中国神经再生研究(英文版) ›› 2021, Vol. 16 ›› Issue (3): 514-522.doi: 10.4103/1673-5374.293149

• 原著:脑损伤修复保护与再生 • 上一篇    下一篇

微小RNA-146a纳米颗粒可减轻创伤性脑损伤12h后的炎症反应

  

  • 出版日期:2021-03-15 发布日期:2020-12-17
  • 基金资助:

    这项工作得到了肯塔基州脊髓和头部损伤研究基金会的资助(15-12A)

Temporal changes in inflammatory mitochondria-enriched microRNAs following traumatic brain injury and effects of miR-146a nanoparticle delivery

Wang-Xia Wang1, 2, 3, *, #, Paresh Prajapati2, 4, #, Hemendra J. Vekaria2, 4, Malinda Spry2, 4, Amber L. Cloud2, 4, Patrick G. Sullivan2, 4, Joe E. Springer2, 4, *   

  1. 1 Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA;  2 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA;  3 Department of Pathology & Laboratory Medicine, University of Kentucky, Lexington, KY, USA;  4 Department of Neuroscience, University of Kentucky, Lexington, KY, USA
  • Online:2021-03-15 Published:2020-12-17
  • Contact: Wang-Xia Wang, PhD, wwangc@uky.edu; Joe E. Springer, PhD, jspring@uky.edu.
  • Supported by:
    This work is supported by a grant (15-12A) from the Kentucky Spinal Cord and Head Injury Research Trust to JES and WXW. 

摘要:

外伤性脑损伤引起快速而明显的神经炎症反应。因此,有效减轻有害的促炎反应,对于保持巨噬细胞的神经修复潜力具有积极的作用。一种新颖的可能减轻神经炎性反应的策略是使用纳米技术来递送靶向调控炎症信号的基因microRNA(miRNA)。作者既往研究显示,严重创伤性脑损伤后3-12h,海马中几种调节炎症的线粒体富集的miR-142-3p,miR-142-5p和miR-146a的活性发生了变化,但这几种症性miRNA在伤后更长时间的变化尚缺乏研究。实验研究了在大鼠受到严重的控制性脑皮质撞击损伤12 h后,炎症性miRNA在线粒体和细胞溶胶组分中的时间表达谱,发现虽然miR-142-3p,miR-142-5p和miR-146a在细胞质中表达水平升高,但线粒体富集的miRNA,即miR-142-3p和miR-142-5p在损伤海马体线粒体中持续至少3天明显减少,并于伤后7天恢复到接近正常水平。miR-146a在伤后3天内保持降低趋势,并在7天后恢复正常。相反,伤后7天恢线粒体内未富集的miR-124a,miR-150,miR-19b,miR-155和miR-223出现增加或者无变化。实验利用新型基于肽纳米颗粒进行了miR-146a模拟物的体外和体内递送,靶向调节受损脑中核因子κB的表达。发现miR-146a纳米颗粒可明显降低BV-2或SH-SY5Y细胞中核因子κB促炎途径的两个重要调节因子TNF受体相关因子6和白介素1受体相关激酶1的表达。在伤后立即给大脑注射miR-146a纳米颗粒,可明显降低海马TNF受体相关因子6和白介素1受体相关激酶1的水平。该实验显示了创伤性脑损伤12 h后炎性miRNA的亚细胞改变;同时证实了miR-146a纳米颗粒递送有减轻损伤大脑炎性反应的潜力。

https://orcid.org/0000-0002-8104-3779 (Wang-Xia Wang); 

https://orcid.org/0000-0001-9611-8107 (Joe E. Springer)

关键词: 细胞渗透性肽传递, 控制性脑皮质撞击, 炎症途径, 线粒体相关的microRNA, 纳米颗粒, 核因子κB, 创伤性脑损伤

Abstract: MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate post-transcriptional gene expression and contribute to all aspects of cellular function. We previously reported that the activities of several mitochondria-enriched miRNAs regulating inflammation (i.e., miR-142-3p, miR-142-5p, and miR-146a) are altered in the hippocampus at 3–12 hours following a severe traumatic brain injury. In the present study, we investigated the temporal expression profile of these inflammatory miRNAs in mitochondria and cytosol fractions at more chronic post-injury times following severe controlled cortical impact injury in rats. In addition, several inflammatory genes were analyzed in the cytosol fractions. The analysis showed that while elevated levels were observed in cytoplasm, the mitochondria-enriched miRNAs, miR-142-3p and miR-142-5p continued to be significantly reduced in mitochondria from injured hippocampi for at least 3 days and returned to near normal levels at 7 days post-injury. Although not statistically significant, miR-146a also remained at reduced levels for up to 3 days following controlled cortical impact injury, and recovered by 7 days. In contrast, miRNAs that are not enriched in mitochondria, including miR-124a, miR-150, miR-19b, miR-155, and miR-223 were either increased or demonstrated no change in their levels in mitochondrial fractions for 7 days. The one exception was that miR-223 levels were reduced in mitochondria at 1 day following injury. No major alterations were observed in sham operated animals. This temporal pattern was unique to mitochondria-enriched miRNAs and correlated with injury-induced changes in mitochondrial bioenergetics as well as expression levels of several inflammatory markers. These observations suggested a potential compartmental re-distribution of the mitochondria-enriched inflammatory miRNAs and may reflect an intracellular mechanism by which specific miRNAs regulate injury-induced inflammatory signaling. To test this, we utilized a novel peptide-based nanoparticle strategy for in vitro and in vivo delivery of a miR-146a mimic as a potential therapeutic strategy for targeting nuclear factor-kappaB inflammatory modulators in the injured brain. Nanoparticle delivery of miR-146a to BV-2 or SH-SY5Y cells significantly reduced expression of TNF receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1), two important modulators of the nuclear factor-kappaB (NF-κB) pro-inflammatory pathway. Moreover, injections of miR-146a containing nanoparticles into the brain immediately following controlled cortical impact injury significantly reduced hippocampal TNF receptor-associated factor 6 and interleukin-1 receptor-associated kinase 1 levels. Taken together, our studies demonstrate the subcellular alteration of inflammatory miRNAs after traumatic brain injury and establish proof of principle that nanoparticle delivery of miR-146a has therapeutic potential for modulating pro-inflammatory effectors in the injured brain. All of the studies performed were approved by the University of Kentucky Institutional Animal Care and Usage Committee (IACUC protocol # 2014-1300) on August 17, 2017.

Key words: cell permeable peptide-delivery, controlled cortical impact, inflammatory pathway, mitochondria-associated microRNA, nanoparticle, nuclear factor-kappaB, traumatic brain injury