Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Ther Med ; 23(3): 223, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35222700

RESUMEN

The present study aimed to explore the effects and clinical importance of serum interleukin (IL) IL-1ß, IL-6, C-reactive protein (CRP), intercellular adhesion molecule (ICAM)-1 and matrix metalloproteinase (MMP)-2 in patients with acute cerebral infarction undergoing intravenous thrombolysis during simultaneous hypothermia therapy. A total of 80 patients with acute cerebral infarction who were treated at our hospital were randomly selected. They were divided into groups A and B. The two groups were treated with intravenous thrombolysis, while group B received sub-hypothermia treatment. Prior to treatment and at 7 days after treatment, 5 ml of venous blood was collected and stored in a freezer at -80˚C. IL-1ß, IL-6, CRP, ICAM-1 and MMP-2 levels were detected by ELISA and compared between the groups and time-points. The results were as follows: i) At 7 days after treatment, the levels of IL-1ß, IL-6, CRP, ICAM-1 and MMP-2 in group B were significantly decreased compared with those in group A (P<0.05), while there was no significant difference of these levels between group A and B before treatment (P>0.05). The incidence of adverse reactions in group A and group B was 35 and 20% respectively, and the mortality rate was 10 and 5%, respectively. There were no significant differences in adverse events and mortality between the two groups (P>0.05). In addition, a positive correlation of the level of IL-1ß, IL-6, CRP, ICAM-1 and MMP-2 with the National Institutes of Health Stroke Scale score was determined in the patients prior to treatment. In conclusion, mild hypothermia treatment in addition to intravenous thrombolysis significantly reduced the levels of IL-1ß, IL-6, CRP, ICAM-1 and MMP-2 in patients with acute cerebral infarction and reduced inflammation, and should therefore be incorporated in clinical practice.

2.
Inflammation ; 43(6): 2109-2118, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32725513

RESUMEN

Panaxatriol saponins (PTS) have a long history in the treatment of stroke. In our previous experiments, PTS has been found to alleviate ischemic stroke and play a role through regulating the inflammatory response, but the specific mechanism of its regulation is still unclear. Cell viability was determined by MTT assay. Expressions of polarization-related proteins CD16, CD68, ARG1 and CD206; inflammatory factors interleukin-1ß (IL-1ß); inducible nitric oxide synthase (iNOS); monocyte chemotactic protein 1(MCP-1) and cyclooxygenase-2 (COX-2); apoptosis-related proteins pro-caspase3; bax; caspase3 and bcl-2; and STAT3 and p-STAT3 were detected by western blot. ELISA was used to detect the expression of inflammatory-related factors in cells. The apoptosis rate was detected by flow cytometry. We found that the survival rate of oxygen sugar deprivation/reoxygenation (OGD/R) cells increased obviously after PTS treatment in a dose-dependent manner. PTS can promote M2 polarization of microglial cells (BV2) and inhibit inflammatory response of OGD/R cells, accompanied by decreased expression of inflammatory factors IL-1ß, iNOS, MCP-1, and COX-2. PTS inhibited apoptosis of OGD/R cells and was accompanied by decreased expression of apoptotic proteins Bax and caspase3 and increased expression of Bcl-2. We also found that PTS activated STAT3 levels in BV2 cells. After the addition of STAT3 inhibitor Stattic, it was found that PTS could promote M2 polarization of BV2 cells by activating the STAT3 pathway, thus inhibiting cell inflammation and apoptosis. PTS promoted M2 polarization in microglia cells by activating the STAT3 pathway, thereby reducing cell inflammation and apoptosis after glucose/oxygen deprivation.


Asunto(s)
Apoptosis/efectos de los fármacos , Ginsenósidos/farmacología , Glucosa/metabolismo , Macrófagos/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Saponinas/farmacología , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Ratones , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal/efectos de los fármacos
3.
Exp Ther Med ; 19(3): 2119-2126, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32104274

RESUMEN

Effect of micro ribonucleic acid (miR)-130a on neuronal apoptosis in rats with cerebral infarction (CI) was studied to explore whether phosphatase and tensin homolog deleted on chromosome ten (PTEN)/phosphatidylinositol 3-hydroxy kinase (PI3K)/protein kinase B (Akt) is involved in the regulation of neuronal apoptosis. Thirty-six Sprague-Dawley (SD) rats were randomly divided into blank control group, model group and miR-130a low-expression group. miR-130a was determined by quantitative polymerase chain reaction (qPCR), the content of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-10 was detected using the enzyme-linked immunosorbent assay (ELISA) kits, and the neuronal apoptosis level in each group was determined through terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. The neurobehavioral score was significantly lower in model group than that in blank control group (P<0.01), while it was significantly higher in miR-130a low-expression group than that in model group (P<0.01). Compared with blank control group, the model group had obviously increased content of TNF-α and IL-6 (P<0.01), decreased content of IL-10 (P<0.01), more apoptotic neurons (P<0.01), higher expression of caspase-3 (P<0.01), and obviously lower Bcl-2/Bax (P<0.01). Moreover, expression of phosphorylated (p)-PTEN, PI3K and p-Akt in brain tissues was remarkably lower in the model group than those in the blank control group (P<0.01). The expression level of miR-130a in brain tissues of CI rats is significantly increased. miR-130a promotes the release of inflammatory factors and facilitates neuronal apoptosis through suppressing the PTEN/PI3K/Akt signaling pathway.

4.
Exp Ther Med ; 19(1): 658-664, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31885705

RESUMEN

Differentially expressed miRNAs in the GEO profile of ischemic stroke were analyzed to clarify the specific role of microRNA-324-5p (miRNA-324-5p) in ischemic stroke and the potential mechanism. After screening out miRNA-324-5p, its level in peripheral blood of stroke patients and in vitro oxygen-glucose deprivation (OGD)-induced primary rat neurons was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Regulatory effects of miRNA-324-5p on viability, and apoptosis of OGD-induced neurons were evaluated by CCK-8 and Annexin V fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining, respectively. Glucose uptake and caspase-3 activity in OGD-induced neurons transfected with miRNA-324-5p mimics or inhibitor were also examined. The binding of miRNA-324-5p to its target gene RAN was analyzed by dual-luciferase reporter gene assay and western blot analysis. By analyzing the data of GSE46266 profile, miRNA-324-5p expression was shown markedly lower in MCAO rats relative to controls. Identically, we also observed the downregulated miRNA-324-5p in peripheral blood of stroke patients and in vitro OGD-induced primary neurons. Overexpression of miRNA-324-5p accelerated viability, induced apoptosis and strengthened glucose uptake ability of OGD-induced neurons. Knockdown of miRNA-324-5p, conversely, obtained the opposite results. Furthermore, we confirmed the binding of miRNA-324-5p to RAN, the target gene that was negatively regulated by miRNA-324-5p. Importantly, RAN overexpression partially reversed the regulatory effect of miRNA-324-5p on viability and glucose uptake of OGD-induced neurons. miRNA-324-5p is downregulated after ischemic stroke, which aggravates the disease condition by inhibiting neuronal proliferation and glucose uptake via upregulating RAN.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...