OGD

OGD
  • 文章类型: Journal Article
    血管性痴呆(VaD)是老年人痴呆的最常见原因。由于缺乏有效的治疗方案,迫切需要找到一种有效的药物化合物来对抗VaD。据报道,吡拉西坦可以改善人类和动物模型中各种疾病中受损的认知功能。然而,吡拉西坦在VaD中的作用和机制尚不清楚。因此,本研究旨在阐明吡拉西坦对体外VaD细胞模型的影响。我们发现吡拉西坦增强了OGD刺激的SH-SY5Y细胞的生长。此外,吡拉西坦抑制OGD刺激的SH-SY5Y细胞的氧化应激。Further,吡拉西坦改善OGD刺激的SH-SY5Y细胞的线粒体功能。机械上,吡拉西坦抑制OGD刺激的SH-SY5Y细胞中的PI3K/Akt/mTOR途径。总的来说,吡拉西坦通过PI3K/Akt/mTOR轴改善OGD刺激的SH-SY5Y细胞的氧化应激和线粒体功能障碍。因此,吡拉西坦有可能成为VaD的有前途的药物。
    Vascular dementia (VaD) is the most common cause of dementia in older adults. Due to the lack of effective treatment options, there is an urgent need to find an effective pharmaceutical compound to combat VaD. Piracetam has been reported to improve impaired cognitive function in a variety of conditions in both human and animal models. However, the role and mechanism of Piracetam in VaD remain unclear. Therefore this study aimed to elucidate the effect of Piracetam on a cellular model of VaD in vitro. We found that Piracetam enhanced the growth of OGD-stimulated SH-SY5Y cells. In addition, Piracetam inhibited the oxidative stress of OGD-stimulated SH-SY5Y cells. Further, Piracetam improved mitochondrial function of OGD-stimulated SH-SY5Y cells. Mechanistically, Piracetam inhibited the PI3K/Akt/mTOR pathway in OGD-stimulated SH-SY5Y cells. Collectively, Piracetam improved oxidative stress and mitochondrial dysfunction of OGD-stimulated SH-SY5Y cells through PI3K/Akt/mTOR axis. Hence, Piracetam has the potential to serve as a promising drug of VaD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    脑微血管内皮细胞(BMEC)功能丧失是缺血性卒中发展的关键事件,可能受到环状RNA(circRNAs)失调的影响。我们的目的是揭示circRNAFKBP脯氨酸异构酶3(circFKBP3)在缺血性卒中细胞模型中的作用。
    在氧葡萄糖剥夺(OGD)治疗的人BEMC(HBMECs)中构建缺血性中风的细胞模型。进行定量实时PCR(qPCR)和蛋白质印迹以分析circFKBP3,miR-766-3p和TNF受体相关因子3(TRAF3)的表达。CCK-8Transwell,伤口愈合,流式细胞术,实施试管形成和ELISA测定以监测细胞活力,迁移,凋亡,血管生成和炎症产生。通过双荧光素酶验证了miR-766-3p与circFKBP3或TRAF3之间的假定结合关系,RIP和下拉测定。
    CircFKBP3表达在OGD处理的HBMECs中升高。OGD抑制HBMEC活力,迁移,血管生成,并引起细胞凋亡和炎症的产生,而circFKBP3的敲除减弱了这些作用。CircFKBP3与miR-766-3p相互作用,和circFKBP3缺失抑制的HBMEC功能丧失和炎症通过miR-766-3p抑制恢复。CircFKBP3靶向miR-766-3p调控TRAF3表达。通过TRAF3过表达恢复MiR-766-3p富集抑制的HBMEC功能丧失和炎症。
    CircFKBP3缺失通过控制miR-766-3p/TRAF3轴减轻了OGD诱导的HBMECs功能丧失和炎症反应。
    UNASSIGNED: Brain microvascular endothelial cell (BMEC) functions loss is a key event in the development of ischemic stroke, which may be affected by the dysregulation of circular RNAs (circRNAs). We aimed to unveil the role of circRNA FKBP Prolyl Isomerase 3 (circFKBP3) in cell models of ischemic stroke.
    UNASSIGNED: Cell models of ischemic stroke were constructed in human BEMCs (HBMECs) with the treatment of oxygen glucose deprivation (OGD). Quantitative real-time PCR (qPCR) and western blotting were conducted for expression analysis of circFKBP3, miR-766-3p and TNF receptor associated factor 3 (TRAF3). CCK-8, transwell, wound healing, flow cytometry, tube formation and ELISA assays were implemented to monitor cell viability, migration, apoptosis, angiogenesis and inflammation production. The putative binding relationship between miR-766-3p and circFKBP3 or TRAF3 was validated by dual-luciferase, RIP and pull-down assays.
    UNASSIGNED: CircFKBP3 expression was elevated in OGD-treated HBMECs. OGD suppressed HBMEC viability, migration, angiogenesis, and provoked cell apoptosis and inflammation production, while knockdown of circFKBP3 attenuated these effects. CircFKBP3 interacted with miR-766-3p, and circFKBP3 absence-repressed HBMEC function loss and inflammation were recovered by miR-766-3p inhibition. CircFKBP3 targeted miR-766-3p to regulate TRAF3 expression. MiR-766-3p enrichment-repressed HBMEC function loss and inflammation were recovered by TRAF3 overexpression.
    UNASSIGNED: CircFKBP3 absence alleviated OGD-induced function loss and inflammatory responses of HBMECs via governing the miR-766-3p/TRAF3 axis.
    CircFKBP3 expression is elevated in OGD-treated HBMECs.OGD-induced HBMEC function loss and inflammation are alleviated by circFKBP3 absence.CircFKBP3 directly targets miR-766-3p to regulate TRAF3 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    我们使用人脑免疫微环境的体外模型来模拟缺氧缺血性脑损伤(HIBI),并使用人脐带间充质干细胞(hUMSCs)进行治疗,以解决临床前研究中动物和人之间基因差异的转化障碍。共同文化系统,被称为hNAME,由人海马神经元(N)组成,星形胶质细胞(A),小胶质细胞(M),和脑微血管内皮细胞(E)。流式细胞术检测神经元和内皮细胞的凋亡率。hNAME-神经元和内皮细胞经历了比单层细胞更严重的损伤,特别是在复氧48小时和24小时后(OGD48/R24)。蛋白质印迹确定了神经炎症反应标志物,包括HIF-1α,C1q,C3,TNF-α,和iNOS。炎症因子起源于神经胶质小室而不是神经元和血管内皮小室。随着OGD和复氧时间的增加,观察到炎症因子的释放逐渐增加,峰值在OGD48/R24。在人脐带间充质干细胞(hUMSCs)中证实了hNAME值。用hUMSC处理导致hNAME中神经元和内皮细胞损伤的严重程度显著降低。由于神经元之间的相互作用,hNAME是模拟人脑免疫微环境的理想体外模型,船只,星形胶质细胞,和小胶质细胞.
    We used an in vitro model of the human brain immune microenvironment to simulate hypoxic-ischemic brain injury (HIBI) and treatment with human umbilical cord mesenchymal stem cells (hUMSCs) to address the transformation barriers of gene differences between animals and humans in preclinical research. A co-culture system, termed hNAME, consisted of human hippocampal neurons (N), astrocytes (A), microglia (M), and brain microvascular endothelial cells (E). Flow cytometry measured the apoptosis rates of neurons and endothelial cells. hNAME-neurons and endothelial cells experienced more severe damage than monolayer cells, particularly after 48 h and 24 h of reoxygenation (OGD48/R24). Western blotting identified neuroinflammatory response markers, including HIF-1α, C1q, C3, TNF-α, and iNOS. Inflammatory factors originated from the glial chamber rather than the neurons and vascular endothelial chambers. A gradual increase in the release of inflammatory factors was observed as the OGD and reoxygenation times increased, peaking at OGD48/R24. The hNAME value was confirmed in human umbilical cord mesenchymal stem cells (hUMSCs). Treatment with hUMSCs resulted in a notable decrease in the severity of neuronal and endothelial cell damage in hNAME. The hNAME is an ideal in vitro model for simulating the immune microenvironment of the human brain because of the interactions between neurons, vessels, astrocytes, and microglia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    兴奋毒性脑损伤后的神经元水肿导致神经元损伤和死亡。旨在减轻水肿的渗透和手术干预产生较差的临床结果。强调需要探索其他机制。同时伴有神经元肿胀,过量的Ca2+负载可能是有害的,但仍未得到充分的研究,尤其是在新生儿期。我们使用离体多光子Ca2成像来评估在不同和短暂的兴奋性毒性损伤后,新生儿GCaMP6表达神经元的细胞毒性水肿与Ca2负荷之间的关系。我们报告了各种短的兴奋性毒性损伤后,胞质GCaMP6急性易位到新生儿神经元的细胞核中,这些损伤被量化为核:胞质强度比(N/C比)。N/C比的增加与神经元肿胀无关。透射电子显微镜显示,N/C比升高与神经元核孔径增加有关。在体内和离体抑制钙蛋白酶可防止N/C比率增加并减少神经元肿胀。我们的结果表明,在早期大脑发育过程中,短暂的兴奋毒性损伤可以通过钙蛋白酶介导的机制扩大神经元的核孔并失调核运输。此外,N/C比测量可用于实时检测急性神经元损伤。
    Neuronal swelling after excitotoxic insults is implicated in neuronal injury and death in the developing brain, yet mitigating brain edema with osmotic and surgical interventions yields poor clinical outcomes. Importantly, neuronal swelling and its downstream consequences during early brain development remain poorly investigated. Using multiphoton Ca2+ imaging in vivo (P12-17) and in acute brain slices (P8-12), we explored Ca2+-dependent downstream effects after neuronal cytotoxic edema. We observed the translocation of cytosolic GCaMP6s into the nucleus of a subpopulation of neurons minutes after various excitotoxic insults. We used automated morphology-detection algorithms for neuronal segmentation and quantified the nuclear translocation of GCaMP6s as the ratio of nuclear and cytosolic intensity (N/C ratio). Elevated neuronal N/C ratios were correlated to higher Ca2+ loads and could occur independently of neuronal swelling. Electron microscopy revealed that the nuclear translocation was associated with increased nuclear pore size. Inhibiting calpains prevented elevated N/C ratios and neuronal swelling. Thus, our results indicate altered nuclear transport in a subpopulation of neurons shortly after injury in the developing brain, which can be used as an early biomarker of acute neuronal injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    miRNAs在心肌纤维化的发病机制中起着重要作用。本研究旨在确定miR-212-5p在氧糖剥夺(OGD)诱导的人心脏成纤维细胞(HCFs)激活中的新通路。首先,我们发现KLF4蛋白在OGD诱导的HCFs中显著降低。然后,生物信息学分析和验证实验用于鉴定KLF4与miR-212-5p的相互作用的存在。功能实验表明OGD显著上调HCFs中缺氧诱导因子-1α(HIF-1α)的表达,通过与其启动子结合来正向调节miR-212-5p转录。MiR-212-5p通过与KLF4mRNA的3'非翻译编码区(UTR)结合来抑制Krüppel样因子4(KLF4)蛋白的表达。miR-212-5p的抑制通过上调KLF4的表达有效抑制OGD诱导的HCFs的激活,并在体内和体外抑制心脏纤维化。
    It is widely accepted that miRNAs play an important role in the pathogenesis of myocardial fibrosis. This study aimed to identify a new pathway of miR-212-5p in the activation of human cardiac fibroblasts (HCFs) induced by oxygen-glucose deprivation (OGD). First, we found that KLF4 protein was markedly decreased in OGD-induced HCFs. Then, bioinformatics analysis and verification experiments were used to identify the existence of an interaction of KLF4 with miR-212-5p. Functional experiments indicated that OGD significantly upregulated the expression of hypoxia inducible factor-1 alpha (HIF-1α) in HCFs, which positively regulated miR-212-5p transcription by binding to its promoter. MiR-212-5p inhibited the expression of Krüppel-like factor 4 (KLF4) protein by binding to the 3\' untranslated coding regions (UTRs) of KLF4 mRNA. Inhibition of miR-212-5p effectively inhibited the activation of OGD-induced HCFs by upregulating KLF4 expression and inhibited cardiac fibrosis in vivo and in vitro.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    再灌注是缺血性中风的基本治疗方法;然而,许多缺血性卒中患者不能接受再灌注治疗.此外,再灌注可引起缺血再灌注损伤。这项研究旨在确定大鼠嗜铬细胞瘤(PC12)细胞和皮质神经元在体外缺血性中风模型-氧和葡萄糖剥夺(OGD)(0.3%O2)中的再灌注作用。在PC12单元格中,OGD导致细胞毒性和细胞凋亡的时间依赖性增加,从2小时开始,MTT活性降低。在OGD恢复的凋亡PC12细胞的较短时间(4和6小时)后再灌注,而在12小时后,OGD增加LDH释放。在初级神经元中,6hOGD导致细胞毒性显著增加,MTT活性和树突状MAP2染色降低。6小时OGD后的再灌注增加了细胞毒性。HIF-1a在PC12细胞中通过4和6小时OGD稳定,在原代神经元中通过2小时OGD稳定。一组缺氧基因通过OGD处理根据持续时间上调。总之,OGD的持续时间决定了线粒体的活性,细胞活力,HIF-1a稳定,以及两种细胞类型中的缺氧基因表达。短期OGD后的再灌注具有神经保护作用,而长持续时间的OGD是细胞毒性的。
    Reperfusion is the fundamental treatment for ischaemic stroke; however, many ischaemic stroke patients cannot undergo reperfusion treatment. Furthermore, reperfusion can cause ischaemic reperfusion injuries. This study aimed to determine the effects of reperfusion in an in vitro ischaemic stroke model-oxygen and glucose deprivation (OGD) (0.3% O2)-with rat pheochromocytoma (PC12) cells and cortical neurons. In PC12 cells, OGD resulted in a time-dependent increase in cytotoxicity and apoptosis, and reduction in MTT activity from 2 h onwards. Reperfusion following shorter periods (4 and 6 h) of OGD recovered apoptotic PC12 cells, whereas after 12 h, OGD increased LDH release. In primary neurons, 6 h OGD led to significant increase in cytotoxicity, reduction in MTT activity and dendritic MAP2 staining. Reperfusion following 6 h OGD increased the cytotoxicity. HIF-1a was stabilised by 4 and 6 h OGD in PC12 cells and 2 h OGD onwards in primary neurons. A panel of hypoxic genes were upregulated by the OGD treatments depending on the duration. In conclusion, the duration of OGD determines the mitochondrial activity, cell viability, HIF-1a stabilization, and hypoxic gene expression in both cell types. Reperfusion following OGD of short duration is neuroprotective, whereas OGD of long duration is cytotoxic.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    先前的研究表明,在细胞周期调节中起生理作用的细胞周期蛋白依赖性激酶(Cdks)在缺血性中风后的有丝分裂后神经元中被激活,导致凋亡性神经元死亡。在这篇文章中,我们报告了我们的结果,使用广泛使用的氧-葡萄糖剥夺(OGD)在原发性小鼠皮层神经元的缺血性中风的体外模型,以研究Cdk7是否作为激活细胞周期的Cdk激活激酶(CAK)复合物的一部分Cdks,可能是缺血性神经元死亡的调节剂,并可能构成神经保护的治疗靶标。我们发现没有证据表明Cdk7的药理或遗传无效具有神经保护作用。尽管有公认的观点认为细胞凋亡有助于缺血半暗带的细胞死亡,我们在OGD模型中也没有发现细胞凋亡的证据.这可以解释在该模型中Cdk7失效后缺乏神经保护。暴露于OGD的神经元似乎倾向于以NMDA受体依赖性方式死亡,无法在下游进一步预防。鉴于神经元直接暴露于缺氧或严重缺氧,OGD与缺血半暗带建模的相关性值得怀疑。由于OGD后细胞死亡的不确定性,当使用这种体外模型来识别新的中风疗法时,需要谨慎。
    Previous research has shown that cyclin-dependent kinases (Cdks) that play physiological roles in cell cycle regulation become activated in post-mitotic neurons after ischemic stroke, resulting in apoptotic neuronal death. In this article, we report our results using the widely used oxygen-glucose deprivation (OGD) in vitro model of ischemic stroke on primary mouse cortical neurons to investigate whether Cdk7, as part of the Cdk-activating kinase (CAK) complex that activates cell cycle Cdks, might be a regulator of ischemic neuronal death and may potentially constitute a therapeutic target for neuroprotection. We found no evidence of neuroprotection with either pharmacological or genetic invalidation of Cdk7. Despite the well-established idea that apoptosis contributes to cell death in the ischemic penumbra, we also found no evidence of apoptosis in the OGD model. This could explain the absence of neuroprotection following Cdk7 invalidation in this model. Neurons exposed to OGD seem predisposed to die in an NMDA receptor-dependent manner that could not be prevented further downstream. Given the direct exposure of neurons to anoxia or severe hypoxia, it is questionable how relevant OGD is for modeling the ischemic penumbra. Due to remaining uncertainties about cell death after OGD, caution is warranted when using this in vitro model to identify new stroke therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脑缺血再灌注损伤(IRI),在此期间,神经元经历氧糖剥夺/复氧(OGD/R),是许多神经系统疾病中值得注意的病理过程。N1-甲基腺苷(m1A)是一种RNA修饰,可以影响基因表达和RNA稳定性。神经元中m1A修饰的m1A景观和潜在功能仍然知之甚少。我们探索了RNA(mRNA,lncRNA,和circRNA)正常和OGD/R处理的小鼠神经元中的m1A修饰以及m1A对多种RNA的影响。我们研究了原代神经元的m1A景观,鉴定了m1A修饰的RNA,发现OGD/R增加了m1ARNA的数量。m1A修饰也可能影响非编码RNA的调控机制,例如,lncRNA-RNA结合蛋白(RBPs)相互作用和circRNA翻译。我们表明m1A修饰介导circRNA/lncRNA-miRNA-mRNA竞争内源性RNA(ceRNA)机制,mRNA的3'非翻译区(3'UTR)修饰可以阻碍miRNA-mRNA结合。确定了三种修饰模式,并且具有不同模式的基因具有潜在的m1A调节特异性的内在机制。对正常和OGD/R神经元中m1A景观的系统分析为理解RNA修饰奠定了关键基础,并为治疗和开发OGD/R病理相关疾病的药物提供了新的观点和理论基础。
    Cerebral ischaemia‒reperfusion injury (IRI), during which neurons undergo oxygen-glucose deprivation/reoxygenation (OGD/R), is a notable pathological process in many neurological diseases. N1-methyladenosine (m1A) is an RNA modification that can affect gene expression and RNA stability. The m1A landscape and potential functions of m1A modification in neurons remain poorly understood. We explored RNA (mRNA, lncRNA, and circRNA) m1A modification in normal and OGD/R-treated mouse neurons and the effect of m1A on diverse RNAs. We investigated the m1A landscape in primary neurons, identified m1A-modified RNAs, and found that OGD/R increased the number of m1A RNAs. m1A modification might also affect the regulatory mechanisms of noncoding RNAs, e.g., lncRNA-RNA binding proteins (RBPs) interactions and circRNA translation. We showed that m1A modification mediates the circRNA/lncRNA‒miRNA-mRNA competing endogenous RNA (ceRNA) mechanism and that 3\' untranslated region (3\'UTR) modification of mRNAs can hinder miRNA-mRNA binding. Three modification patterns were identified, and genes with different patterns had intrinsic mechanisms with potential m1A-regulatory specificity. Systematic analysis of the m1A landscape in normal and OGD/R neurons lays a critical foundation for understanding RNA modification and provides new perspectives and a theoretical basis for treating and developing drugs for OGD/R pathology-related diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:已发现环状RNA(circularRNA)在许多疾病的进展中起重要作用,包括缺血性中风.然而,circSEC11A在缺血性卒中进展中的调控机制有待进一步研究.
    方法:通过氧糖剥夺(OGD)刺激人脑微血管内皮细胞(HBMECs)。CircSEC11A,通过定量实时PCR(qRT-PCR)定量SEC11AmRNA和miR(微小RNA)-29a-3p。SEMA3A,通过蛋白质印迹定量BAX和BCL2蛋白水平。氧化应激,细胞增殖,血管生成和细胞凋亡能力通过氧化应激检测试剂盒,5-乙炔基-2'-脱氧尿苷(EdU)染色,管形成测定和流式细胞术测定,分别。miR-29a-3p与circSEC11A或SEMA3A之间的直接关系通过双荧光素酶报告基因试验得到验证,RIP测定和RNA下拉测定。
    结果:CircSEC11A在OGD诱导的HBMECs中上调。OGD促进氧化应激和凋亡,抑制细胞增殖和血管生成,而circSEC11A敲除减轻了影响。CircSEC11A充当miR-29a-3p的海绵,和miR-29a-3p抑制剂逆转了si-circSEC11A对OGD诱导的HBMECs氧化损伤的影响。此外,SEMA3A作为miR-29a-3p的靶基因。MiR-29a-3p抑制改善OGD诱导的HBMECs氧化损伤,而SEMA3A过表达拯救了miR-29a-3p模拟物的影响。
    结论:CircSEC11A通过介导miR-29a-3p/SEMA3A轴促进OGD诱导的HBMECs的恶性进展。这项研究为circSEC11A在缺血性卒中细胞模型中的潜在应用提供了新的见解。
    BACKGROUND: Circular RNA (circRNA) has been found to play an important role in the progression of many diseases, including ischemic stroke. However, the regulatory mechanism of circSEC11A in ischemic stroke progression need to further investigation.
    METHODS: Human brain microvascular endothelial cells (HBMECs) were stimulated by oxygen glucose deprivation (OGD). CircSEC11A, SEC11A mRNA and miR (microRNA)-29a-3p were quantified by quantitative real-time PCR (qRT-PCR). SEMA3A, BAX and BCL2 protein level was quantified by western blot. Oxidative stress, cell proliferation, angiogenesis and apoptosis abilities were gauged by oxidative stress assay kit, 5-Ethynyl-2\'-Deoxyuridine (EdU) staining, tube formation assay and flow cytometry assays, respectively. Direct relationship between miR-29a-3p and circSEC11A or SEMA3A was validated by dual-luciferase reporter assay, RIP assay and RNA pull-down assay.
    RESULTS: CircSEC11A was upregulated in OGD-induced HBMECs. OGD promoted the oxidative stress and apoptosis and inhibited cell proliferation and angiogenesis, while circSEC11A knockdown relieved the effects. CircSEC11A functioned as the sponge for miR-29a-3p, and miR-29a-3p inhibitor reversed the effects of si-circSEC11A on OGD-induced HBMECs oxidative injuries. Moreover, SEMA3A served as the target gene of miR-29a-3p. MiR-29a-3p inhibition ameliorated OGD-induced HBMECs oxidative injuries, while SEMA3A overexpression rescued the impacts of miR-29a-3p mimic.
    CONCLUSIONS: CircSEC11A promoted the malignant progression in OGD-induced HBMECs through the mediation of miR-29a-3p/SEMA3A axis. This study has provided the new insight into the underlying application of circSEC11A in cell model of ischemic stroke.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Carnosol is a phytopolyphenol (diterpene) found and extracted from plants of Mediterranean diet, which has anti-tumor, anti-inflammatory and antioxidant effects. However, its role in ischemic stroke has not been elucidated.
    Primary neurons subjected to oxygen-glucose deprivation (OGD) was used to investigate the effect of carnosol in vitro. A mouse MCAO model was used to evaluate the effect of carnosol on ischemic stroke in vivo. The mRNA level of inflammatory and apoptosis-related genes was determined by RT-PCR. The protein level of total and phosphorylated AMPK was determined by WB. H&E and Immunofluorescent assay was used to investigate the necrosis, inflammation and apoptosis in brain tissue.
    Carnosol protected the activity of primary neurons subjected to oxygen-glucose deprivation (OGD) in vitro, as well as inhibited inflammation and apoptosis. Furthermore, carnosol could significantly reduce the infarct and edema volume and protect against neurological deficit in vivo, and had a significant inhibitory effect on brain neuroinflammation and apoptosis. Mechanically, carnosol could activate AMPK, and the effect of carnosol on cerebral ischemia-reperfusion injury cell model could be abolished by AMPK phosphorylation inhibitor.
    Carnosol has a protective effect on ischemic stroke, and this effect is achieved through AMPK activation. Our study demonstrates the protective effect of carnosol on cerebral ischemia-reperfusion injury and provides a new perspective for the clinical treatment of ischemic stroke.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号