...
首页> 外文期刊>Cellular Physiology and Biochemistry >HMGB1 a-Box Reverses Brain Edema and Deterioration of Neurological Function in a Traumatic Brain Injury Mouse Model
【24h】

HMGB1 a-Box Reverses Brain Edema and Deterioration of Neurological Function in a Traumatic Brain Injury Mouse Model

机译:HMGB1 a-Box逆转脑水肿和创伤性脑损伤小鼠模型中神经功能的恶化。

获取原文

摘要

Background/Aims Traumatic brain injury (TBI) is a complex neurological injury in young adults lacking effective treatment. Emerging evidences suggest that inflammation contributes to the secondary brain injury following TBI, including breakdown of the blood brain barrier (BBB), subsequent edema and neurological deterioration. High mobility group box-1 (HMGB1) has been identified as a key cytokine in the inflammation reaction following TBI. Here, we investigated the therapeutic efficacy of HMGB1 A-box fragment, an antagonist competing with full-length HMGB1 for receptor binding, against TBI. Methods TBI was induced by controlled cortical impact (CCI) in adult male mice. HMGB1 A-box fragment was given intravenously at 2 mg/kg/day for 3 days after CCI. HMGB1 A-box-treated CCI mice were compared with saline-treated CCI mice and sham mice in terms of BBB disruption evaluated by Evan’s blue extravasation, brain edema by brain water content, cell death by propidium iodide staining, inflammation by Western blot and ELISA assay for cytokine productions, as well as neurological functions by the modified Neurological Severity Score, wire grip and beam walking tests. Results HMGB1 A-box reversed brain damages in the mice following TBI. It significantly reduced brain edema by protecting integrity of the BBB, ameliorated cell degeneration, and decreased expression of pro-inflammatory cytokines released in injured brain after TBI. These cellular and molecular effects were accompanied by improved behavioral performance in TBI mice. Notably, HMGB1 A-box blocked IL-1β-induced HMGB1 release, and preferentially attenuated TLR4, Myd88 and P65 in astrocyte cultures. Conclusion Our data suggest that HMGB1 is involved in CCI-induced TBI, which can be inhibited by HMGB1 A-box fragment. Therefore, HMGB1 A-box fragment may have therapeutic potential for the secondary brain damages in TBI.
机译:背景/目的颅脑外伤(TBI)是缺乏有效治疗的年轻人的复杂神经系统损伤。新兴证据表明,炎症是TBI后继发性继发性脑损伤的原因,包括血脑屏障(BBB)破裂,随后的水肿和神经系统恶化。高迁移率族box-1(HMGB1)已被确定为TBI后炎症反应中的关键细胞因子。在这里,我们研究了HMGB1 A-box片段(与全长HMGB1竞争受体结合的拮抗剂)对TBI的治疗效果。方法成年雄性小鼠通过控制皮质撞击(CCI)诱导TBI。 CCI后,以2 mg / kg /天的剂量静脉内给予HMGB1 A-box片段,持续3天。 HMGB1 A-box处理的CCI小鼠与盐水处理的CCI小鼠和假小鼠在BBB破坏方面进行了比较,通过Evan的蓝色渗出评估,通过脑水含量评估脑水肿,通过​​碘化丙啶染色检测细胞死亡,通过Western blot和ELISA评估炎症通过改进的神经系统严重性评分,线握和束步测试对细胞因子产生以及神经功能进行检测。结果TBI后,HMGB1 A-box逆转了小鼠的脑损伤。它通过保护BBB的完整性,改善的细胞变性以及减少TBI后在受伤的脑中释放的促炎性细胞因子的表达而大大减少了脑水肿。这些细胞和分子效应伴随着TBI小鼠行为表现的改善。值得注意的是,HMGB1 A-box可阻断星形胶质细胞培养物中IL-1β诱导的HMGB1释放,并优先减弱TLR4,Myd88和P65。结论我们的数据表明HMGB1参与CCI诱导的TBI,可被HMGB1 A-box片段抑制。因此,HMGB1 A-box片段可能具有治疗TBI中继发性脑损伤的潜力。

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