首页> 外文期刊>Frontiers in Neuropharmacology >PPAR γ Prevents Neuropathic Pain by Down-Regulating CX3CR1 and Attenuating M1 Activation of Microglia in the Spinal Cord of Rats Using a Sciatic Chronic Constriction Injury Model
【24h】

PPAR γ Prevents Neuropathic Pain by Down-Regulating CX3CR1 and Attenuating M1 Activation of Microglia in the Spinal Cord of Rats Using a Sciatic Chronic Constriction Injury Model

机译:PPARγ通过坐标慢性收缩损伤模型通过下调节CX3CR1并通过曲线慢性收缩损伤模型衰减大鼠脊髓中微胶质细胞的M1活化的神经疗法

获取原文
       

摘要

Previous studies have proved that peripheral nerve injury is involved in the pathogenesis of neuropathic pain (NP). The peripheral nerve injury primes spinal M1 microglia phenotype and produces pro-inflammatory cytokines, which are responsible for neurotoxic and neuronal hyper-excitable outcomes. Spinal peroxisome proliferatoractivated receptor gamma (PPAR γ) has been shown to play an anti-inflammatory role in the development of NP. However, the role of PPAR γ in attenuating the pathological pathway of spinal microgliosis is still unknown. Methods: Sprague-Dawley rats (male, aged 8–10 weeks) were randomly divided into three groups, i.e., a control group, a NP group, and a NP + lentivirus encoding PPAR γ (LV-PPAR γ) group. The sciatic chronic constriction injury (CCI) model was used to induce NP in rats. Pain behavior was assessed by monitoring the rat hind-paw withdrawal threshold to mechanical stimuli and withdrawal latency to radiant heat. The LV-PPAR γ was intrathecally infused 1 day before CCI. Western blot analysis and realtime qPCR were used to detect the microglia phenotypic molecules and CX3CR1 expression in the spinal cord. In vitro, BV-2 microglia cells were transfected with LVPPAR γ and incubated with lipopolysaccharides (LPS), and the levels of M1 microglia phenotypic molecules and CX3CR1 in BV-2 microglia cells were assessed by western blot analysis, real-time qPCR, and enzyme-linked immunosorbent assay. Results: Preoperative intrathecal infusion of LV-PPAR γ attenuated pain in rats 7 days post-CCI. The M1-microglia marker, CX3CR1, and pro-inflammatory signaling factors were increased in the spinal cord of CCI rats, while the preoperative intrathecal infusion of LV-PPAR γ attenuated these changes and increased the expression of IL-10. In vitro, the overexpression of PPAR γ in BV-2 cells reduced LPS-induced M1 microglia polarization and the levels of CX3CR1 and pro-inflammatory cytokines.Conclusion: Intrathecal infusion of LV-PPAR γ exerts a protective effect on the development of NP induced by CCI in rats. The overexpression of PPAR γ may produce both analgesic and anti-inflammatory effects due to inhibition of the M1 phenotype and CX3CR1 signaling pathway in spinal microglia.
机译:先前的研究证明,周围神经损伤涉及神经性疼痛的发病机制(NP)。周围神经损伤引发脊髓M1微胶质细胞表型并产生促炎细胞因子,其负责神经毒性和神经元超易激发成果。已显示脊柱过氧化物酶促增生受体γ(PPARγ)在NP的发育中发挥抗炎作用。然而,PPARγ在衰减脊髓小叶细胞症病理途径方面的作用仍然未知。方法:将Sprague-Dawley大鼠(男性,8-10次)随机分为三组,即对照组,NP组和编码PPARγ(LV-PPARγ)组的NP +慢病毒。坐骨慢性收缩损伤(CCI)模型用于诱导大鼠NP。通过监测大鼠后爪抽出阈值对机械刺激和辐射延迟来评估疼痛行为。在CCI之前,LV-PPARγ在鞘内注入1天。 Western印迹分析和实时QPCR用于检测脊髓中的小胶质细胞表型分子和CX3CR1表达。体外,用LVPPARγ转染BV-2微胶质细胞并与脂多糖(LPS)孵育,通过Western印迹分析,实时QPCR和BV-2微胶质细胞中的M1微凝血性表型分子和CX3Cr1的水平。酶联免疫吸附测定。结果:在CCI后7天大鼠术前鞘内输注LV-PPARγ衰减疼痛。在CCI大鼠的脊髓中增加了M1-MICROGLIA标记,CX3CR1和促炎信号传导因子,而LV-PPARγ的术前鞘内输注抑制这些变化并增加了IL-10的表达。在体外,在BV-2细胞中的PPARγ的过度表达降低了LPS诱导的M1微胶质细胞偏振和CX3CR1和促炎细胞因子的水平。结论:LV-PPARγ的鞘内输注对NP诱导的影响产生保护作用通过CCI在大鼠。 PPARγ的过度表达由于脊髓微胶中的M1表型和CX3CR1信号通路的抑制,可以产生镇痛和抗炎作用。

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