3 research outputs found

    The protective mechanism of Ginkgolides and Ginkgo flavonoids on the TNF-α induced apoptosis of rat hippocampal neurons and its mechanisms in vitro

    Get PDF
    Objective: To explore the neuroprotective mechanism of Ginkgolides or Ginkgo flavonoids on the TNF-α induced apoptosis of cultured rat hippocampal neurons. Materials and methods: Primary hippocampal neurons were isolated from rat brains and cultured with (model group) or without (control group) addition of tumor necrosis factor-α (TNF-α, final concentration of 40 ng/ml) to induce apoptosis. TNF-α induced cultures were divided into model group, Ginkgolides pre-treatment group (20 μg/ml) and Ginkgo flavonoids pre-treatment group (12 μg/ml). CCK8 was used to assess cell viability while Hoechst 33258 staining, Flow cytometry and TUNEL kits were all employed to determine apoptotic neurons. Expression levels of Bcl-2, Bax, Caspase-3, Caspase-7, Caspase-8, Caspase-9 and Cytc were estimated by qRT-PCR. Results: Cell viability was significantly improved in Ginkgolides pre-treatment group or Ginkgo flavonoids pre-treatment group compared with that in model group. Apoptotic neurons were significantly less in Ginkgolides pre-treatment group or Ginkgo flavonoids pre-treatment group than those in model group. Transcription levels of caspase-7, caspase-8, caspase-3, caspase-9, Bax and Cytc were down-regulated, while transcription levels of Bcl-2 was up-regulated in Ginkgolides pre-treatment or Ginkgo flavonoids pre-treatment group than those in model group. Conclusions: Ginkgolides and Ginkgo flavonoids might protect against apoptosis of hippocampal neurons through inhibiting death receptor pathway or mitochondrial pathway under TNF-α background

    Sodium Danshensu stabilizes atherosclerotic vulnerable plaques by targeting IKKβ mediated inflammation in macrophages

    No full text
    Background: The primary cause of acute cardiovascular events with high mortality is the rupture of atherosclerotic plaque followed by thrombosis. Sodium Danshensu (SDSS) has shown potential in inhibiting the inflammatory response in macrophages and preventing early plaque formation in atherosclerotic mice. However, the specific targets and detailed mechanism of action of SDSS are still unclear. Objective: This study aims to investigate the efficacy and mechanism of SDSS in inhibiting inflammation in macrophages and stabilizing vulnerable plaques in atherosclerosis (AS). Materials and Methods: The efficacy of SDSS in stabilizing vulnerable plaques was demonstrated using various techniques such as ultrasound, Oil Red O staining, HE staining, Masson staining, immunohistochemistry, and lipid analysis in ApoE-/- mice. Subsequently, IKKβ was identified as a potential target of SDSS through protein microarray, network pharmacology analysis, and molecular docking. Additionally, ELISA, RT-qPCR, Western blotting, and immunofluorescence were employed to measure the levels of inflammatory cytokines, IKKβ, and NF-κB pathway-related targets, thereby confirming the mechanism of SDSS in treating AS both in vivo and in vitro. Finally, the impact of SDSS was observed in the presence of an IKKβ-specific inhibitor. Results: Initially, the administration of SDSS led to a decrease in the formation and area of aortic plaque, while also stabilizing vulnerable plaques in ApoE-/- mice. Furthermore, it was identified that IKKβ serves as the primary binding target of SDSS. Additionally, both in vivo and in vitro experiments demonstrated that SDSS effectively inhibits the NF-κB pathway by targeting IKKβ. Lastly, the combined use of the IKKβ-specific inhibitor IMD-0354 further enhanced the beneficial effects of SDSS. Conclusions: SDSS stabilized vulnerable plaques and suppressed inflammatory responses by inhibiting the NF-κB pathway through its targeting of IKKβ

    Matrix Metalloproteinase 9 Secreted by Hypoxia Cardiac Fibroblasts Triggers Cardiac Stem Cell Migration In Vitro

    No full text
    Cessation of blood supply due to myocardial infarction (MI) leads to complicated pathological alteration in the affected regions. Cardiac stem cells (CSCs) migration plays a major role in promoting recovery of cardiac function and protecting cardiomyocytes in post-MI remodeling. Despite being the most abundant cell type in the mammalian heart, cardiac fibroblasts (CFs) were underestimated in the mechanism of CSCs migration. Our objective in this study is therefore to investigate the migration related factors secreted by hypoxia CFs in vitro and the degree that they contribute to CSCs migration. We found that supernatant from hypoxia induced CFs could accelerate CSCs migration. Four migration-related cytokines were reported upregulated both in mRNA and protein levels. Upon adding antagonists of these cytokines, the number of migration cells significantly declined. When the cocktail antagonists of all above four cytokines were added, the migration cells number reduced to the minimum level. Besides, MMP-9 had an important effect on triggering CSCs migration. As shown in our results, MMP-9 induced CSCs migration and the underlying mechanism might involve TNF-α signaling which induced VEGF and MMP-9 expression
    corecore