中国神经再生研究(英文版) ›› 2018, Vol. 13 ›› Issue (11): 1974-1980.doi: 10.4103/1673-5374.239445

• 原著:退行性病与再生 • 上一篇    下一篇

金丝桃苷可保护血脑屏障免受Aβ1-42的神经毒性

  

  • 收稿日期:2018-07-09 出版日期:2018-11-15 发布日期:2018-11-15
  • 基金资助:

     中国国家自然科学基金项目(81573771),江苏省自然科学基金项目(BK20151599

Hyperoside protects the blood-brain barrier from neurotoxicity of amyloid beta 1–42

Chen-Yang Liu1, Kuan Bai2, Xiao-Hui Liu1, Li-Mi Zhang1, Gu-Ran Yu1   

  1. 1 Department of Neurology, Jiangsu Traditional Chinese Medicine Hospital, the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
    2 Graduate School of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
  • Received:2018-07-09 Online:2018-11-15 Published:2018-11-15
  • Contact: Gu-Ran Yu,yushengzh@126.com.
  • Supported by:

    This study was financially supported by the National Natural Science Foundation of China, No. 81573771; the Natural Science Foundation of Jiangsu Province of China, No. BK20151599.

摘要:

已有研究显示淀粉样β蛋白在阿尔茨海默病血脑屏障的破坏中发挥神经毒性,而金丝桃苷在体内外均具有抗淀粉样β蛋白的神经保护作用,作者团队既往研究也发现金丝桃苷能抑制Aβ1-42诱导的血脑屏障渗漏,但机制尚不明确。为此,实验设计以50,200,500μM金丝桃苷预处理bEnd.3细胞2h后,再进行24h的Aβ1-42暴露。以MTT检测细胞的存活率,以流式细胞术和TUNEL分析细胞凋亡,以Western blot分析凋亡相关蛋白Bax,Bcl-2,细胞色素c,Caspase-3, Caspase-8,Caspase-9和Caspase-12的表达水平、血脑屏障相关蛋白Occludin,claudin-5和ZO-1的表达水平以及降解细胞连接的基质金属蛋白酶2和基质金属蛋白酶9的表达水平。可见显示,单纯Aβ1-42暴露能显著增加bEnd.3细胞的凋亡,增加细胞中cleaved caspase-9/caspase-9,Bax/Bcl-2,cleaved caspase-8/caspase-8和cleaved caspase-12/caspase-12比值,增加细胞色素C表达及caspase-3的活性,降低ZO-1,Claudin-5和Occludin的水平,并增加基质金属蛋白酶2和基质金属蛋白酶9水平。以金丝桃苷预处理后逆转了上述变化,且作用效果呈浓度依赖性。结果说明金丝桃苷能减轻纤维状Aβ1-42诱导的血脑屏障破坏,为治疗阿尔茨海默病提供实验依据。

orcid:0000-0002-9356-8695(Gu-Ran Yu)

关键词: 阿尔茨海默病, A&beta, 1-42, 血脑屏障, bEnd.3细胞, 紧密连接蛋白, 金丝桃苷, 抗凋亡, 神经再生

Abstract:

Mounting evidence indicates that amyloid β protein (Aβ) exerts neurotoxicity by disrupting the blood-brain barrier (BBB) in Alzheimer’s disease. Hyperoside has neuroprotective effects both in vitro and in vivo against Aβ. Our previous study found that hyperoside suppressed Aβ1–42-induced leakage of the BBB, however, the mechanism remains unclear. In this study, bEnd.3 cells were pretreated with 50, 200, or 500 μM hyperoside for 2 hours, and then exposed to Aβ1–42 for 24 hours. Cell viability was determined using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay were used to analyze cell apoptosis. Western blot assay was carried out to analyze expression levels of Bax, Bcl-2, cytochrome c, caspase-3, caspse-8, caspase-9, caspase-12, occludin, claudin-5, zonula occludens-1, matrix metalloproteinase-2 (MMP-2), and MMP-9. Exposure to Aβ1–42 alone remarkably induced bEnd.3 cell apoptosis; increased ratios of cleaved caspase-9/caspase-9, Bax/Bcl-2, cleav ed caspase-8/caspase-8, and cleaved caspase-12/caspase-12; increased expression of cytochrome c and activity of caspase-3; diminished levels of zonula occludens-1, claudin-5, and occludin; and increased levels of MMP-2 and MMP-9. However, hyperoside pretreatment reversed these changes in a dose-dependent manner. Our findings confirm that hyperoside alleviates fibrillar Aβ1–42-induced BBB disruption, thus offering a feasible therapeutic application in Alzheimer’s disease.

Key words: nerve regeneration, Alzheimer’s disease, amyloid beta 1–42, blood-brain barrier, bEnd.3 cells, tight junction proteins, hyperoside, anti-apoptosis, neural regeneration