacol Toxicol ; 128(2): 322-333, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32991780

RESUMEN

This study investigated the impact of prolonged nicotine administration on myocardial susceptibility to ischaemia-reperfusion (I/R) injury in a rat model and determined whether nicotine affects mitochondrial reactive oxygen species (ROS) production and permeability transition in rat hearts. Sprague-Dawley rats were administered 0.6 or 1.2 mg/kg nicotine for 28 days, and their hearts were isolated at end-point for assessment of myocardial susceptibility to I/R injury ex vivo. Rat heart mitochondria were also isolated from a subset of rats for analysis of mitochondrial ROS production and permeability transition. Compared to the vehicle controls, rat hearts isolated from nicotine-administered rats exhibited poorer left ventricular function that worsened over the course of I/R. Coronary flow rate was also severely impaired in the nicotine groups at baseline and this worsened after I/R. Nicotine administration significantly increased mitochondrial ROS production and permeability transition relative to the vehicle controls. Interestingly, pre-incubation of isolated mitochondria with ROS scavengers (superoxide dismutase and mitoTEMPO) significantly abolished nicotine-induced increase in mitochondria permeability transition in isolated rat heart mitochondria. Overall, our data showed that prolonged nicotine administration enhances myocardial susceptibility to I/R injury in rats and this is associated with mitochondrial ROS-driven increase in mitochondrial permeability transition.


Asunto(s)
Daño por Reperfusión Miocárdica/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Animales , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Esquema de Medicación , Preparación de Corazón Aislado , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
13.
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
14.
Peptides ; 137: 170474, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359394

RESUMEN

Isoprenaline-induced cardiac hypertrophy can deteriorate to heart failure, which is a leading cause of mortality. Endogenous vasonatrin peptide (VNP) has been reported to be cardioprotective against myocardial ischemia/reperfusion injury in diabetic rats. However, little is known about the effect of exogenous VNP on cardiac hypertrophy. We further explored whether VNP attenuated isoprenaline-induced cardiomyocyte hypertrophy by examining the levels and activities of cGMP and PKG. In this study, we found that VNP significantly attenuated isoprenaline-induced myocardial hypertrophy and cardiac fibroblast activation in vivo. Moreover, VNP effectively halted the activation of apoptosis and oxidative stress in the isoprenaline-treated myocardium. VNP promoted superoxide dismutase (SOD) activity. Further study revealed that the protective effects of VNP might be mediated by the activity of the cGMP-PKG signaling pathway in vivo or in vitro, while the use of agonists and antagonists confirmed these results. Therefore, we demonstrated that the antiapoptosis and antioxidative stress effects of VNP depends on elevated cGMP-PKG signaling activity both in vivo and in vitro. These results suggest that VNP may be used in the treatment of myocardial hypertrophy.


Asunto(s)
Factor Natriurético Atrial/farmacología , Cardiomegalia/tratamiento farmacológico , Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , GMP Cíclico/genética , Humanos , Isoproterenol/toxicidad , Ratones , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Cultivo Primario de Células , Ratas , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/genética
15.
Drug Des Devel Ther ; 14: 5275-5288, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33299300

RESUMEN

INTRODUCTION: Cyclophosphamide (CP) causes redox imbalance and its use is associated with marked cardiotoxicity that limits its clinical applications. The present study investigated the protective effects of acetovanillone (AV) and edaravone (ED) against CP-induced oxidative stress and cardiac damage, emphasizing the role of PI3K/Akt/mTOR and Nrf2 signaling. MATERIALS AND METHODS: Rats received either AV (100 mg/kg) or ED (20 mg/kg) orally for 10 days and CP (200 mg/kg) on day 7. At day 11, the rats were sacrificed, and samples were collected for analysis. RESULTS: AV and ED ameliorated serum troponin I, CK-MB, LDH, AST and ALP, and prevented cardiac histological alterations in CP-intoxicated rats. Both treatments decreased cardiac lipid peroxidation and enhanced GSH, SOD and cytoglobin in CP-induced rats. AV and ED downregulated Keap1, whereas increased the expression of PI3K, Akt, mTOR and Nrf2 in the heart of rats received CP. Additionally, the binding modes of AV and ED to Keap1 were pinpointed in silico using molecular docking simulations. CONCLUSION: AV and ED prevent CP cardiotoxicity by attenuating oxidative stress and tissue injury, and modulating cytoglobin, and PI3K/Akt/mTOR and Keap1/Nrf2 signaling. Therefore, AV and ED may represent promising agents that can prevent cardiac injury in patients receiving CP.


Asunto(s)
Acetofenonas/farmacología , Antioxidantes/farmacología , Edaravona/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Acetofenonas/administración & dosificación , Administración Oral , Animales , Antioxidantes/administración & dosificación , Ciclofosfamida , Edaravona/administración & dosificación , Masculino , Simulación del Acoplamiento Molecular , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/prevención & control , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
16.
Eur Rev Med Pharmacol Sci ; 24(22): 11740-11746, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33275272

RESUMEN

OBJECTIVE: The purpose of this study was to explore the influence of Ghrelin on myocardial injury of septic rats through the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway. MATERIALS AND METHODS: A total of 36 Sprague-Dawley rats were randomly divided into normal group (n=12), model group (n=12), and Ghrelin group (n=12). The rats in the normal group were fed normally, while those in the model group were intraperitoneally injected with endotoxin to establish the sepsis model. The rats in the Ghrelin group were given intraperitoneal injection of Ghrelin solution to prepare the sepsis model. 9 h later, the specimens were obtained. Then, the expressions of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were detected via immunohistochemistry, and the protein expressions of phosphorylated JAK (p-JAK) and STAT3 were determined by Western blotting (WB). Next, enzyme-linked immunosorbent assay (ELISA) was performed to measure the content of IL-6 and TNF-α, and quantitative Polymerase Chain Reaction (qPCR) was applied to examine the messenger ribonucleic acid (mRNA) expressions of JAK and STAT3. Finally, the cell apoptosis was detected through terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. RESULTS: The results of immunohistochemistry showed that compared with those in the normal group, the positive expression levels of IL-6 and TNF-α were markedly increased in other groups (p<0.05), while in comparison with those in the model group, the positive expression levels of IL-6 and TNF-α were decreased significantly in the Ghrelin group (p<0.05). The WB results indicated that the model group and Ghrelin group had remarkably higher protein expression levels of p-JAK and STAT3 than the normal group (p<0.05), and Ghrelin group exhibited notably lower protein expression levels of p-JAK and STAT3 than the model group (p<0.05). According to the results of qPCR, the relative mRNA expression levels of JAK and STAT3 were distinctly raised in the model group and Ghrelin group in comparison with those in the normal group (p<0.05), while they were reduced evidently in the Ghrelin group compared with those in the model group (p<0.05). Furthermore, it was manifested in the results of ELISA that the model group and Ghrelin group had prominently elevated content of TNF-α and IL-6 compared with normal group (p<0.05), and Ghrelin group displayed significantly lowered content of TNF-α and IL-6 in comparison with the model group (p<0.05). Moreover, the TUNEL results revealed that the apoptosis rate was remarkably higher in the other two groups than that in the normal group (p<0.05), while it was evidently lower in the Ghrelin group than that in the model group (p<0.05). CONCLUSIONS: Ghrelin can inhibit inflammatory response and apoptosis in the process of myocardial injury in septic rats by repressing the JAK/STAT signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Ghrelina/farmacología , Inflamación/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Ghrelina/administración & dosificación , Inflamación/inducido químicamente , Inflamación/patología , Inyecciones Intraperitoneales , Quinasas Janus/metabolismo , Lipopolisacáridos/administración & dosificación , Masculino , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/patología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Sepsis/inducido químicamente , Sepsis/patología , Transducción de Señal/efectos de los fármacos
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(11): 1009-1015, 2020 Nov.
Artículo en Chino | MEDLINE | ID: mdl-33210595

RESUMEN

Objective To investigate the role and mechanism of endothelin receptor antagonist CPU0213 in myocardial ischemia-reperfusion (I/R) injury and oxidative stress injury. Methods In order to investigate the role of CPU0213 in I/R, SD rats were randomly divided into sham operation group, ischemia reperfusion injury (I/R) group, CPU0213 treatment group after I/R, CPU0213 and Janus kinase 2 (JAK2) specific inhibitor AG490 treatment group after I/R. In order to investigate the role of CPU0213 in oxidative stress damage, the isolated and characterized cardiomyocytes were cultured and divided into control group, H2O2 oxidative stress group (H2O2 group), oxidative stress damaged group treated with CPU0213, and oxidative stress damaged group treated with CPU0213 and AG490. The rat myocardial I/R models were constructed, and the rats and cardiomyocytes were treated with different treatments according to the experimental requirements. The rat heart was stained with triphenyltetrazolium chloride (TTC) to observe the area of myocardial infarction and the lactate dehydrogenase (LDH) and creatine kinase (CK) activity, flow cytometry to detect the apoptosis rate of cardiomyocytes, CCK-8 method to detect cell growth viability, Western blotting to detect the expression of Bcl2, JAK2, phosphorylated JAK2 (p-JAK2), caspase-3 and caspase-9, STAT3 and phosphorylated STAT3 (p-STAT3). Results After I/R injury in mice, CPU0213 treatment reduced myocardial infarction area, LDH, CK activity and apoptosis rate, but increased the phosphorylation level of JAK2 and STAT3. Compared with I/R combined with CPU0213, I/R combined with CPU0213 and AG490 increased the expression of caspase-3 and caspase-9, decreased significantly the expression of Bcl2 and the cell viability. After oxidative stress damage to cardiomyocytes, CPU0213 treatment reduced LDH, CK activity and cell apoptosis rate, and increased the phosphorylation level of JAK2 and STAT3. In the oxidative stress damaged group treated with CPU0213 and AG490, caspase-3 and caspase-9 expression increased, Bcl2 expression dropped significantly, cell viability decreased significantly. Conclusion The activation of JAK2/STAT3 pathway by CPU0213 can inhibit the apoptosis of cardiomyocytes induced by I/R and oxidative stress.


Asunto(s)
Janus Quinasa 2 , Daño por Reperfusión Miocárdica , Pirazoles , Factor de Transcripción STAT3 , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Peróxido de Hidrógeno/toxicidad , Janus Quinasa 2/metabolismo , Ratones , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Pirazoles/farmacología , Pirazoles/uso terapéutico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Eur Rev Med Pharmacol Sci ; 24(17): 9055-9062, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32964996

RESUMEN

OBJECTIVE: Oxidative stress is one of the main factors leading to myocardial cell damage, and the redox imbalance promotes apoptosis. Therefore, the purpose of this study was to explore the protective effect of PrxII on H2O2-induced H9c2 cell injury. MATERIALS AND METHODS: The overexpressed PrxII cell model was constructed by virus. The H9c2 cells were treated with H2O2, and the supernatant and cells were collected. Then, the chymotrypsin-like activity, caspase-like activity, and trypsin-like activity were detected by the kit, and the expressions of P21, P27, and P53 were detected by the ELISA method. Finally, the expressions of antioxidant factors, apoptosis-related factors, and AMPK/Sirt1 signaling pathway were detected by Western blot and Real-time polymerase chain reaction (PCR). RESULTS: Overexpression of PrxII inhibited the decrease of enzyme activity induced by H2O2, promoted the expressions of anti-oxidation factors GPX1, GPX2, and GSX, and inhibited the expressions of apoptosis-related factors P21, P27, and P53, and activated AMPK/Sirt1 pathway. CONCLUSIONS: Overexpression of PrxII can activate the AMPK/Sirt1 pathway, thereby inhibiting H2O2-induced oxidative stress and slowing apoptosis.


Asunto(s)
Antioxidantes/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Peroxirredoxinas/metabolismo , Sustancias Protectoras/metabolismo , Animales , Células Cultivadas , Peróxido de Hidrógeno , Daño por Reperfusión Miocárdica/inducido químicamente , Peroxirredoxinas/genética , Ratas
19.
Eur J Med Res ; 25(1): 29, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32738910

RESUMEN

BACKGROUND: Electroacupuncture is well known for its advantageous neuroanalgesic and therapeutic effects on myocardial ischemia-reperfusion injury. The purpose of the present research was to verify whether electroacupuncture can alleviate bupivacaine-induced myocardial injury. METHODS: Specific pathogen-free Wistar rats were used to establish the bupivacaine-induced myocardial injury model. Western blot, PCR, transmission electron microscope and enzyme-linked immunosorbent (ELISA) methods were used to evaluate bupivacaine-induced structure injury and dysfunction of the mitochondria as well as the alleviating effects of lipid emulsion, acupoint injection, and electroacupuncture pre-treatment of the oxidase stress response. RESULTS: Bupivacaine caused structural damage, degradation, and swelling of mitochondria. Furthermore, it reduced adenosine triphosphate (ATP) synthesis and impaired energy metabolism in the mitochondria. Structural and functional impairment of the mitochondria was alleviated via lipid emulsion injection, acupoint injection, and electroacupuncture pre-treatment. Electroacupuncture pre-treatment of PC6 yielded a greater alleviating effect than others approaches. Following electroacupuncture pre-treatment of PC6 point, the number of mitochondria increased; apoptosis was reduced, enzymatic activity of cytochrome C oxidase (COX) and superoxide dismutase and expression of uncoupling protein 2, voltage-dependent anion channel 1, and Bcl 2 were upregulated and SLC25A6, MDA levels were downregulated. Additionally, our findings indicated that electroacupuncture pre-treatment of PC6 point exerted an effect on the mitochondria via the mitochondrial-transcription-factor-A/nuclear-respiratory-factor-1/proliferator-activated-receptor-gamma-coactivator-1 pathway. CONCLUSION: The present study revealed that electroacupuncture pre-treatment of PC6 could effectively alleviate bupivacaine-induced myocardial mitochondrial damage, thereby providing a theoretical basis for clinical studies and applications of this treatment method.


Asunto(s)
Bupivacaína/toxicidad , Electroacupuntura/métodos , Mitocondrias Cardíacas/fisiología , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Anestésicos Locales/toxicidad , Animales , Apoptosis , Masculino , Proteínas Mitocondriales/genética , Daño por Reperfusión Miocárdica/inducido químicamente , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Estrés Oxidativo , Ratas , Ratas Wistar
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA