...
首页> 外文期刊>Journal of neuroinflammation >Early activation of nSMase2/ceramide pathway in astrocytes is involved in ischemia-associated neuronal damage via inflammation in rat hippocampi
【24h】

Early activation of nSMase2/ceramide pathway in astrocytes is involved in ischemia-associated neuronal damage via inflammation in rat hippocampi

机译:星形胶质细胞中nSMase2 /神经酰胺途径的早期激活与大鼠海马区炎症引起的缺血相关神经元损伤有关

获取原文

摘要

Background Ceramide accumulation is considered a contributing factor to neuronal dysfunction and damage. However, the underlying mechanisms that occur following ischemic insult are still unclear. Methods In the present study, we established cerebral ischemia models using four-vessel occlusion and oxygen-glucose deprivation methods. The hippocampus neural cells were subjected to immunohistochemistry and immunofluorescence staining for ceramide and neutral sphingomyelinase 2 (nSMase2) levels; immunoprecipitation and immunoblot analysis for nSMase2, receptor for activated C kinase 1 (RACK1), embryonic ectoderm development (EED), p38 mitogen-activated protein kinase (p38MAPK) and phosphorylated p38MAPK expression; SMase assay for nSMase and acid sphingomyelinase (aSMase) activity; real-time reverse transcription polymerase chain reaction for cytokine expression; and Nissl, microtubule-associated protein 2 and terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling staining. Results We found considerable production of ceramide in astrocytes, but not in neurons, during early cerebral ischemia. This was accompanied by the induction of nSMase (but not aSMase) activity in the rat hippocampi. The inhibition of nSMase2 activity effectively reduced ceramide accumulation in astrocytes and alleviated neuronal damage to some extent. Meanwhile, the expression levels of proinflammatory cytokines, including tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β) and IL-6, were found to be upregulated, which may have played an import role in neuronal damage mediated by the nSMase2/ceramide pathway. Although enhanced binding of nSMase2 with RACK1 and EED were also observed after cerebral ischemia, nSMase2 activity was not blocked by the TNF-α receptor inhibitor through RACK1/EED signaling. p38MAPK, but not protein kinase Cζ or protein phosphatase 2B, was able to induce nSMase2 activation after ischemia. p38MAPK can be induced by A2B adenosine receptor (A2BAR) activity. Conclusions These results indicate that the inhibition of ceramide production in astrocytes by targeting A2BAR/p38MAPKSMase2 signaling may represent a viable approach for attenuating inflammatory responses and neuronal damage after cerebral ischemia.
机译:背景神经酰胺积聚被认为是神经元功能障碍和损害的一个成因。但是,尚不清楚缺血性损伤后发生的潜在机制。方法在本研究中,我们采用四血管阻塞和氧葡萄糖剥夺方法建立了脑缺血模型。对海马神经细胞进行了神经酰胺和中性鞘磷脂酶2(nSMase2)水平的免疫组化和免疫荧光染色。 nSMase2的免疫沉淀和免疫印迹分析,活化的C激酶1的受体(RACK1),胚外胚层发育(EED),p38促丝裂原活化的蛋白激酶(p38MAPK)和磷酸化的p38MAPK表达; SMase检测nSMase和酸性鞘磷脂酶(aSMase)的活性;实时逆转录聚合酶链反应用于细胞因子表达; Nissl,微管相关蛋白2和末端脱氧核苷酸转移酶介导的脱氧尿苷三磷酸缺口末端标记染色。结果我们发现,在早期脑缺血期间,星形胶质细胞中产生了大量神经酰胺,但在神经元中却没有。这伴随着在大鼠海马体中诱导nSMase(而不是aSMase)活性。抑制nSMase2活性可有效减少神经酰胺在星形胶质细胞中的积累,并在一定程度上减轻神经元损伤。同时,发现肿瘤坏死因子α(TNF-α),白细胞介素1β(IL-1β)和IL-6等促炎细胞因子的表达水平上调,这可能在由β介导的神经元损伤中起重要作用。 nSMase2 /神经酰胺途径。尽管在脑缺血后也观察到nSMase2与RACK1和EED的结合增强,但是TNF-α受体抑制剂通过RACK1 / EED信号传导并未阻断nSMase2的活性。 p38MAPK,但不是蛋白激酶Cζ或蛋白磷酸酶2B,却能够在缺血后诱导nSMase2激活。 p38MAPK可由A2B腺苷受体(A2BAR)活性诱导。结论这些结果表明,通过靶向A2BAR / p38MAPK / nSMase2信号通路抑制星形胶质细胞中神经酰胺的产生可能是减轻脑缺血后炎症反应和神经元损伤的可行方法。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号