4.8 Article

Maternal immune activation dysregulation of the fetal brain transcriptome and relevance to the pathophysiology of autism spectrum disorder

期刊

MOLECULAR PSYCHIATRY
卷 23, 期 4, 页码 1001-1013

出版社

NATURE PUBLISHING GROUP
DOI: 10.1038/mp.2017.15

关键词

-

资金

  1. University of California, San Diego Clinical and Translational Research Institute [KL2TR00099, 1KL2TR001444]
  2. Simons Foundation Autism Research Initiative [176540, 323220]
  3. National Institute of Health [R01MH110558]
  4. Child Health Research Institute at Stanford University School of Medicine
  5. NATIONAL CENTER FOR ADVANCING TRANSLATIONAL SCIENCES [UL1TR001442, KL2TR001444, KL2TR000099] Funding Source: NIH RePORTER
  6. NATIONAL INSTITUTE OF MENTAL HEALTH [R01MH096815, R01MH110558] Funding Source: NIH RePORTER

向作者/读者索取更多资源

Maternal immune activation (MIA) via infection during pregnancy is known to increase risk for autism spectrum disorder (ASD). However, it is unclear how MIA disrupts fetal brain gene expression in ways that may explain this increased risk. Here we examine how MIA dysregulates rat fetal brain gene expression (at a time point analogous to the end of the first trimester of human gestation) in ways relevant to ASD-associated pathophysiology. MIA downregulates expression of ASD-associated genes, with the largest enrichments in genes known to harbor rare highly penetrant mutations. MIA also downregulates expression of many genes also known to be persistently downregulated in the ASD cortex later in life and which are canonically known for roles in affecting prenatally late developmental processes at the synapse. Transcriptional and translational programs that are downstream targets of highly ASD-penetrant FMR1 and CHD8 genes are also heavily affected by MIA. MIA strongly upregulates expression of a large number of genes involved in translation initiation, cell cycle, DNA damage and proteolysis processes that affect multiple key neural developmental functions. Upregulation of translation initiation is common to and preserved in gene network structure with the ASD cortical transcriptome throughout life and has downstream impact on cell cycle processes. The cap-dependent translation initiation gene, EIF4E, is one of the most MIA-dysregulated of all ASD-associated genes and targeted network analyses demonstrate prominent MIA-induced transcriptional dysregulation of mTOR and EIF4E-dependent signaling. This dysregulation of translation initiation via alteration of the Tsc2-mTor-Eif4e axis was further validated across MIA rodent models. MIA may confer increased risk for ASD by dysregulating key aspects of fetal brain gene expression that are highly relevant to pathophysiology affecting ASD.

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

4.8
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