4.8 Article

Cyclic GMP-AMP synthase promotes the inflammatory and autophagy responses in Huntington disease

出版社

NATL ACAD SCIENCES
DOI: 10.1073/pnas.2002144117

关键词

striatal vulnerability; HTT-copy number; cotranslational cleavage; autophagy flux; pro-inflammatory response

资金

  1. Lottie French Lewis Fund of the Community Foundation for Palm Beach and Martin Counties
  2. NIH/National Institute of Neurological Disorders and Stroke [R01-NS087019-01A1, R01-NS094577-01A1]
  3. Cure Huntington Disease Initiative Foundation

向作者/读者索取更多资源

Huntington disease (HD) is caused by an expansion mutation of the N -terminal polyglutamine of huntingtin (mHTT). mHTT is ubiq- uitously present, but it induces noticeable damage to the brain 's striatum, thereby affecting motor, psychiatric, and cognitive func- tions. The striatal damage and progression of HD are associated with the inflammatory response; however, the underlying molec- ular mechanisms remain unclear. Here, we report that cGMP-AMP synthase (cGAS), a DNA sensor, is a critical regulator of inflamma- tory and autophagy responses in HD. Ribosome profiling revealed that the cGAS mRNA has high ribosome occupancy at exon 1 and codon-specific pauses at positions 171 (CCG) and 172 (CGT) in HD striatal cells. Moreover, the protein levels and activity of cGAS (based on the phosphorylated STING and phosphorylated TBK1 levels), and the expression and ribosome occupancy of cGAS- dependent inflammatory genes ( Ccl5 and Cxcl10 ) are increased in HD striatum. Depletion of cGAS diminishes cGAS activity and de- creases the expression of inflammatory genes while suppressing the up -regulation of autophagy in HD cells. In contrast, reinstating cGAS in cGAS-depleted HD cells activates cGAS activity and pro- motes inflammatory and autophagy responses. Ribosome profil- ing also revealed that LC3A and LC3B , the two major autophagy initiators, show altered ribosome occupancy in HD cells. We also detected the presence of numerous micronuclei, which are known to induce cGAS, in the cytoplasm of neurons derived from human HD embryonic stem cells. Collectively, our results indicate that cGAS is up -regulated in HD and mediates inflammatory and auto- phagy responses. Thus, targeting the cGAS pathway may offer therapeutic benefits in HD.

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