Transposable element-derived double-stranded RNAs stimulate pro-inflammatory activation in human astrocytes

Physiology(2024)

引用 0|浏览0
暂无评分
摘要
Astrocyte-related inflammation contributes to neuroinflammation with brain aging and Alzheimer’s disease (AD), but the causes of pro-inflammatory astrocyte activation in this context are incompletely understood. One driver of pro-inflammatory signaling in astrocytes may be endogenous double-stranded RNA (dsRNA) derived from transposable elements (TEs). TEs make up more than 50% of the human genome, and recent evidence suggests a role for TEs in pro-inflammatory signaling with aging and neurodegenerative diseases. However, the role of TEs and/or TE-derived dsRNA in astrocyte-related inflammation, and the specific TEs involved, have yet to be investigated. Here, we increased the expression of TE transcripts in human astrocytes by suppressing ADAR1 (an enzyme involved in the suppression/regulation of endogenous dsRNA) and performed RNA-seq to broadly characterize resulting biological changes. Using a computational pipeline (SPRINT) to detect potential dsRNA transcripts through adenosine-to-inosine (A-to-I; a marker of ADAR1 activity) edits, we found that dsRNA transcripts were more likely to come from TEs compared to coding genes. Of these TEs, highly edited and increased transcripts consisted of the TE families Alu and LINE-1 (L1) elements. We confirmed the increase in L1 transcripts in vitro by characterizing colocalization of J2 (antibody for dsRNA) and an L1 element via fluorescence in situ hybridization. Increases in these TEs were associated with increases in pro-inflammatory signaling and decreases in expression of genes relating to astrocyte identity/function (e.g., synapse modifying and cholesterol-related genes). Lastly, we found similar increases in Alu and L1 transcripts in brain tissue of AD patients and older subjects. Together, these data suggest a role for TE-derived dsRNA in pro-inflammatory activation in astrocytes, and possibly in aging and AD. To expand on these findings, we have performed RNA immunoprecipitation sequencing to identify the specific TEs that form dsRNA with ADAR1 suppression in astrocytes, and we are developing an approach to mitigate their effects by increasing ADAR1 expression in the brains of old mice. NIA: F31AG084330, R01AG078859. This is the full abstract presented at the American Physiology Summit 2024 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
更多
查看译文
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要