...
首页> 外文期刊>Journal of neuroinflammation >P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite
【24h】

P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite

机译:P2X7R阻滞剂可在大鼠脑出血模型中阻止NLRP3炎性体活化和脑损伤:过氧亚硝酸盐的参与

获取原文

摘要

Background The NLR family, pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in intracerebral hemorrhage (ICH)-induced inflammatory injury, and the purinergic 2X7 receptor (P2X7R) is upstream of NLRP3 activation. This study aimed to investigate how P2X7R functions in ICH-induced inflammatory injury and how the receptor interacts with the NLRP3 inflammasome. Methods Rats were treated with P2X7R small interfering RNA (siRNA) 24 h before undergoing collagenase-induced ICH. A selective P2X7R inhibitor (blue brilliant G, BBG) or a peroxynitrite (ONOO ? ) decomposition catalyst (5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron(III) [FeTPPS]) was injected 30 min after ICH. Brain water content, hemorrhagic lesion volume, and neurological deficits were evaluated, and western blot, immunofluorescence, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) were carried out. Results Striatal P2X7R and NLRP3 inflammasomes were activated after ICH. Gene silencing of P2X7R suppressed NLRP3 inflammasome activation and interleukin (IL)-1β/IL-18 release and significantly ameliorated brain edema and neurological deficits. Additionally, enhanced NADPH oxidase 2 (NOX2, gp91 phox ) and inducible nitric oxide synthase (iNOS), as well as their cytotoxic product (ONOO ? ) were markedly attenuated by BBG treatment following ICH. This was accompanied by downregulations of the inflammasome components, IL-1β/IL-18 and myeloperoxidase (MPO, a neutrophil marker). Most importantly, inflammasome activation and IL-1β/IL-18 release were significantly inhibited by ONOO ? decomposition with FeTPPS. Conclusions Our findings implicate that P2X7R exacerbated inflammatory progression and brain damage in ICH rats possibly via NLRP3 inflammasome-dependent IL-1β/IL-18 release and neutrophil infiltration. ONOO ? , a potential downstream signaling molecule of P2X7R, may play a critical role in triggering NLRP3 inflammasome activation.
机译:背景NLR家族,含吡啶结构域3(NLRP3)炎性小体在脑出血(ICH)引起的炎症性损伤中起关键作用,而嘌呤能2X7受体(P2X7R)在NLRP3激活的上游。这项研究旨在研究P2X7R如何在ICH引起的炎症性损伤中起作用,以及该受体如何与NLRP3炎性小体相互作用。方法大鼠于胶原酶诱导的ICH前24小时接受P2X7R小干扰RNA(siRNA)治疗。 ICH注射30分钟后,注入了选择性的P2X7R抑制剂(蓝色亮光G,BBG)或过亚硝酸盐(ONOO?)分解催化剂(5,10,15,20-四(4-磺基苯基)卟啉铁(III)[FeTPPS]) 。评价脑含水量,出血病灶体积和神经功能缺损,并进行蛋白质印迹,免疫荧光和末端脱氧核苷酸转移酶dUTP缺口末端标记(TUNEL)。结果ICH后纹状体P2X7R和NLRP3炎性小体被激活。 P2X7R的基因沉默抑制了NLRP3炎性体的激活和白介素(IL)-1β/ IL-18的释放,并显着改善了脑水肿和神经功能缺损。另外,ICH后,BBG处理显着减弱了增强的NADPH氧化酶2(NOX2,gp91phox)和诱导型一氧化氮合酶(iNOS)及其细胞毒性产物(ONOOβ)。这伴随着炎性体成分IL-1β/ IL-18和髓过氧化物酶(MPO,嗜中性粒细胞标记物)的下调。最重要的是,ONOO能够显着抑制炎症小体的活化和IL-1β/ IL-18的释放。 FeTPPS分解。结论我们的发现暗示P2X7R可能通过NLRP3炎性小体依赖性IL-1β/ IL-18释放和嗜中性白细胞浸润加重了ICH大鼠的炎症进程和脑损伤。 ONOO? P2X7R的潜在下游信号分子可能在触发NLRP3炎性体激活中起关键作用。

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