...
首页> 外文期刊>Oxidative Medicine and Cellular Longevity >Hydrogen Gas Attenuates Hypoxic-Ischemic Brain Injury via Regulation of the MAPK/HO-1/PGC-1a Pathway in Neonatal Rats
【24h】

Hydrogen Gas Attenuates Hypoxic-Ischemic Brain Injury via Regulation of the MAPK/HO-1/PGC-1a Pathway in Neonatal Rats

机译:氢气通过调节新生大鼠的MAPK / HO-1 / PGC-1A途径衰减缺氧缺血性脑损伤

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Neonatal hypoxic-ischemic encephalopathy (HIE) is a leading cause of death in neonates with no effective treatments. Recent advancements in hydrogen (H2) gas offer a promising therapeutic approach for ischemia reperfusion injury; however, the impact of this approach for HIE remains a subject of debate. We assessed the therapeutic effects of H2 gas on HIE and the underlying molecular mechanisms in a rat model of neonatal hypoxic-ischemic brain injury (HIBI). H2 inhalation significantly attenuated neuronal injury and effectively improved early neurological outcomes in neonatal HIBI rats as well as learning and memory in adults. This protective effect was associated with initiation time and duration of sustained H2 inhalation. Furthermore, H2 inhalation reduced the expression of Bcl-2-associated X protein (BAX) and caspase-3 while promoting the expression of Bcl-2, nuclear factor erythroid-2-related factor 2, and heme oxygenase-1 (HO-1). H2 activated extracellular signal-regulated kinase and c-Jun N-terminal protein kinase and dephosphorylated p38 mitogen-activated protein kinase (MAPK) in oxygen-glucose deprivation/reperfusion (OGD/R) nerve growth factor-differentiated PC12 cells. Inhibitors of MAPKs blocked H2-induced HO-1 expression. HO-1 small interfering RNA decreased the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and sirtuin 1 (SIRT1) and reversed the protectivity of H2 against OGD/R-induced cell death. These findings suggest that H2 augments cellular antioxidant defense capacity through activation of MAPK signaling pathways, leading to HO-1 expression and subsequent upregulation of PGC-1α and SIRT-1 expression. Thus, upregulation protects NGF-differentiated PC12 cells from OGD/R-induced oxidative cytotoxicity. In conclusion, H2 inhalation exerted protective effects on neonatal rats with HIBI. Early initiation and prolonged H2 inhalation had better protective effects on HIBI. These effects of H2 may be related to antioxidant, antiapoptotic, and anti-inflammatory responses. HO-1 plays an important role in H2-mediated protection through the MAPK/HO-1/PGC-1α pathway. Our results support further assessment of H2 as a potential therapeutic for neurological conditions in which oxidative stress and apoptosis are implicated.
机译:新生儿缺氧缺血性脑病(HIE)是NeoNates死亡的主要原因,没有有效的治疗。氢气(H2)气体的最新进展为缺血再灌注损伤提供了有希望的治疗方法;然而,这种方法对HIE的影响仍然是辩论的主题。我们评估了H2气体对新生儿缺氧脑损伤(HIBI)大鼠模型中HIE和潜在分子机制的治疗效果。 H2吸入显着减弱了神经元损伤,并有效改善了新生儿Hibi大鼠的早期神经原因,以及成人中的学习和记忆。该保护作用与持续的H2吸入的开始时间和持续时间有关。此外,H 2吸入降低了Bcl-2相关X蛋白(Bax)和Caspase-3的表达,同时促进Bcl-2,核因子红外血清2-相关系数2和血红素氧合酶-1的表达(HO-1 )。 H2活化细胞外信号调节激酶和C-JUN N-末端蛋白激酶和去磷酸化的P38丝裂剂活化蛋白激活蛋白激酶(MAPK)在氧葡萄糖剥夺/再灌注(OGD / R)神经生长因子分化的PC12细胞中。 MAPK的抑制剂阻断了H2诱导的HO-1表达。 HO-1小干扰RNA降低过氧化物体增殖物激活受体γ1-α(PGC-1α)和Sirtuin1(SIRT1)的表达,并反转H2对OGD / R诱导的细胞死亡的保护性。这些发现表明,H2通过激活MAPK信号传导途径增强了细胞抗氧化防御能力,导致HO-1表达和随后的PGC-1α和SIRT-1表达的上调。因此,上调保护来自OGD / R诱导的氧化细胞毒性的NGF分化的PC12细胞。总之,H2吸入对Hibi对新生大鼠的保护作用。早期开始和延长的H2吸入对Hibi具有更好的保护作用。 H2的这些效果可能与抗氧化剂,抗曝光和抗炎反应有关。 HO-1通过MAPK / HO-1 / PGC-1α途径在H2介导的保护中起重要作用。我们的研究结果支持对H2的进一步评估作为神经系统条件的潜在治疗方法,其中氧化应激和细胞凋亡是牵连的。

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