...
首页> 外文期刊>BMC Neuroscience >Glutamate-induced apoptosis in primary cortical neurons is inhibited by equine estrogens via down-regulation of caspase-3 and prevention of mitochondrial cytochrome c release
【24h】

Glutamate-induced apoptosis in primary cortical neurons is inhibited by equine estrogens via down-regulation of caspase-3 and prevention of mitochondrial cytochrome c release

机译:马雌激素通过下调caspase-3并阻止线粒体细胞色素c释放来抑制谷氨酸诱导的原代皮层神经元凋亡。

获取原文
           

摘要

Background Apoptosis plays a key role in cell death observed in neurodegenerative diseases marked by a progressive loss of neurons as seen in Alzheimer's disease. Although the exact cause of apoptosis is not known, a number of factors such as free radicals, insufficient levels of nerve growth factors and excessive levels of glutamate have been implicated. We and others, have previously reported that in a stable HT22 neuronal cell line, glutamate induces apoptosis as indicated by DNA fragmentation and up- and down-regulation of Bax (pro-apoptotic), and Bcl-2 (anti-apoptotic) genes respectively. Furthermore, these changes were reversed/inhibited by estrogens. Several lines of evidence also indicate that a family of cysteine proteases (caspases) appear to play a critical role in neuronal apoptosis. The purpose of the present study is to determine in primary cultures of cortical cells, if glutamate-induced neuronal apoptosis and its inhibition by estrogens involve changes in caspase-3 protease and whether this process is mediated by Fas receptor and/or mitochondrial signal transduction pathways involving release of cytochrome c. Results In primary cultures of rat cortical cells, glutamate induced apoptosis that was associated with enhanced DNA fragmentation, morphological changes, and up-regulation of pro-caspase-3. Exposure of cortical cells to glutamate resulted in a time-dependent cell death and an increase in caspase-3 protein levels. Although the increase in caspase-3 levels was evident after 3 h, cell death was only significantly increased after 6 h. Treatment of cells for 6 h with 1 to 20 mM glutamate resulted in a 35 to 45% cell death that was associated with a 45 to 65% increase in the expression of caspase-3 protein. Pretreatment with caspase-3-protease inhibitor z-DEVD or pan-caspase inhibitor z-VAD significantly decreased glutamate-induced cell death of cortical cells. Exposure of cells to glutamate for 6 h in the presence or absence of 17β-estradiol or Δ8, 17β-estradiol (10 nM-10 μM) resulted in the prevention of cell death and was associated with a significant dose-dependent decrease in caspase-3 protein levels, with Δ8, 17β-E2 being more potent than 17β-E2. Protein levels of Fas receptor remained unchanged in the presence of glutamate. In contrast, treatment with glutamate induced, in a time-dependent manner, the release of cytochrome c into the cytosol. Cytosolic cytochrome c increased as early as 1.5 h after glutamate treatment and these levels were 5 fold higher after 6 h, compared to levels in the untreated cells. Concomitant with these changes, the levels of cytochrome c in mitochondria decreased significantly. Both 17β-E2 and Δ8, 17β-E2 reduced the release of cytochrome c from mitochondria into the cytosol and this decrease in cytosolic cytochrome c was associated with inhibition of glutamate-induced cell death. Conclusion In the primary cortical cells, glutamate-induced apoptosis is accompanied by up-regulation of caspase-3 and its activity is blocked by caspase protease inhibitors. These effects of glutamate on caspase-3 appear to be independent of changes in Fas receptor, but are associated with the rapid release of mitochondrial cytochrome c, which precedes changes in caspase-3 protein levels leading to apoptotic cell death. This process was differentially inhibited by estrogens with the novel equine estrogen Δ8, 17β-E2 being more potent than 17β-E2. To our knowledge, this is the first study to demonstrate that equine estrogens can prevent glutamate-induced translocation of cytochrome c from mitochondria to cytosol in rat primary cortical cells.
机译:背景凋亡在神经退行性疾病中观察到的细胞死亡中起着关键作用,其特征是如在阿尔茨海默氏病中所见,神经元逐渐丧失。尽管尚不清楚细胞凋亡的确切原因,但涉及许多因素,例如自由基,神经生长因子水平不足和谷氨酸水平过高。我们和其他人以前曾报道过,在稳定的HT22神经元细胞系中,谷氨酸诱导凋亡,这分别由DNA片段和Bax(促凋亡)和Bcl-2(抗凋亡)基因的上调和下调表示。 。此外,这些变化被雌激素逆转/抑制。几条证据还表明,半胱氨酸蛋白酶(胱天蛋白酶)家族似乎在神经元凋亡中起关键作用。本研究的目的是确定皮质细胞的原代培养物中,谷氨酸诱导的神经元凋亡及其是否受到雌激素的抑制是否涉及caspase-3蛋白酶的变化,以及该过程是否由Fas受体和/或线粒体信号转导途径介导。涉及细胞色素的释放c。结果在大鼠皮质细胞的原代培养物中,谷氨酸诱导凋亡,这与增强的DNA片段化,形态学变化和pro-caspase-3上调相关。皮质细胞暴露于谷氨酸导致时间依赖性细胞死亡和caspase-3蛋白水平增加。尽管3小时后caspase-3水平明显增加,但6小时后细胞死亡仅显着增加。用1至20 mM谷氨酸处理细胞6小时导致35至45%的细胞死亡,这与caspase-3蛋白表达增加45至65%有关。用caspase-3-蛋白酶抑制剂z-DEVD或pan-caspase抑制剂z-VAD预处理可显着降低谷氨酸诱导的皮质细胞死亡。在存在或不存在17β-雌二醇或Δ 8 ,17β-雌二醇(10 nM-10μM)的情况下,将细胞暴露于谷氨酸6小时可防止细胞死亡,并与细胞凋亡有关。 caspase-3蛋白水平显着剂量依赖性降低,其中Δ 8 ,17β-E 2 比17β-E 2 更有效。谷氨酸存在时,Fas受体的蛋白水平保持不变。相反,用谷氨酸盐处理以时间依赖性方式诱导细胞色素c释放到细胞质中。谷氨酸处理后,细胞溶质的细胞色素c最早在1.5 h时就升高了,与未处理的细胞相比,这些水平在6 h后提高了5倍。伴随这些变化,线粒体中细胞色素c的水平显着下降。 17β-E 2 和Δ 8 ,17β-E 2 都减少了线粒体中细胞色素c释放到细胞质中,并降低了细胞质细胞色素c与谷氨酸诱导的细胞死亡的抑制有关。结论在原代皮层细胞中,谷氨酸诱导的细胞凋亡伴随caspase-3的上调,其活性被caspase蛋白酶抑制剂阻断。谷氨酸对caspase-3的作用似乎与Fas受体的变化无关,但与线粒体细胞色素c的快速释放有关,后者在caspase-3蛋白水平变化之前导致凋亡细胞死亡。新型马雌激素Δ 8 比17β-E 2 更有效,雌激素可不同程度地抑制这一过程。据我们所知,这是第一项证明马雌激素可以防止谷氨酸诱导的大鼠原代皮层细胞中细胞色素c从线粒体向细胞质易位的转运。

著录项

相似文献

  • 外文文献
  • 中文文献
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