Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
BMC Complement Med Ther ; 23(1): 277, 2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37542250

RESUMEN

BACKGROUND AND AIMS: The scientific community is concerned about cardiovascular disease mortality and morbidity, especially myocardial infarction (MI). Schisantherin A (SCA), a dibenzocyclooctadiene lignan monomer found in S. chinensis fruits has cardiovascular advantages such as increasing NO production in isolated rat thoracic aorta and reducing heart damage caused by ischemia-reperfusion (I/R) through decreasing apoptosis. The present study was undertaken to explore the potential effects of SCA on ISO-induced myocardial infarction in rats. METHODS: Rats were randomly allocated to four groups: control; ISO-treated, and two additional groups of ISO + SCA (5 or 10 mg/kg body weight). All SCA-treated groups were administered with SCA for 20 days and all ISO groups were challenged with ISO on days 19 and 20. RESULTS: SCA significantly attenuated ISO-induced rise in heart/body weight ratio, myocardial infarct size, and cardiac functional biomarkers (CK-MB, cTnI and BNP). SCA pre- and co-treatment resulted in a significant reduction in oxidative stress (via MDA, NO and GSH and increased activities of SOD, CAT and GPx) and inflammation (via decreased levels of TNF-α, IL-6 and IL-1ß) markers when compared to the same levels in cardiac tissue of ISO-treated rats. This study also showed that SCA protects ISO-induced oxidative stress and inflammation by activating the PI3K-AKT/Nrf2/ARE pathway and suppressing TLR4/MAPK/NF-κB pathways. Furthermore, SCA treatment protected histopathological alterations observed in only ISO-treated cardiac transverse sections of rats. CONCLUSION: In conclusion, the findings of this study suggest that SCA protects against cardiac injury in the ISO-induced MI model of rats.


Asunto(s)
Infarto del Miocardio , FN-kappa B , Ratas , Animales , Isoproterenol/efectos adversos , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Toll-Like 4/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Estrés Oxidativo , Inflamación/tratamiento farmacológico , Peso Corporal
2.
J Thorac Dis ; 14(3): 607-613, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35399236

RESUMEN

Background: Coronary angiography (CAG) is "gold standard" for the diagnosis of coronary heart disease (CHD). This study aimed to explore the diagnostic value of cardiopulmonary exercise testing (CPET) and the oxygen uptake kinetics indexes of CPET. Methods: One hundred thirty-one patients with chest pain who underwent coronary angiography in the Department of Cardiology of our hospital from April to September 2021 were selected. According to the results of angiography, the patients were divided into an observation group (patients with coronary heart disease, n=80) and a control group (patients without coronary heart disease, n=75). Both groups underwent CPET before angiography. The differences of peak oxygen uptake, anaerobic threshold, peak kilogram oxygen uptake, peak oxygen pulse, maximum exercise load, maximum metabolic equivalent, and exercise time between the two groups were compared. Also, the correlation between the above indexes and the degree of coronary artery stenosis was analyzed, and the clinical value of the CPET in the diagnosis of CHD was evaluated. Results: The peak oxygen uptake, anaerobic threshold, peak kilogram oxygen uptake, peak oxygen pulse, maximum exercise load, maximum metabolic equivalent, and exercise time in the observation group were lower than those in the control group (P<0.01), and were negatively correlated with the Gensini score (P<0.01). The area under the receiver operating characteristic (ROC) curve of the above seven indexes in the combined diagnosis of CHD was 0.974, the sensitivity was 86.40%, and the specificity was 98.50%, which was better than the clinical value of any of the above indexes alone. Conclusions: CPET is an effective non-invasive examination in the diagnosis of CHD, and has a certain clinical value in the evaluation of the severity of coronary artery stenosis.

3.
Kaohsiung J Med Sci ; 38(2): 149-156, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34741409

RESUMEN

Previous studies have demonstrated that the levels of asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide (NO) synthesis, are strongly associated with hypertension, diabetes, and cardiovascular diseases. Profilin-1, an actin-binding protein, has been documented to be involved in endothelial injury and in the proliferation of vascular smooth muscle cells resulting from hypertension. However, the role of profilin-1 in ADMA-induced vascular injury in hypertension remains largely unknown. Forty healthy subjects and forty-two matched patients with essential hypertension were enrolled, and the related indexes of vascular injury in plasma were detected. Rat aortic smooth muscle cells (RASMCs) were treated with different concentrations of ADMA for different periods of time and transfected with profilin-1 small hairpin RNA to interrupt the expression of profilin-1. To determine the role of the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway, RASMCs were pretreated with AG490 or rapamycin. The expression of profilin-1 was tested using real-time polymerase chain reaction (PCR) and western blot analysis. Cell proliferation was measured by flow cytometry and 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazoliumbromide assays. Compared with healthy subjects, the levels of ADMA and profilin-1 were markedly elevated in hypertensive individuals, while the levels of NO were significantly decreased (p < 0.05). In vitro, studies showed ADMA-induced profilin-1 expression in a concentration- and time-dependent manner in RASMCs (p < 0.05), concomitantly with promoting the proliferation of RASMCs. Furthermore, ADMA-mediated proliferation of RASMCs and upregulation expression of profilin-1 were inhibited by blockade of the JAK2/STAT3 pathway or knockdown of profilin-1. Profilin-1 implicated in the ADMA-mediated vascular lesions in hypertension.


Asunto(s)
Arginina/análogos & derivados , Endotelio Vascular/efectos de los fármacos , Hipertensión/etiología , Miocitos del Músculo Liso/efectos de los fármacos , Profilinas/fisiología , Animales , Arginina/farmacología , Arginina/fisiología , Proliferación Celular , Endotelio Vascular/patología , Humanos , Miocitos del Músculo Liso/patología , Ratas
4.
Comput Struct Biotechnol J ; 19: 1567-1578, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868594

RESUMEN

Heart failure with preserved ejection fraction (HFpEF) is associated with multiple etiologic and pathophysiologic factors. HFpEF leads to significant cardiovascular morbidity and mortality. There are various reasons that fail to identify effective therapeutic interventions for HFpEF, primarily due to its clinical heterogeneity causing significant difficulties in determining physiologic and prognostic implications for this syndrome. Thus, identifying clinical subtypes using multi-omics data has great implications for efficient treatment and prognosis of HFpEF patients. Here we proposed to integrate mRNA, DNA methylation and microRNA (miRNA) expression data of HFpEF with a similarity network fusion (SNF) method following a network enhancement (ne-SNF) denoising technique to form a fused network. A spectral clustering method was then used to obtain clusters of patient subtypes. Experiments on HFpEF datasets demonstrated that ne-SNF significantly outperforms single data subtype analysis and other integrated methods. The identified subgroups were shown to have statistically significant differences in survival. Two HFpEF subtypes were defined: a high-risk group (16.8%) and a low-risk group (83.2%). The 5-year mortality rates were 63.3% and 33.0% for the high- and low-risk group, respectively. After adjusting for the effects of clinical covariates, HFpEF patients in the high-risk group were 2.43 times more likely to die than the low-risk group. A total of 157 differentially expressed (DE) mRNAs, 2199 abnormal methylations and 121 DE miRNAs were identified between two subtypes. They were also enriched in many HFpEF-related biological processes or pathways. The ne-SNF method provides a novel pipeline for subtype identification in integrated analysis of multi-omics data.

5.
Endocrine ; 48(2): 472-82, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24888764

RESUMEN

Retinol-binding protein 4 (RBP4) is a newly discovered adipocytokine related to insulin resistance (IR). Hyperinsulinemia and IR are the major risk factors for cardiovascular diseases (CVD). The role of RBP4 in CVD has not yet been determined. The present study was designed to analyze the correlation of RBP4 and CVD risk factors and to evaluate the role of RBP4 in proliferation of vascular smooth muscle cells during hyperinsulinemia and the underlying mechanisms. Plasma RBP4 concentration, IR-related indexes, and cardiovascular risk factors were measured from blood samples of hyperinsulinemic rats (HIns) and control SD rats (Cons). The vascular morphology and the expression of ERK1/2, p-ERK1/2 in arterial tissues of rats were assessed. Different concentrations of RBP4 (1, 4 µg/ml) were used as intervention factor during insulin-induced aortic smooth muscle cells (RASMCs) proliferation. The expression of cell growth signaling pathways was assessed to identify the active pathway during this proliferation. Specifically, ERK1/2 inhibitor PD98059 and JAK2 inhibitor AG490 were used to detect it. RBP4 expression was higher in HIns compared with Cons (p < 0.01). Plasma RBP4 concentrations were positively correlated with TG (r = 0.490), hsCRP (r = 0.565), media thickness (r = 0.890), and p-ERK1/2 protein (r = 0.746) (p < 0.05 each). In cultured RASMCs, RBP4 enhanced insulin-induced proliferation of cells and expression of p-ERK1/2 and p-JAK2. Blockade of ERK1/2 signaling pathway inhibited RBP4-induced proliferation of RASMCs, while suppressing JAK2 remains unchanged. These results suggest that plasma RBP4 concentrations were associated with CVD. In addition, RBP4 increases the proliferation of VSMCs induced by hyperinsulinism via activation of MAPK signaling pathway.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Hiperinsulinismo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Músculo Liso Vascular/metabolismo , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Animales , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Resistencia a la Insulina , Janus Quinasa 2/metabolismo , Masculino , Miocitos del Músculo Liso/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Riesgo
6.
Mol Med Rep ; 9(5): 1634-40, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24604418

RESUMEN

Recent studies have suggested that retinol binding protein 4 (RBP4), an adipocytokine related to insulin resistance (IR), may play an important role in the development of atherosclerosis and cardiovascular diseases (CVD). Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is one of the most common causes of atherosclerosis. Hyperinsulinism promotes proliferation of VSMCs through the MAPK pathway. However, whether RBP4 is involved in insulin-induced proliferation of VSMCs leading to atherosclerosis remains unclear. In the present study, we evaluated the role of RBP4 and the potential relevance of signaling pathways in this process. Different concentrations of RBP4 (1 and 4 µg/ml) were added to rat aortic smooth muscle cells (RASMCs) during insulin-induced proliferation. The levels of cell growth signaling pathway proteins ERK1/2, p-ERK1/2, JAK2, p-JAK2, STAT3 and p-STAT3 were assessed by western blotting in order to identify the pathway(s) that are activated during insulin-induced proliferation. The specific inhibitors of ERK1/2 (PD98059) and JAK2 (AG490) were used to confirm our findings. Insulin induced proliferation of RASMCs in a concentration- and time-dependent manner, and increased the expression of ERK1/2, p-ERK1/2, JAK2, p-JAK2, STAT3 and p-STAT3 in a time-dependent manner. RBP4 enhanced insulin-induced proliferation of RASMCs and expression of p-ERK1/2 and p-JAK2. RBP4­induced proliferation of RASMCs was reduced by the ERK1/2 inhibitor, while it was unaffected by the JAK2 inhibitor. These results suggest that RBP4 mediates VSMC proliferation induced by insulin via activation of the MAPK pathway, and highlight RBP4 as a modulator of atherosclerosis in hyperinsulinemia, therby enhancing our understanding on a number of unexpected aspects of CVD.


Asunto(s)
Aorta/metabolismo , Hiperinsulinismo/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Animales , Aorta/citología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Insulina/metabolismo , Insulina/farmacología , Janus Quinasa 2/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Proteínas Plasmáticas de Unión al Retinol/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Vascul Pharmacol ; 55(1-3): 34-41, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21586339

RESUMEN

Profilin-1, a regulator of actin polymerization, has recently been linked to vascular hypertrophy and remodeling. Whether profilin-1 is involved in angiotensin (Ang) II-induced proliferation of vascular smooth muscle cells leading to vascular remodeling in hypertension remains unclear. The present study was designed to analyze the correlation of profilin-1 and vascular remodeling during hypertension and to evaluate the role of profilin-1 in proliferation of vascular smooth muscle cells and the underlying mechanisms. The vascular morphology and the expression of profilin-1 in arterial tissues of spontaneously hypertensive rats and Wistar-Kyoto rats were assessed. The profilin-1 expression was significantly increased concomitantly with definite vascular remodeling by evaluating the media thickness, lumen diameter, media thickness-to-lumen diameter ratio and mean nuclear area in artery media in spontaneously hypertensive rats, which was inhibited by treatment with losartan. In cultured rat aortic smooth muscle cells (RASMCs), Ang II induced profilin-1 expression in a dose- and time-dependent manner. Knockdown of profilin-1 using small hairpin RNA inhibited Ang II-induced proliferation of RASMCs. Moreover, blockade of JAK2/STAT3 signaling pathway also inhibited Ang II-induced proliferation of RASMCs and profilin-1 expression. These results suggest that profilin-1 mediates the proliferation of RASMCs induced by Ang II via activation of Ang II type 1 receptor/JAK2/STAT3 signaling pathway, which may contribute to vascular remodeling in hypertension.


Asunto(s)
Angiotensina II/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Profilinas/metabolismo , Angiotensina II/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Presión Sanguínea/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Técnicas de Silenciamiento del Gen/métodos , Hipertensión/metabolismo , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Losartán/farmacología , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Profilinas/antagonistas & inhibidores , Profilinas/genética , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA