4.7 Article

Prior nasal delivery of antagomiR-122 prevents radiation-induced brain injury

期刊

MOLECULAR THERAPY
卷 29, 期 12, 页码 3465-3483

出版社

CELL PRESS
DOI: 10.1016/j.ymthe.2021.06.019

关键词

-

资金

  1. National Natural Science Foundation of China [82071475, 81671181]
  2. Guangdong Province Universities and Colleges Pearl River Scholar Funded Scheme (2017)
  3. Guangdong Province: Special Support Plan for HighLevel Talents Grant (2016)

向作者/读者索取更多资源

The study revealed that miR-122-5p plays a crucial role in regulating neuroinflammation in radiation-induced brain injury (RBI), suggesting that inhibiting miR-122-5p could alleviate radiation-induced cognitive impairment, neuronal injury, and neuroinflammation. The results also indicated that miR-122-5p inhibition in microglia may reduce proinflammatory cytokine levels and enhance phagocytic function to protect against radiation-induced neuronal injury, with Tensin 1 identified as a potential target of miR-122-5p in RBI. Additional safety assessments, optimization of drug delivery, and clarification of mechanisms are needed for clinical translation.
Radiation-induced brain injury is a major adverse event in head and neck tumor treatment, influencing the quality of life for the more than 50% of patients who undergo radiation therapy and experience long-term survival. However, no effective treatments are available for these patients, and preventative drugs and effective drug-delivery methods must be developed. Based on our results, miR-122-5p was upregulated in the mouse radiation-induced brain injury (RBI) model and patients with nasopharyngeal carcinoma (NPC) who received radiation therapy. Intranasal administration of a single antagomiR-122-5p dose before irradiation effectively alleviated radiation-induced cognitive impairment, neuronal injury, and neuroinflammation in the mouse RBI model. Results further indicated that miR-122-5p inhibition in microglia reduced the levels of proinflammatory cytokines and enhanced the phagocytic function to protect against radiation-induced neuronal injury in cell models. Further, we profiled transcriptome data and verified that Tensin 1 (TNS1) may be the target of miR-122-5p in RBI. In summary, our results reveal a distinct role for miR-122-5p in regulating neuroinflammation in RBI, indicating that a non-invasive strategy for intranasal miR-122-5p administration may be an attractive therapeutic target in RBI, providing new insights for clinical trials. Further systematic safety assessment, optimization of drug administration, and clarity of mechanism will accelerate the process into clinical practice.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据