中国神经再生研究(英文版) ›› 2021, Vol. 16 ›› Issue (6): 1037-1043.doi: 10.4103/1673-5374.300453

• 原著:脑损伤修复保护与再生 • 上一篇    下一篇

P53诱导糖酵解和凋亡调节因子可减轻缺氧缺血小胶质细胞焦亡和缺血性脑损伤

  

  • 出版日期:2021-06-15 发布日期:2020-12-31
  • 基金资助:

    国家自然科学基金项目(81872845,81771625);江苏省自然科学基金(BK20180207);江苏省医学青年人才项目(QNRC2016762);苏州市儿科临床中心项目(Szzx201504);江苏省研究生科研与实践创新计划(KYCX19_1998);江苏省政府留学奖学金(JS-2017-127);第五批固苏卫生人才计划

TP53-induced glycolysis and apoptosis regulator alleviates hypoxia/ischemia-induced microglial pyroptosis and ischemic brain damage

Lan-Lan Tan1, #, Xiao-Lu Jiang1, #, Li-Xiao Xu2, Gen Li2, Chen-Xi Feng2, Xin Ding1, Bin Sun1, Zheng-Hong Qin3, Zu-Bin Zhang3, *, Xing Feng1, *, Mei Li2, *#br#   

  1. 1 Department of Neonatology, Children’s Hospital of Soochow University, Suzhou, Jiangsu Province, China;  2 Department of Pediatrics Research Institute, Children’s Hospital of Soochow University, Suzhou, Jiangsu Province, China;  3 Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province, China 
  • Online:2021-06-15 Published:2020-12-31
  • Contact: Mei Li, PhD, meili_edu@163.com; Xing Feng, PhD, xing_feng66@suda.edu.cn; Zu-Bin Zhang, PhD, zubinzhang.2008@163.com.
  • Supported by:
    This work was supported by the National Natural Science Foundation of China, Nos. 81872845 (to ML), 81771625 (to XF); the Natural Science Foundation of Jiangsu Province of China, No. BK20180207 (to ML); Jiangsu Provincial Medical Youth Talent of China, No. QNRC2016762(to ML); the Pediatric Clinical Center of Suzhou City of China, No. Szzx201504 (to XF); Postgraduate Research & Practice Innovation Program of Jiangsu Province of China, No. KYCX19_1998 (to LLT); Jiangsu Government Scholarship for Overseas Studies of China, No. JS-2017-127 (to ML); and the Fifth Batch of Gusu Health Talent Plan of China (to ML).

摘要:

作者既往研究表明,P53诱导糖酵解和凋亡调节因子(TIGAR)可保护脑缺血再灌注后的神经元,但其对新生儿缺氧缺血性脑损伤的作用尚不明确。实验首先对7d龄SD大鼠行左侧颈总动脉永久闭塞后,缺氧2h建立体内缺氧缺血性脑损伤模型。在缺氧缺血性脑损伤6d前将LV-sh_TIGAR或LV-sh_GSDMD注射至大鼠左侧脑室和纹状体。然后通过对永生化小神经胶质细胞(HAPI小胶质细胞)行氧糖剥夺2h,并24h复氧建立体外缺氧缺血性脑损伤模型,在造模前3d行LV-sh_TIGAR或LV-sh_GSDMD转染。结果显示,缺氧缺血性脑损伤大鼠皮质和氧糖剥夺/复氧HAPI小胶质细胞中TIGAR表达增加,而慢病毒介导的TIGAR敲低可明显加重大鼠脑组织中的细胞焦亡和脑损伤。而外源性NADPH可增加氧糖剥夺/复氧细胞NADPH水平和谷胱甘肽/氧化型谷胱甘肽比例,降低活性氧水平。外源性NADPH可在体内外阻断TIGAR敲低对新生儿缺氧缺血性脑损伤的影响。表明TIGAR可抑制小胶质细胞焦亡,在新生儿缺氧缺血性脑损伤中起保护作用。实验于2017年经苏州大学伦理委员会批准,批准号:2017LW003。

https://orcid.org/0000-0001-9828-5531 (Mei Li)

关键词:

P53诱导糖酵解和凋亡调节因子, 细胞焦亡, NADPH, 小胶质细胞, 活性氧, 缺氧缺血性脑损伤, 体内, 体外

Abstract: Our previous studies have demonstrated that TP53-induced glycolysis and apoptosis regulator (TIGAR) can protect neurons after cerebral ischemia/reperfusion. However, the role of TIGAR in neonatal hypoxic-ischemic brain damage (HIBD) remains unknown. In the present study, 7-day-old Sprague-Dawley rat models of HIBD were established by permanent occlusion of the left common carotid artery followed by 2-hour hypoxia. At 6 days before induction of HIBD, a lentiviral vector containing short hairpin RNA of either TIGAR or gasdermin D (LV-sh_TIGAR or LV-sh_GSDMD) was injected into the left lateral ventricle and striatum. Highly aggressively proliferating immortalized (HAPI) microglial cell models of in vitro HIBD were established by 2-hour oxygen/glucose deprivation followed by 24-hour reoxygenation. Three days before in vitro HIBD induction, HAPI microglial cells were transfected with LV-sh_TIGAR or LV-sh_GSDMD. Our results showed that TIGAR expression was increased in the neonatal rat cortex after HIBD and in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Lentivirus-mediated TIGAR knockdown in rats markedly worsened pyroptosis and brain damage after hypoxia/ischemia in vivo and in vitro. Application of exogenous nicotinamide adenine dinucleotide phosphate (NADPH) increased the NADPH level and the glutathione/oxidized glutathione ratio and decreased reactive oxygen species levels in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Additionally, exogenous NADPH blocked the effects of TIGAR knockdown in neonatal HIBD in vivo and in vitro. These findings show that TIGAR can inhibit microglial pyroptosis and play a protective role in neonatal HIBD. The study was approved by the Animal Ethics Committee of Soochow University of China (approval No. 2017LW003) in 2017. 

Key words: hypoxic-ischemic brain damage, in vitro, in vivo, microglia, NADPH, pyroptosis, ROS, TIGAR