Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 607
Filtrar
Más filtros

Intervalo de año de publicación
1.
Can J Physiol Pharmacol ; 102(5): 331-341, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38118123

RESUMEN

Extracellular potassium concentration might modify electrophysiological properties in the border zone of ischemic myocardium. We evaluated the depolarization and repolarization characteristics across the ischemic-normal border under [K+] variation. Sixty-four-lead epicardial mapping was performed in 26 rats ([K+] 2.3-6.4 mM) in a model of acute ischemia/reperfusion. The animals with [K+] < 4.7 mM (low-normal potassium) had an ischemic zone with ST-segment elevation and activation delay, a border zone with ST-segment elevation and no activation delay, and a normal zone without electrophysiological abnormalities. The animals with [K+] >4.7 mM (normal-high potassium) had only the ischemic and normal zones and no transitional area. Activation-repolarization intervals and local conduction velocities were inversely associated with [K+] in linear regression analysis with adjustment for the zone of myocardium. The reperfusion extrasystolic burden (ESB) was greater in the low-normal as compared to normal-high potassium animals. Ventricular tachycardia/fibrillation incidence did not differ between the groups. In patch-clamp experiments, hypoxia shortened action potential duration at 5.4 mM but not at 1.3 mM of [K+]. IK(ATP) current was lower at 1.3 mM than at 5.4 mM of [K+]. We conclude that the border zone formation in low-normal [K+] was associated with attenuation of IK(ATP) response to hypoxia and increased reperfusion ESB.


Asunto(s)
Potenciales de Acción , Isquemia Miocárdica , Potasio , Animales , Potasio/sangre , Potasio/metabolismo , Masculino , Ratas , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/sangre , Isquemia Miocárdica/metabolismo , Potenciales de Acción/fisiología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Ratas Wistar
2.
Scand Cardiovasc J ; 58(1): 2347290, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38733316

RESUMEN

Objectives. The aim of this study was to investigate the expression of long non-coding RNA (lncRNA) brain and reproductive organ-expressed protein (BRE) antisense RNA 1 (BRE-AS1) in patients with acute myocardial infarction (AMI) and its effect on ischemia/reperfusion (I/R)-induced oxidative stress and apoptosis of cardiomyocytes. Methods. Serum BRE-AS1 levels in patients with AMI was detected using quantitative real-time polymerase chain reaction (qRT-PCR). The diagnostic and prognostic values of BRE-AS1 were evaluated. H9c2 cells were treated with hypoxia/reoxygenation to establish an in vitro myocardial infarction cell model. The levels of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 were detected by enzyme-linked immunosorbent assay (ELISA). Levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were determined by commercial kits. Cell counting kit-8 (CCK-8) and flow cytometry were used to evaluate the cell viability and cell apoptosis. Results. The expression of BRE-AS1 in serum of patients with AMI is upregulated, which shows the clinical diagnostic value for AMI. In the I/R injury cell model, the knockout of BRE-AS1 can significantly alleviate the increase in TNF-α, IL-1ß, and IL-6 levels, inhibit the production of LDH and MDA, increase the activities of SOD and GSH-Px, promote the cell viability and suppress cell apoptosis. Conclusions. Abnormally elevated BRE-AS1 has a high diagnostic value for AMI as well as a prognostic value for major adverse cardiovascular events (MACEs). The elevation of BRE-AS1 promoted oxidative stress injury and cell apoptosis in vitro.


Asunto(s)
Apoptosis , Mediadores de Inflamación , Infarto del Miocardio , Miocitos Cardíacos , Estrés Oxidativo , ARN Largo no Codificante , Animales , Femenino , Humanos , Masculino , Ratas , Estudios de Casos y Controles , Línea Celular , Citocinas/metabolismo , Citocinas/sangre , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/sangre , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Infarto del Miocardio/diagnóstico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Largo no Codificante/sangre , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Regulación hacia Arriba
3.
J Cell Physiol ; 236(9): 6657-6665, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33554327

RESUMEN

Oxygen is often administered to patients and occasionally to healthy individuals as well; however, the cellular toxicity of oxygen, especially following prolonged exposure, is widely known. To evaluate the potential effect of oxygen exposure on circulating stem/progenitor cells and cardiac ischemia/reperfusion (I/R) injury, we exposed healthy adult mice to 100% oxygen for 20 or 60 min. We then examined the c-kit-positive stem/progenitor cells and colony-forming cells and measured the cytokine/chemokine levels in peripheral blood. We also induced cardiac I/R injury in mice at 3 h after 60 min of oxygen exposure and examined the recruitment of inflammatory cells and the fibrotic area in the heart. The proportion of c-kit-positive stem/progenitor cells significantly increased in peripheral blood at 3 and 24 h after oxygen exposure for either 20 or 60 min (p < .01 vs. control). However, the abundance of colony-forming cells in peripheral blood conversely decreased at 3 and 24 h after oxygen exposure for only 60 min (p < .05 vs. control). Oxygen exposure for either 20 or 60 min resulted in significantly decreased plasma vascular endothelial growth factor levels at 3 h, whereas oxygen exposure for only 60 min reduced plasma insulin-like growth factor 1 levels at 24 h (p < .05 vs. control). Protein array indicated the increase in the levels of some cytokines/chemokines, such as CXCL6 (GCP-2) at 24 h after 60 min of oxygen exposure. Moreover, oxygen exposure for 60 min enhanced the recruitment of Ly6g- and CD11c-positive inflammatory cells at 3 days (p < .05 vs. control) and increased the fibrotic area at 14 days in the heart after I/R injury (p < .05 vs. control). Prolonged oxygen exposure induced the mobilization and functional impairment of stem/progenitor cells and likely enhanced inflammatory responses to exacerbate cardiac I/R injury in healthy mice.


Asunto(s)
Daño por Reperfusión Miocárdica/patología , Oxígeno/efectos adversos , Células Madre/patología , Animales , Quimiocina CXCL12/sangre , Ensayo de Unidades Formadoras de Colonias , Mediadores de Inflamación/sangre , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/sangre , Miocardio/metabolismo , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor A de Crecimiento Endotelial Vascular/sangre
4.
Microcirculation ; 28(1): e12657, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32892409

RESUMEN

OBJECTIVE: This study examined the correlation between serum miR-98-5p levels and indices of microvascular reperfusion in patients undergoing primary percutaneous coronary intervention (pPCI) after ST-segment elevation myocardial infarction (STEMI). Additionally, we evaluated the mechanisms by which miR-98-5p promoted ischemia/reperfusion (I/R)-induced injury in both cultured cell lines and an animal model. METHODS: Circulating miR-98-5p levels were measured and compared from 171 STEMI patients undergoing pPCI, who were divided into two groups: no-reflow and reflow. The levels of miR-98-5p, nerve growth factor (NGF), and transient receptor potential vanilloid 1 (TRPV1) were analyzed in cultured human coronary endothelial cells (HCECs) exposed to hypoxia/reoxygenation (H/R). The effects of antagomir-98-5p on myocardial I/R-induced microvascular dysfunction in vivo were evaluated. Target gene expression and activity were assessed. RESULTS: Higher miR-98-5p levels were associated with compromised indices of microvascular reperfusion. In vitro experiments on HCECs showed that exposure to H/R significantly increased miR-98-5p levels. We identified NGF as a novel target of miR-98-5p. Further, antagomir-98-5p relieved microvascular dysfunction and enhanced the expression of NGF and TRPV1 in the rat myocardial I/R model. CONCLUSIONS: MiR-98-5p promotes microvascular dysfunction by targeting the NGF-TRPV1 axis. Serum miR-98-5p serves as a potential biomarker for microvascular reperfusion.


Asunto(s)
Vasos Coronarios/metabolismo , MicroARNs/sangre , Microvasos/metabolismo , Daño por Reperfusión Miocárdica/sangre , Factor de Crecimiento Nervioso/sangre , Anciano , Biomarcadores/sangre , Células Cultivadas , Vasos Coronarios/patología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Femenino , Estudios de Seguimiento , Regulación de la Expresión Génica , Humanos , Masculino , Microvasos/patología , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/patología
5.
Basic Res Cardiol ; 116(1): 36, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34037861

RESUMEN

We studied the translational cardioprotective potential of P2Y12 inhibitors against acute myocardial ischemia/reperfusion injury (IRI) in an animal model of acute myocardial infarction and in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI). P2Y12 inhibitors may have pleiotropic effects to induce cardioprotection against acute myocardial IRI beyond their inhibitory effects on platelet aggregation. We compared the cardioprotective effects of clopidogrel, prasugrel, and ticagrelor on infarct size in an in vivo rat model of acute myocardial IRI, and investigated the effects of the P2Y12 inhibitors on enzymatic infarct size (48-h area-under-the-curve (AUC) troponin T release) and clinical outcomes in a retrospective study of STEMI patients from the CONDI-2/ERIC-PPCI trial using propensity score analyses. Loading with ticagrelor in rats reduced infarct size after acute myocardial IRI compared to controls (37 ± 11% vs 52 ± 8%, p < 0.01), whereas clopidogrel and prasugrel did not (50 ± 11%, p > 0.99 and 49 ± 9%, p > 0.99, respectively). Correspondingly, troponin release was reduced in STEMI patients treated with ticagrelor compared to clopidogrel (adjusted 48-h AUC ratio: 0.67, 95% CI 0.47-0.94). Compared to clopidogrel, the composite endpoint of cardiac death or hospitalization for heart failure within 12 months was reduced in STEMI patients loaded with ticagrelor (HR 0.63; 95% CI 0.42-0.94) but not prasugrel (HR 0.84, 95% CI 0.43-1.63), prior to PPCI. Major adverse cardiovascular events did not differ between clopidogrel, ticagrelor, or prasugrel. The cardioprotective effects of ticagrelor in reducing infarct size may contribute to the clinical benefit observed in STEMI patients undergoing PPCI.


Asunto(s)
Plaquetas/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Intervención Coronaria Percutánea , Inhibidores de Agregación Plaquetaria/uso terapéutico , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Receptores Purinérgicos P2Y12/efectos de los fármacos , Infarto del Miocardio con Elevación del ST/terapia , Investigación Biomédica Traslacional , Anciano , Animales , Plaquetas/metabolismo , Clopidogrel/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/fisiopatología , Intervención Coronaria Percutánea/efectos adversos , Inhibidores de Agregación Plaquetaria/efectos adversos , Clorhidrato de Prasugrel/uso terapéutico , Antagonistas del Receptor Purinérgico P2Y/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Receptores Purinérgicos P2Y12/metabolismo , Estudios Retrospectivos , Infarto del Miocardio con Elevación del ST/fisiopatología , Ticagrelor/uso terapéutico , Resultado del Tratamiento
6.
Microvasc Res ; 138: 104235, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34453991

RESUMEN

Electroacupuncture (EA) intervention has a remarkable cardioprotection against myocardial ischemia reperfusion injury (MIRI). Recently, it has been suggested that the gut microbiota plays an important role in regulating the progression and prognosis of MIRI. The purpose of this study was to illustrate the relationship between gut microbiota and cardioprotection of EA on MIRI. We conducted a MIRI model by ligating the left anterior descending coronary artery for 30 min followed by reperfusion in male Sprague Dawley rats, which then received 7 days of EA intervention. Echocardiography was employed to evaluate left ventricular function. Fecal samples were collected for microbial analysis by 16S rDNA high-throughput sequencing. Blood samples and myocardium were collected for inflammatory cytokine detection by enzyme linked immunosorbent assay (ELISA) and Western blot. Hematoxylin & eosin (HE) staining and immunofluorescence of ileum tissue were performed for intestinal damage evaluation. After 7 days of EA intervention, the left ventricular function was improved with significantly increased ejection fraction and fractional shortening. Furthermore, we found that EA intervention reversed the changed gut microbiota induced by MIRI, including Clostridiales, RF39, S24-7, Desulfovibrio, and Allobaculum, improved the impaired gut barrier, reduced the production and circulation of lipopolysaccharide (LPS), inhibited the level of interleukin 6 (IL-6) and interleukin 12 (IL-12) in periphery and decreased the expression of Toll like receptor 4 (TLR4) and IL-6 in myocardium. EA intervention could improve the impaired gut mucosal barrier and reduce the production and circulation of LPS after MIRI through regulating gut microbiota, thus inhibiting the circulation and myocardium inflammation and finally exerted the cardioprotective effect.


Asunto(s)
Bacterias/metabolismo , Electroacupuntura , Microbioma Gastrointestinal , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/microbiología , Lipopolisacáridos/sangre , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Proteínas de Fase Aguda , Animales , Bacterias/crecimiento & desarrollo , Proteínas Portadoras/sangre , Modelos Animales de Enfermedad , Disbiosis , Masculino , Glicoproteínas de Membrana/sangre , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/microbiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Sprague-Dawley , Función Ventricular Izquierda
7.
Cell Biochem Funct ; 39(2): 287-295, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32767595

RESUMEN

Ischaemia/reperfusion (I/R) injury can lead to severe arrhythmia and aggravate myocardial damage. Exosomes are small-membrane vesicles that play a protective role in myocardial I/R injury. This study aimed to explore the protective effects of ischaemic preconditioning (IPC)-induced serum exosomes (IPC-Exo) on myocardial I/R injury in rats and its underlying mechanism. Serum exosomes were extracted from IPC rats and quantified using a bicinchoninic acid assay kit. IPC-Exo (50 µg) was injected into the infarcted myocardium immediately after ligation. Rats were randomly divided into Sham, I/R, IPC-Exo + I/R, I/R + LY294002, and I/R + IPC-Exo + LY294002 groups. Haemodynamic parameters were measured by physiological recording. Transthoracic echocardiography was used to detect cardiac function. The serum levels of creatine kinase isomer-MB, lactate dehydrogenase, aspartate transaminase, tumour necrosis factor-alpha, interleukin (IL)-1ß, and IL-10 were detected by enzyme-linked immunosorbent assay. Triphenyl tetrazolium chloride staining was used to measure the myocardial infarct size. Apoptosis in myocardial tissues was detected by TUNEL staining. Western blotting was used to detect the levels of PI3K/AKT and apoptosis-related proteins. Our results showed that treatment with IPC-Exo ameliorated cardiac function and reduced inflammatory factor production, cardiomyocyte apoptosis, and myocardial infarct size. Moreover, IPC-Exo treatment promoted the protein expression of Bcl-2, p-PI3K, and p-AKT but inhibited that of caspase-3 and Bax. However, treatment with LY294002 significantly reversed that IPC-Exo-induced increase in p-PI3K and p-AKT levels, improvement of haemodynamics, and decrease of inflammatory factor production and apoptosis in the I/R + IPC-Exo group. Taken together, our results suggest that IPC-Exo may alleviate I/R injury via activating the PI3K/AKT signalling pathway.


Asunto(s)
Exosomas/metabolismo , Precondicionamiento Isquémico , Daño por Reperfusión Miocárdica/patología , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Cromonas/farmacología , Forma MB de la Creatina-Quinasa/metabolismo , Hemodinámica/efectos de los fármacos , Masculino , Morfolinas/farmacología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tetraspanina 30/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Función Ventricular Izquierda/fisiología
8.
Circulation ; 139(7): 918-931, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30586717

RESUMEN

BACKGROUND: Platelets store large amounts of serotonin that they release during thrombus formation or acute inflammation. This facilitates hemostasis and modulates the inflammatory response. METHODS: Infarct size, heart function, and inflammatory cell composition were analyzed in mouse models of myocardial reperfusion injury with genetic and pharmacological depletion of platelet serotonin. These studies were complemented by in vitro serotonin stimulation assays of platelets and leukocytes in mice and men, and by measuring plasma serotonin levels and leukocyte activation in patients with acute coronary syndrome. RESULTS: Platelet-derived serotonin induced neutrophil degranulation with release of myeloperoxidase and hydrogen peroxide (H2O2) and increased expression of membrane-bound leukocyte adhesion molecule CD11b, leading to enhanced inflammation in the infarct area and reduced myocardial salvage. In patients hospitalized with acute coronary syndrome, plasmatic serotonin levels correlated with CD11b expression on neutrophils and myeloperoxidase plasma levels. Long-term serotonin reuptake inhibition-reported to protect patients with depression from cardiovascular events-resulted in the depletion of platelet serotonin stores in mice. These mice displayed a reduction in neutrophil degranulation and preserved cardiac function. In line, patients with depression using serotonin reuptake inhibition, presented with suppressed levels of CD11b surface expression on neutrophils and lower myeloperoxidase levels in blood. CONCLUSIONS: Taken together, we identify serotonin as a potent therapeutic target in neutrophil-dependent thromboinflammation during myocardial reperfusion injury.


Asunto(s)
Plaquetas/metabolismo , Degranulación de la Célula , Infarto del Miocardio/sangre , Daño por Reperfusión Miocárdica/sangre , Miocardio/metabolismo , Neutrófilos/metabolismo , Serotonina/sangre , Síndrome Coronario Agudo/sangre , Animales , Antígeno CD11b/sangre , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Humanos , Peróxido de Hidrógeno/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Neutrófilos/patología , Peroxidasa/sangre , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/genética
9.
Circulation ; 139(23): 2654-2663, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905171

RESUMEN

BACKGROUND: Nitrosation of a conserved cysteine residue at position 93 in the hemoglobin ß chain (ß93C) to form S-nitroso (SNO) hemoglobin (Hb) is claimed to be essential for export of nitric oxide (NO) bioactivity by the red blood cell (RBC) to mediate hypoxic vasodilation and cardioprotection. METHODS: To test this hypothesis, we used RBCs from mice in which the ß93 cysteine had been replaced with alanine (ß93A) in a number of ex vivo and in vivo models suitable for studying export of NO bioactivity. RESULTS: In an ex vivo model of cardiac ischemia/reperfusion injury, perfusion of a mouse heart with control RBCs (ß93C) pretreated with an arginase inhibitor to facilitate export of RBC NO bioactivity improved cardiac recovery after ischemia/reperfusion injury, and the response was similar with ß93A RBCs. Next, when human platelets were coincubated with RBCs and then deoxygenated in the presence of nitrite, export of NO bioactivity was detected as inhibition of ADP-induced platelet activation. This effect was the same in ß93C and ß93A RBCs. Moreover, vascular reactivity was tested in rodent aortas in the presence of RBCs pretreated with S-nitrosocysteine or with hemolysates or purified Hb treated with authentic NO to form nitrosyl(FeII)-Hb, the proposed precursor of SNO-Hb. SNO-RBCs or NO-treated Hb induced vasorelaxation, with no differences between ß93C and ß93A RBCs. Finally, hypoxic microvascular vasodilation was studied in vivo with a murine dorsal skin-fold window model. Exposure to acute systemic hypoxia caused vasodilatation, and the response was similar in ß93C and ß93A mice. CONCLUSIONS: RBCs clearly have the fascinating ability to export NO bioactivity, but this occurs independently of SNO formation at the ß93 cysteine of Hb.


Asunto(s)
Plaquetas/metabolismo , Eritrocitos/metabolismo , Hemoglobinas/metabolismo , Daño por Reperfusión Miocárdica/sangre , Óxido Nítrico/sangre , Piel/irrigación sanguínea , Globinas beta/metabolismo , Alanina , Sustitución de Aminoácidos , Animales , Transporte Biológico , Cisteína , Modelos Animales de Enfermedad , Hemoglobinas/genética , Humanos , Hipoxia/sangre , Hipoxia/fisiopatología , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Daño por Reperfusión Miocárdica/fisiopatología , Activación Plaquetaria , Ratas Sprague-Dawley , Vasodilatación , Función Ventricular Izquierda , Presión Ventricular , Globinas beta/genética
10.
Am J Physiol Heart Circ Physiol ; 319(1): H32-H41, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32412785

RESUMEN

Disruption of the normal gut microbiome (dysbiosis) is implicated in the progression and severity of myriad disorders, including hypercholesterolemia and cardiovascular disease. Probiotics attenuate and reverse gut dysbiosis to improve cardiovascular risk factors like hypertension and hypercholesterolemia. Lactobacillus reuteri is a well-studied lactic acid-producing probiotic with known cholesterol-lowering properties and anti-inflammatory effects. In the present study, we hypothesized that L. reuteri delivered to hypercholesterolemic low-density lipoprotein receptor knockout (LDLr KO) mice will reduce cholesterol levels and minimize cardiac injury from an ischemic insult. L. reuteri [1 × 109 or 50 × 106 colony-forming units (CFU)/day] was administered by oral gavage to wild-type mice and LDLr KO for up to 6 wk followed by an ischemia-reperfusion (I/R) protocol. After 4 wk of gavage, total serum cholesterol in wild-type mice receiving saline was 113.5 ± 5.6 mg/dL compared with 113.3 ± 6.8 and 101.9 ± 7.5 mg/dL in mice receiving 1 × 109 or 50 × 106 CFU/day, respectively. Over the same time frame, administration of L. reuteri at 1 × 109 or 50 × 106 CFU/day did not lower total serum cholesterol (283.0 ± 11.1, 263.3 ± 5.0, and 253.1 ± 7.0 mg/dL; saline, 1 × 109 or 50 × 106 CFU/day, respectively) in LDLr KO mice. Despite no impact on total serum cholesterol, L. reuteri administration significantly attenuated cardiac injury following I/R, as evidenced by smaller infarct sizes compared with controls in both wild-type and LDLr KO groups. In conclusion, daily L. reuteri significantly protected against cardiac injury without lowering cholesterol levels, suggesting anti-inflammatory properties of L. reuteri uncoupled from improvements in serum cholesterol.NEW & NOTEWORTHY We demonstrated that daily delivery of Lactobacillus reuteri to wild-type and hypercholesterolemic lipoprotein receptor knockout mice attenuated cardiac injury following ischemia-reperfusion without lowering total serum cholesterol in the short term. In addition, we validated protection against cardiac injury using histology and immunohistochemistry techniques. L. reuteri offers promise as a probiotic to mitigate ischemic cardiac injury.


Asunto(s)
Colesterol/sangre , Daño por Reperfusión Miocárdica/microbiología , Probióticos/uso terapéutico , Animales , Microbioma Gastrointestinal , Limosilactobacillus reuteri/patogenicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/prevención & control , Probióticos/administración & dosificación , Receptores de LDL/deficiencia , Receptores de LDL/genética
11.
Cell Mol Life Sci ; 76(20): 4103-4115, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31250032

RESUMEN

Cardiovascular diseases (CVDs) are among the leading threats to human health. The advanced glycation end product (AGE) and receptor for AGE (RAGE) signaling pathway regulates the pathogenesis of CVDs, through its effects on arterial stiffness, atherosclerosis, mitochondrial dysfunction, oxidative stress, calcium homeostasis, and cytoskeletal function. Targeting the AGE/RAGE pathway is a potential therapeutic strategy for ameliorating CVDs. Vitamin D has several beneficial effects on the cardiovascular system. Experimental findings have shown that vitamin D regulates AGE/RAGE signaling and its downstream effects. This article provides a comprehensive review of the mechanistic insights into AGE/RAGE involvement in CVDs and the modulation of the AGE/RAGE signaling pathways by vitamin D.


Asunto(s)
Cardiomiopatías Diabéticas/prevención & control , Productos Finales de Glicación Avanzada/genética , Daño por Reperfusión Miocárdica/prevención & control , Miocarditis/prevención & control , Receptor para Productos Finales de Glicación Avanzada/genética , Vitamina D/uso terapéutico , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Animales , Calcio/metabolismo , Cardiotónicos/uso terapéutico , Cardiomiopatías Diabéticas/sangre , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Regulación de la Expresión Génica , Productos Finales de Glicación Avanzada/sangre , Guanidinas/uso terapéutico , Humanos , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocarditis/sangre , Miocarditis/genética , Miocarditis/patología , Estrés Oxidativo/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/sangre , Transducción de Señal , Tiazoles/uso terapéutico , Rigidez Vascular/efectos de los fármacos , Vitamina D/sangre
12.
Thorac Cardiovasc Surg ; 68(3): 223-231, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31022735

RESUMEN

BACKGROUND: The aim of this study is to compare the combined use of the Myocardial Protection System and our microplegia (Basel Microplegia Protocol) with Cardioplexol® in coronary artery bypass grafting using the minimal extracorporeal circulation. METHODS: The analysis focused on propensity score matched pairs of patients in whom microplegia or Cardioplexol® was used. Primary efficacy endpoints were high-sensitivity cardiac troponin-T on postoperative day 1 and peak values during hospitalization. Furthermore, we assessed creatine kinase and creatinine kinase-myocardial type, as well as safety endpoints. RESULTS: A total of 56 patients who received microplegia and 155 patients who received Cardioplexol® were included. The use of the microplegia was associated with significantly lower geometric mean (confidence interval) peak values of high-sensitivity cardiac troponin-T (233 ng/L [194-280 ng/L] vs. 362 ng/L [315-416 ng/L]; p = 0.001), creatinine kinase (539 U/L [458-633 U/L] vs. 719 U/L [645-801 U/L]; p = 0.011), and creatinine kinase-myocardial type (13.8 µg/L [9.6-19.9 µg/L] vs. 21.6 µg/L [18.9-24.6 µg/L]; p = 0.026), and a shorter length of stay on the intensive care unit (1.5 days [1.2-1.8 days] vs. 1.9 days [1.7-2.1 days]; p = 0.011). Major adverse cardiac and cerebrovascular events occurred with roughly equal frequency (1.8 vs. 5.2%; p = 0.331). CONCLUSIONS: The use of the Basel Microplegia Protocol was associated with lower peak values of high-sensitivity cardiac troponin-T, creatinine kinase, and creatinine kinase-myocardial type and with a shorter length of stay on the intensive care unit, as compared with the use of Cardioplexol® in isolated coronary artery bypass surgery using minimal extracorporeal circulation.


Asunto(s)
Soluciones Cardiopléjicas/administración & dosificación , Puente Cardiopulmonar , Puente de Arteria Coronaria , Paro Cardíaco Inducido/métodos , Daño por Reperfusión Miocárdica/prevención & control , Anciano , Biomarcadores/sangre , Soluciones Cardiopléjicas/efectos adversos , Puente Cardiopulmonar/efectos adversos , Puente de Arteria Coronaria/efectos adversos , Forma MB de la Creatina-Quinasa/sangre , Femenino , Paro Cardíaco Inducido/efectos adversos , Humanos , Unidades de Cuidados Intensivos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/etiología , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Troponina T/sangre
13.
Perfusion ; 35(8): 819-825, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32308124

RESUMEN

BACKGROUND: Rho-associated protein kinase (ROCK) plays a vital role in the pathogenesis of many cardiovascular diseases. Previous studies have demonstrated that ROCK is overactivated and involved in myocardial ischemia/reperfusion in vivo. But the role of ROCK in circulating leukocytes during myocardial ischemia/reperfusion is not well studied. MATERIAL AND METHODS: This study was performed to evaluate ROCK activity in circulating leukocytes in rats with myocardial ischemia/reperfusion injury. Myocardial ischemia/reperfusion Wistar rats were subjected to 30-min ischemia followed by 180-min reperfusion. ROCK activity in circulating leukocytes was examined by the phosphorylation state of myosin phosphatase targeting subunit 1, a substrate of ROCK. RESULTS: ROCK activity significantly increased in leukocytes in rat ischemia/reperfusion models compared to the sham group. ROCK1 not ROCK2 level in circulating leukocytes was significantly elevated in ischemia/reperfusion. Administration of the selective inhibitor of ROCK, fasudil, significantly reduced myocardial infarct size, myocyte apoptosis, and inflammatory cytokine, including interleukin 6 and tumor necrosis factor α. Furthermore, fasudil upregulated ischemia/reperfusion-induced reduction of nitric oxide production. CONCLUSION: Increased ROCK1 not ROCK2 in circulating leukocytes plays a role in the pathogenesis of myocardial ischemia/reperfusion injury. Inhibition of ROCK1 in circulating leukocytes has an important role in fasudil-induced cardioprotective effects. ROCK1 in circulating leukocytes might be a new biomarker in myocardial ischemia/reperfusion injury.


Asunto(s)
Leucocitos/metabolismo , Daño por Reperfusión Miocárdica/patología , Quinasas Asociadas a rho/metabolismo , Animales , Femenino , Leucocitos/enzimología , Daño por Reperfusión Miocárdica/sangre , Ratas , Ratas Wistar , Quinasas Asociadas a rho/sangre
14.
J Cell Biochem ; 120(8): 12912-12923, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30861181

RESUMEN

The goal of this study was to explore the role of tumor associated long noncoding RNA expressed on chromosome 2 (TALNEC2) in protecting against myocardial ischemic injury, as well as its underlying molecular mechanism. We established a cell model of myocardial injury through treating H9c2 cells with hypoxia, and the expression level of TALNEC2 was analyzed. Further, in vitro studies investigated the functional role of TALNEC2 dysregulation in hypoxia injury by assessing cell proliferation, migration, invasion, and apoptosis. Moreover, the expression of miR-21 was determined after dysregulation of TALNEC2, and whether TALNEC2-regulated hypoxia injury in H9c2 cells via regulating miR-21 expression were explored. Furthermore, the regulatory relationship between TALNEC2 and Wnt/ß-catenin pathway was also investigated. TALNEC2 was highly expressed in the serum from patients with myocardial ischemic compared with that in healthy persons. Hypoxia-induced injury in H9c2 cells. Overexpression of TALNEC2 aggravated hypoxia injury in H9c2 cells. TALNEC2 could negative regulate the miR-21 expression, and overexpression of TALNEC2 aggravated hypoxia injury by downregulation of miR-21. Moreover, miR-21 negatively regulated the PDCD4 expression, and PDCD4 was a target of miR-21. Further studies disclosed that the overexpression of TALNEC2 further activated the Wnt/ß-catenin pathway in hypoxia-treated H9c2 cells, implying that the Wnt/ß-catenin pathway was a downstream mechanism mediating the role of TALNEC2 in regulating hypoxia injury in H9c2 cells. These findings confirmed the key functions of TALNEC2 in regulating myocardial ischemic injury. Upregulation of TALNEC2 may aggravate hypoxia injury in H9c2 cells via regulating miR-21/PDCD4-medited activation of the Wnt/ß-catenin pathway. TALNEC2 may serve as a promising therapeutic target in myocardial ischemia.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Regulación de la Expresión Génica , MicroARNs/genética , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , Anciano , Animales , Secuencia de Bases , Hipoxia de la Célula , Línea Celular , Supervivencia Celular/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/genética , Ratas , beta Catenina/metabolismo
15.
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
16.
Crit Care Med ; 47(3): e250-e255, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30608281

RESUMEN

OBJECTIVES: Remote ischemic preconditioning (RIPC) is a practicable and noninvasive method to protect the heart against ischemia reperfusion injury. Unfortunately results from clinical studies are not convincing. Propofol is suggested to be an inhibiting factor of cardioprotection by RIPC, but the underlying mechanism is still unknown. We investigated whether after RIPC the release of humoral factors and/or the direct cardioprotective effect at the myocardium is inhibited by propofol. DESIGN: Randomized, prospective, blinded laboratory investigation. SETTING: Experimental laboratory. PATIENTS/SUBJECTS: Male Wistar rats. INTERVENTIONS: Repetitive hind limb ischemia in rats-blood plasma transfers to isolated rat heart. MEASUREMENTS AND MAIN RESULTS: In male Wistar rats (six groups, each n = 6/group), RIPC was induced by four cycles of 5 minutes bilateral hind limb ischemia alternately with 5 minutes of reperfusion. Blood samples were taken with (RIPC) and without RIPC (Con). Rats received continuous anesthesia with pentobarbital (Pento, 40 mg/kg body weight/hr) or propofol (Prop, 12 mg/kg body weight/hr), respectively. Cardioprotective properties of the blood plasma was investigated in the rat heart in vitro (six groups, each n = 6/group) perfused with Krebs-Henseleit buffer alone or with propofol (10 µM). Plasma was administered over 10 minutes before myocardial ischemia. All hearts underwent 33 minutes of global ischemia followed by 1 hour of reperfusion. At the end of the experiments, infarct size was determined by triphenyl-tetrazolium-chloride staining. RIPC plasma from pentobarbital anesthetized rats (Pento-RIPC) reduced infarct size from 64% (62-71%) (Pento-Con) to 34% (30-39%) (p < 0.0001). Infarct size with control plasma from propofol anesthetized rats was 59% (58-64%) (Prop-Con). RIPC plasma could not induce cardioprotection (Prop-RIPC: 63% [56-70%] ns vs Prop-Con). In contrast, RIPC plasma from pentobarbital anesthetized rats induced a significant infarct size reduction under propofol perfusion (Pento-RIPC: 34% [30-42%] vs Pento-Con: 54% [53-63%]; p < 0.0001). CONCLUSIONS: Loss of cardioprotection by RIPC during propofol anesthesia depends on inhibition of release of humoral factors.


Asunto(s)
Anestésicos Intravenosos/efectos adversos , Precondicionamiento Isquémico , Daño por Reperfusión Miocárdica/prevención & control , Propofol/efectos adversos , Anestesia/efectos adversos , Animales , Hemodinámica , Miembro Posterior/irrigación sanguínea , Precondicionamiento Isquémico/métodos , Masculino , Daño por Reperfusión Miocárdica/sangre , Distribución Aleatoria , Ratas , Ratas Wistar
17.
Cardiovasc Diabetol ; 18(1): 66, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31151453

RESUMEN

BACKGROUND: The mechanisms underlying increased mortality in patients with diabetes and admission hyperglycemia after an acute coronary syndrome may involve reduced capacity for cardioprotection. We investigated the impact of hyperglycemia on exogenously activated cardioprotection by ischemic preconditioning (IPC) in hearts from rats with type 2 diabetes mellitus (T2DM) that were endogenously cardioprotected by an inherent mechanism, and the involvement of myocardial glucose uptake (MGU) and myocardial O-linked ß-N-acetylglucosamine (O-GlcNAc). METHODS AND RESULTS: In isolated, perfused rat hearts subjected to ischemia-reperfusion, infarct size (IS) was overall larger during hyper- ([Glucose] = 22 mmol/L]) than normoglycemia ([Glucose] = 11 mmol/L]) (p < 0.001). IS was smaller in 12-week old Zucker diabetic fatty rats with recent onset T2DM (fa/fa) than in rats without T2DM (fa/+) (n = 8 in each group) both during hyperglycemia (p < 0.05) and normoglycemia (p < 0.05). IPC (2 × 5 min cycles) reduced IS during normo- (p < 0.01 for both groups) but not during hyperglycemia independently of the presence of T2DM. During hyperglycemia, an intensified IPC stimulus (4 × 5 min cycles) reduced IS only in hearts from animals with T2DM (p < 0.05). IPC increased MGU and O-GlcNAc levels during reperfusion in animals with and without T2DM at normoglycemia (MGU: p < 0.05, O-GlcNAc: p < 0.01 for both groups) but not during hyperglycemia. Intensified IPC at hyperglycemia increased MGU (p < 0.05) and O-GlcNAc levels (p < 0.05) only in hearts from animals with T2DM. CONCLUSION: While the effect of IPC is reduced during hyperglycemia in rats without T2DM, endogenous cardioprotection in animals with T2DM is not influenced by hyperglycemia and the capacity for exogenous cardioprotection by IPC is preserved. MGU and O-GlcNAc levels are increased by exogenously induced cardioprotection by IPC but not by endogenous cardioprotection in animals with T2DM reflecting different underlying mechanisms by exogenous and endogenous cardioprotection.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Acetilglucosamina/metabolismo , Animales , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/complicaciones , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Infarto del Miocardio/sangre , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Zucker , beta-N-Acetilhexosaminidasas/metabolismo
18.
Cardiovasc Diabetol ; 18(1): 85, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31262297

RESUMEN

BACKGROUND: Recent large-scale clinical trials have shown that SGLT2-inhibitors reduce cardiovascular events in diabetic patients. However, the regulation and functional role of cardiac sodium-glucose cotransporter (SGLT1 is the dominant isoform) compared with those of other glucose transporters (insulin-dependent GLUT4 is the major isoform) remain incompletely understood. Given that glucose is an important preferential substrate for myocardial energy metabolism under conditions of ischemia-reperfusion injury (IRI), we hypothesized that SGLT1 contributes to cardioprotection during the acute phase of IRI via enhanced glucose transport, particularly in insulin-resistant phenotypes. METHODS AND RESULTS: The hearts from mice fed a high-fat diet (HFD) for 12 weeks or a normal-fat diet (NFD) were perfused with either the non-selective SGLT-inhibitor phlorizin or selective SGLT2-inhibitors (tofogliflozin, ipragliflozin, canagliflozin) during IRI using Langendorff model. After ischemia-reperfusion, HFD impaired left ventricular developed pressure (LVDP) recovery compared with the findings in NFD. Although phlorizin-perfusion impaired LVDP recovery in NFD, a further impaired LVDP recovery and a dramatically increased infarct size were observed in HFD with phlorizin-perfusion. Meanwhile, none of the SGLT2-inhibitors significantly affected cardiac function or myocardial injury after ischemia-reperfusion under either diet condition. The plasma membrane expression of GLUT4 was significantly increased after IRI in NFD but was substantially attenuated in HFD, the latter of which was associated with a significant reduction in myocardial glucose uptake. In contrast, SGLT1 expression at the plasma membrane remained constant during IRI, regardless of the diet condition, whereas SGLT2 was not detected in the hearts of any mice. Of note, phlorizin considerably reduced myocardial glucose uptake after IRI, particularly in HFD. CONCLUSIONS: Cardiac SGLT1 but not SGLT2 plays a compensatory protective role during the acute phase of IRI via enhanced glucose uptake, particularly under insulin-resistant conditions, in which IRI-induced GLUT4 upregulation is compromised.


Asunto(s)
Glucemia/efectos de los fármacos , Resistencia a la Insulina , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Florizina/farmacología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Compuestos de Bencidrilo/farmacología , Glucemia/metabolismo , Canagliflozina/farmacología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Transportador de Glucosa de Tipo 4/metabolismo , Glucósidos/farmacología , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Obesidad/sangre , Obesidad/fisiopatología , Transducción de Señal , Transportador 1 de Sodio-Glucosa/metabolismo , Tiofenos/farmacología
19.
J Cardiovasc Pharmacol ; 73(2): 100-104, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30531437

RESUMEN

Although hypothermia suppresses myocardial ischemia/reperfusion injury, whether it also protects the myocardium against cellular stresses such as chemical anoxia and calcium overload remains unknown. We examined the effect of mild hypothermia (33°C) on myocardial injury during ischemia/reperfusion, local administration of sodium cyanide (chemical anoxia), or local administration of maitotoxin (forced Ca overload) using cardiac microdialysis applied to the feline left ventricle. Baseline myoglobin levels (in ng/mL) were 237 ± 57 and 150 ± 46 under normothermia and hypothermia, respectively (mean ± SE, n = 6 probes each). Coronary artery occlusion increased the myoglobin level to 2600 ± 424 under normothermia, which was suppressed to 1160 ± 149 under hypothermia (P < 0.05). Reperfusion further increased the myoglobin level to 6790 ± 1550 under normothermia, which was also suppressed to 2060 ± 343 under hypothermia (P < 0.05). By contrast, hypothermia did not affect the cyanide-induced myoglobin release (930 ± 130 vs. 912 ± 62, n = 6 probes each) or the maitotoxin-induced myoglobin release (2070 ± 511 vs. 2110 ± 567, n = 6 probes each). In conclusion, mild hypothermia does not make the myocardium resistant to cellular stresses such as chemical anoxia and forced Ca overload.


Asunto(s)
Cardiopatías/prevención & control , Hipotermia Inducida , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Gatos , Hipoxia de la Célula , Modelos Animales de Enfermedad , Cardiopatías/sangre , Cardiopatías/inducido químicamente , Cardiopatías/patología , Toxinas Marinas , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Mioglobina/sangre , Oxocinas , Cianuro de Sodio
20.
J Cardiovasc Pharmacol ; 74(2): 91-97, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31356535

RESUMEN

BACKGROUND: Preventing myocardial ischemia-reperfusion injury in on-pump cardiac surgeries remains an enormous challenge. Sevoflurane postconditioning has been effective at overcoming this challenge by modulating inflammatory mediators and ameliorating antioxidative stress. Dexmedetomidine (DEX) is a commonly used medication for cardiac patients with organ-protective properties that lead to positive outcomes. Whether DEX also has cardiac-protective properties and the associated mechanism in sevoflurane postconditioning-based valve replacement surgeries are unknown. OBJECTIVE: This study was conducted to observe the effect of DEX administration before cardiopulmonary bypass (CPB) on myocardial injury, oxidative stress, and inflammatory response indicators in the peripheral blood. METHODS: Twenty-eight eligible cardiac patients who underwent valve replacement surgery with standard sevoflurane postconditioning were included in the study. The patients were randomly divided into a DEX group and a non-DEX group according to whether DEX (0.5-µg/kg overload dose for 10 minutes and a 0.5-µg/kg/h maintenance dose) or saline was administered from induction to the beginning of CPB. The primary outcome was the cardiac troponin I concentration (cTnI) in the blood 24 hours after CPB. The levels of malondialdehyde (MDA), superoxide dismutase, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) were also measured. RESULTS: The mean cTnI at 24 hours after CPB was clearly decreased in the DEX group compared with that in the non-DEX group (4.16 ± 1.58 vs. 6.90 ± 3.73, P < 0.05). TNF-α levels were lower in the DEX group after CPB (T1-T5), with a significant difference found at 1-6 hours after CPB (1 hour, 19.03 vs. 28.09; 6 hours, 20.74 vs. 30.94, P < 0.05). The IL-6 and IL-8 concentrations in the DEX group were dramatically increased at 6 hours after CPB (P < 0.05). The MDA content and superoxide dismutase activity were comparable between the 2 groups. A lower proportion of anemia cases were noted after CPB in the DEX group than in the non-DEX group (non-DEX, 10% vs. DEX, 5%, P < 0.05). CONCLUSIONS: In valve replacement surgery with sevoflurane postconditioning, pre-CPB administration of DEX can reduce the cTnI level at 24 hours after CPB and brings synergic benefits of the inflammatory response.


Asunto(s)
Antiinflamatorios/administración & dosificación , Puente Cardiopulmonar/efectos adversos , Dexmedetomidina/administración & dosificación , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Inflamación/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Sevoflurano/administración & dosificación , Anciano , Antiinflamatorios/efectos adversos , Biomarcadores/sangre , China , Citocinas/sangre , Dexmedetomidina/efectos adversos , Método Doble Ciego , Esquema de Medicación , Femenino , Humanos , Inflamación/sangre , Inflamación/diagnóstico , Inflamación/etiología , Mediadores de Inflamación/sangre , Masculino , Malondialdehído/sangre , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/etiología , Proyectos Piloto , Estudios Prospectivos , Sevoflurano/efectos adversos , Superóxido Dismutasa/sangre , Factores de Tiempo , Resultado del Tratamiento , Troponina I/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA