...
首页> 外文期刊>Journal of neuroinflammation >TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain
【24h】

TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain

机译:TNFR1-JNK信号传导是LPS致敏的缺氧缺血性脑损伤后神经炎症和神经血管损伤的共享途径

获取原文

摘要

Background Hypoxic-ischemia (HI) and inflammation are the two major pathogenic mechanisms of brain injury in very preterm infants. The neurovascular unit is the major target of HI injury in the immature brain. Systemic inflammation may worsen HI by up-regulating neuroinflammation and disrupting the blood–brain barrier (BBB). Since neurons and oligodendrocytes, microvascular endothelial cells, and microglia may closely interact with each other, there may be a common signaling pathway leading to neuroinflammation and neurovascular damage after injury in the immature brain. TNF-α is a key pro-inflammatory cytokine that acts through the TNF receptor (TNFR), and c-Jun N-terminal kinases (JNK) are important stress-responsive kinases. Objective To determine if TNFR1-JNK signaling is a shared pathway underlying neuroinflammation and neurovascular injury after lipopolysaccharide (LPS)-sensitized HI in the immature brain. Methods Postpartum (P) day-5 mice received LPS or normal saline (NS) injection before HI. Immunohistochemistry, immunoblotting and TNFR1- and TNFR2-knockout mouse pups were used to determine neuroinflammation, BBB damage, TNF-α expression, JNK activation, and cell apoptosis. The cellular distribution of p-JNK, TNFR1/TNFR2 and cleaved caspase-3 were examined using immunofluorescent staining. Results The LPS?+?HI group had significantly greater up-regulation of activated microglia, TNF-α and TNFR1 expression, and increases of BBB disruption and cleaved caspase-3 levels at 24 hours post-insult, and showed more cortical and white matter injury on P17 than the control and NS?+?HI groups. Cleaved caspase-3 was highly expressed in microvascular endothelial cells, neurons, and oligodendroglial precursor cells. LPS-sensitized HI also induced JNK activation and up-regulation of TNFR1 but not TNFR2 expression in the microglia, endothelial cells, neurons, and oligodendrocyte progenitors, and most of the TNFR1-positive cells co-expressed p-JNK. Etanercept (a TNF-α inhibitor) and AS601245 (a JNK inhibitor) protected against LPS-sensitized HI brain injury. The TNFR1-knockout but not TNFR2-knockout pups had significant reduction in JNK activation, attenuation of microglial activation, BBB breakdown and cleaved caspase-3 expression, and showed markedly less cortical and white matter injury than the wild-type pups after LPS-sensitized HI. Conclusion TNFR1-JNK signaling is the shared pathway leading to neuroinflammation and neurovascular damage after LPS-sensitized HI in the immature brain.
机译:背景缺氧缺血(HI)和炎症是极早产儿脑损伤的两个主要致病机制。神经血管单位是未成熟大脑中HI损伤的主要靶标。全身性炎症可能通过上调神经炎症和破坏血脑屏障(BBB)而使HI恶化。由于神经元和少突胶质细胞,微血管内皮细胞和小胶质细胞可能彼此紧密相互作用,因此在未成熟的脑损伤后可能存在导致神经炎症和神经血管损害的常见信号通路。 TNF-α是通过TNF受体(TNFR)起作用的关键促炎细胞因子,而c-Jun N端激酶(JNK)是重要的应激反应激酶。目的确定未成熟脑中脂多糖(LPS)致敏的HI后,TNFR1-JNK信号传导是否是神经炎症和神经血管损伤的共同途径。方法产后(P)第5天小鼠在HI前接受LPS或生理盐水(NS)注射。免疫组织化学,免疫印迹和TNFR1和TNFR2敲除小鼠幼仔用于确定神经炎症,BBB损伤,TNF-α表达,JNK活化和细胞凋亡。使用免疫荧光染色检查p-JNK,TNFR1 / TNFR2和裂解的caspase-3的细胞分布。结果LPSα+ΔHI组在感染后24小时具有明显更高的活化小胶质细胞,TNF-α和TNFR1表达,并增加BBB破坏和caspase-3裂解水平,并显示更多的皮质和白质对P17的伤害高于对照组和NS?+?HI组。劈裂的caspase-3在微血管内皮细胞,神经元和少突胶质前体细胞中高度表达。 LPS敏感的HI还可诱导小胶质细胞,内皮细胞,神经元和少突胶质祖细胞中的TNFR1的JNK激活和TNFR2的表达上调,而大多数TNFR1阳性细胞共表达p-JNK。 Etanercept(一种TNF-α抑制剂)和AS601245(一种JNK抑制剂)可以抵抗LPS敏感的HI脑损伤。 LPS致敏后,TNFR1敲除的幼崽而非TNFR2敲除的幼崽的JNK活化,微胶质激活的减弱,BBB分解和caspase-3表达的裂解均显着降低,并且显示出比野生型幼崽明显更少的皮质和白质损伤嗨结论TNFR1-JNK信号通路是未成熟脑脂多糖致HI后神经炎症和神经血管损伤的共同途径。

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