Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Acta Biochim Biophys Sin (Shanghai) ; 53(4): 430-437, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33686403

RESUMEN

Resveratrol (RES) protects myocardial cells from hypoxia/reoxygenation (H/R)-caused injury. However, the mechanism of this effect has not been clarified. Thus, in this study, we aimed to determine whether RES attenuates H/R-induced cell necroptosis by inhibiting the tumor necrosis factor-alpha (TNF-α)/receptor-interacting protein kinase 1 (RIP1)/RIP3/mixed-lineage kinase domain-like (MLKL) signaling pathway. Rat myocardial ischemia/reperfusion (I/R) models and H/R-injured cell models were constructed. Our study showed that myocardial H/R injury significantly increased the levels of TNF-α, RIP1, RIP3, and p-MLKL/MLKL by western blot analysis. Cell viability assay and 4,6-dianmidino-2-phenylindole (DAPI)-propidium iodide staining showed that the cell viability was decreased, and necroptosis was increased after myocardial H/R injury. The expressions of TNF-α, RIP1, RIP3, and p-MLKL/MLKL in H/R myocardial cells treated with different concentrations of RES were significantly downregulated. In addition, we also found that the cell viability was increased and necroptosis was decreased in dose-dependent manners when H/R-injured cells were treated with RES. In addition, the enhanced effect of TNF-α on necroptosis in myocardial H/R-injured cells was improved by RES, and the effect of RES was confirmed in vivo in I/R rats. This study also showed that RES suppresses necroptosis in H9c2 cells, which may occur through the inhibition of the TNF-α/RIP1/RIP3/MLKL signaling pathway. Our data suggest that necroptosis is a promising therapeutic target and may be a promising therapeutic target for the treatment of myocardial I/R injury.


Asunto(s)
Daño por Reperfusión Miocárdica/metabolismo , Necroptosis/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Resveratrol/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Masculino , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Ratas , Ratas Sprague-Dawley
2.
BMC Med Inform Decis Mak ; 19(1): 193, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615569

RESUMEN

BACKGROUND: Several heart failure (HF) risk models exist, however, most of them perform poorly when applied to real-world situations. This study aimed to develop a convenient and efficient risk model to identify patients with high readmission risk within 90 days of HF. METHODS: A multivariate logistic regression model was used to predict the risk of 90-day readmission. Data were extracted from electronic medical records from January 1, 2017 to December 31, 2017 and follow-up records of patients with HF within 3 months after discharge. Model performance was evaluated using a receiver operating characteristic curve. All statistical analysis was done using R version 3.5.0. RESULTS: A total of 350 patients met the inclusion criterion of being readmitted within in 90 days. All data sets were randomly divided into derivation and validation cohorts at a 7/3 ratio. The baseline data were fairly consistent among the derivation and validation cohorts. The variables most clearly related to readmission were logarithm of serum N-terminal pro b-type natriuretic peptide (NT-proBNP) level, red cell volume distribution width (RDW-CV), and Charlson comorbidity index (CCI). The model had good discriminatory ability (C-statistic = 0.73). CONCLUSIONS: We developed and validated a multivariate logistic regression model to predict the 90-day readmission risk for Chinese patients with HF. The predictors included in the model are derived from electronic medical record (EMR) admission data, making it easier for physicians and pharmacists to identify high-risk patients and tailor more intensive precautionary strategies.


Asunto(s)
Registros Electrónicos de Salud , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/terapia , Readmisión del Paciente , Adulto , Anciano , Femenino , Insuficiencia Cardíaca/diagnóstico , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Péptido Natriurético Encefálico/sangre , Alta del Paciente , Fragmentos de Péptidos/sangre , Curva ROC , Medición de Riesgo
3.
Sci Rep ; 14(1): 15167, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956425

RESUMEN

Selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs) are reported to cause stress cardiomyopathy (SC). This study evaluated the association between SSRI/SNRI use and the occurrence of cardiomyopathy in the publicly available U.S. Food and Drug Administration Adverse Event Reporting System (FAERS) database. Disproportionate analysis and likelihood ratio tests were used to identify risk associated with SSRIs or SNRIs and the incidence of SC, using data from between from 2012 to 2022 acquired from the FAERS database. The study identified 132 individual case safety reports (ICSRs) of SC associated with SSRIs or SNRIs. Venlafaxine (48%) and fluoxetine (27%) were the most common antidepressants of the ICSRs. Approximately 80% of SC cases were reported in females, with individuals aged 45-65 years identified as a high-risk population. Both venlafaxine (ratio-scale information component [RSIC] 2.54, 95% CI 2.06-3.04) and fluoxetine (RSIC 3.20, 95% CI 2.31-4.47) were associated with SC, with likelihood ratio estimates of 3.55 (p = 0.02) for venlafaxine and 4.82 (p = 0.008) for fluoxetine. The median time to cardiomyopathy onset was 20 days, with hospitalization reported in 48.33% of patients. Venlafaxine and fluoxetine were associated with SC risk, particularly in middle-aged women. Caution should be exercised when using SSRIs or SNRIs combined with other serotonergic medications.


Asunto(s)
Farmacovigilancia , Inhibidores Selectivos de la Recaptación de Serotonina , Inhibidores de Captación de Serotonina y Norepinefrina , Cardiomiopatía de Takotsubo , Humanos , Femenino , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Masculino , Persona de Mediana Edad , Anciano , Inhibidores de Captación de Serotonina y Norepinefrina/efectos adversos , Cardiomiopatía de Takotsubo/inducido químicamente , Cardiomiopatía de Takotsubo/epidemiología , Sistemas de Registro de Reacción Adversa a Medicamentos , Adulto , Estados Unidos/epidemiología , Clorhidrato de Venlafaxina/efectos adversos , Fluoxetina/efectos adversos , Bases de Datos Factuales , Factores de Riesgo
4.
PeerJ ; 10: e14111, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36213502

RESUMEN

Background: Clopidogrel is widely used to prevent and treat cardiovascular atherosclerosis and thrombosis. However, disturbance in the expression and activity of liver cytochrome metabolic enzymes significantly changes clopidogrel efficacy. Therefore, the effect of chronic unpredictable mild stress (CUMS)-induced depression on the expression of liver cytochrome metabolic enzymes and clopidogrel pharmacokinetics in rats were explored. Methods: Nine different CUMSs were selected to establish a rat model of depression. Open field experiment and sucrose preference test were applied to explore the depressive behaviors. The concentration of serotonin in the cortex of depressed rats was determined using enzyme linked immunosorbent assay (ELISA). All rats were given 10 mg/kg clopidogrel orally after 12 weeks, and blood samples were collected at different time points. The clopidogrel concentration and CYP2C19/ CYP2C9 activity in rat liver microsomes were assayed by high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). The rat liver drug enzymes expression was determined by Real-Time Quantitative Reverse Transcription PCR (RT-qPCR). Results: Open field experiment and sucrose preference test indicated the successful construction of the CUMS-induced depression model. The concentration of serotonin in the cortex of depressed rats decreased by 42.56% (∗∗ p < 0.01). The area under the curve of clopidogrel pharmacokinetics decreased by 33.13% (∗ p < 0.05) in the depression rats, while distribution volume and clearance increased significantly (∗∗ p < 0.01). The half-time and distribution volume did not significantly differ. The CYP2C19 and CYP2C9 activity of liver microsomes in the CUMS-induced depression group were significantly higher than that in the control group (∗∗ p < 0.01). CYP2C11 and CYP1A2 mRNA expression up-regulated approximately 1.3 - fold in the depressed rat livers compared with that in the control, whereas that of CYP2C13 was down-regulated by 27.43% (∗∗ p < 0.01). CYP3A1 and CYP2C12 expression were slightly up-regulated, and that of CES1 did not change. Conclusions: These results indicated that CUMS-induced depression altered clopidogrel pharmacokinetics, and the change in CYP450 activity and expression in depressed rat livers might contribute to the disturbance of clopidogrel pharmacokinetics.


Asunto(s)
Clopidogrel , Depresión , Estrés Psicológico , Animales , Ratas , Clopidogrel/farmacocinética , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Sistema Enzimático del Citocromo P-450/genética , Depresión/tratamiento farmacológico , Ratas Sprague-Dawley , Serotonina , Sacarosa , Espectrometría de Masas en Tándem
5.
J Mol Histol ; 52(2): 207-217, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33547542

RESUMEN

MicroRNAs (miRNAs) play a key role in various pathological processes like atrial fibrillation (AF). However, the mechanisms remain unclear. Herein, this study was undertaken to probe the roles of ADAM10 and its targeting miR-520d in rapid pacing-induced apoptosis in atrial myocytes. In this study, the atrial myocytes grew adherently with irregular morphology. Immunofluorescence showed that more than 90% of atrial myocytes were α-sarcomeric actin (α-SCA) positive, indicating that the primary cells were positive for α-SCA staining and atrial myocytes were successfully isolated. The pacing atrial myocyte model was established after rapid pacing stimulation and we found the rapid pacing stimulation caused elevated ADAM10 and suppressed miR-520d. CCK-8 assay was applied for evaluation of cell viability, TUNEL staining for assessment of cell apoptosis and dual-luciferase reporter gene assay for verification of the targeting relationship between miR-520d and ADAM10. Overexpression of miR-520d or silencing of ADAM10 could enhance cell viability and reduce cell apoptosis in the rapid pacing-induced atrial myocytes. ADAM10 was a target gene of miR-520d. MiR-520d negatively targeted ADAM10, thereby promoting cell viability and inhibiting apoptosis in rapid pacing atrial myocyte model. In summary, miR-520d enhances atrial myocyte viability and inhibits cell apoptosis in rapid pacing-induced AF mouse model through negative mediation of ADAM10.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Apoptosis/fisiología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Apoptosis/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
Free Radic Res ; 54(1): 76-90, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31909644

RESUMEN

Dexmedetomidine (Dex), a sedative and analgesic agent, is known to have a cardioprotective effect against ischaemia/reperfusion (I/R) injury via regulation of antioxidant and anti-inflammatory signals. In contrast, Se shows a cardioprotective effect against I/R injury, because it is a key component of selenoproteins, most of which are antioxidant enzymes such as GPxs and TrxRs. This study aimed to determine whether the protective effects on myocardial cells against I/R injury were further improved when treatment with Dex and Se in combination. H9C2 cells were treated with Dex and Na2SeO3, alone or in combination, before oxygen glucose deprivation/reoxygenation (OGD/R). OGD/R-induced myocardial cell injury was evaluated using cell viability, apoptosis rate, the release of LDH, and intracellular ROS levels. Both Dex and Na2SeO3 improved cell viability and reduced the apoptosis rate, LDH release, and intracellular ROS. This cytoprotection was higher with Dex and Na2SeO3 cotreatment than their individual treatments. Treatment with Dex increased the SOD1, SOD2, GPx1, and GPx2 expression in H9C2 cells in OGD/R, while Na2SeO3 increased the GPx1-4 and TrxR1-3 mRNA levels. Notably, cotreatment with Dex and Na2SeO3 increased the mRNA expression of all these antioxidant enzymes. Dex treatment attenuated the activation of JNK, p65 (NF-κB), Camk1, and NLRP3 signals. Na2SeO3 enhanced the inhibitory effect of Dex on phosphorylated (p)-p65, p65, and NLRP3 in OGD/R. However, TrxR1 knockdown attenuated the positive effect of Na2SeO3 on Dex-mediated anti-inflammatory effects. In summary, cotreatments with Dex and Na2SeO3 further improved antioxidant and anti-inflammatory protection of myocardial cells from I/R injury compared to their individual treatments.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Dexmedetomidina/uso terapéutico , Hipnóticos y Sedantes/uso terapéutico , Miocardio/metabolismo , Selenio/metabolismo , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Dexmedetomidina/farmacología , Humanos , Hipnóticos y Sedantes/farmacología , Miocardio/citología , Ratas
7.
Biosci Rep ; 39(12)2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31782491

RESUMEN

BACKGROUND: Prostaglandin-E1 (PGE1) is a potent vasodilator with anti-inflammatory and antiplatelet effects. However, the mechanism by which PGE1 contributes to the amelioration of cardiac injury remains unclear. METHODS: The present study was designed to investigate how PGE1 protects against hypoxia/reoxygenation (H/R)-induced injuries by regulating microRNA-21-5p (miR-21-5p) and fas ligand (FASLG). Rat H9C2 cells and isolated primary cardiomyocytes were cultured under hypoxic conditions for 6 h (6H, hypoxia for 6 h), and reoxygenated for periods of 6 (6R, reoxygenation for 6 h), 12, and 24 h, respectively. Cells from the 6H/6R group were treated with various doses of PGE1; after which, their levels of viability and apoptosis were detected. RESULTS: The 6H/6R treatment regimen induced the maximum level of H9C2 cell apoptosis, which was accompanied by the highest levels of Bcl-2-associated X protein (Bax) and cleaved-caspase-3 expression and the lowest level of B-cell lymphoma 2 (Bcl-2) expression. Treatment with PGE1 significantly diminished the cell cytotoxicity and apoptosis induced by the 6H/6R regimen, and also decreased expression of IL-2, IL-6, P-p65, TNF-α, and cleaved-caspase-3. In addition, we proved that PGE1 up-regulated miR-21-5p expression in rat cardiomyocytes exposed to conditions that produce H/R injury. FASLG was a direct target of miR-21-5p, and PGE1 reduced the ability of H/R-injured rat cardiomyocytes to undergo apoptosis by affecting the miR-21-5p/FASLG axis. In addition, we proved that PGE1 could protect primary cardiomyocytes against H/R-induced injuries. CONCLUSIONS: These results indicate that PGE1 exerts cardioprotective effects in H9C2 cells during H/R by regulating the miR-21-5p/FASLG axis.


Asunto(s)
Alprostadil/farmacología , Proteína Ligando Fas/genética , MicroARNs/genética , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Sustancias Protectoras/farmacología , Ratas , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA