4.8 Article

STAT1-induced ASPP2 transcription identifies a link between neuroinflammation, cell polarity, and tumor suppression

出版社

NATL ACAD SCIENCES
DOI: 10.1073/pnas.1407898111

关键词

TP53BP2; TLR4; multiple sclerosis

资金

  1. Ludwig Institute for Cancer Research Ltd.
  2. National Institutes of Health-Oxford Scholars Program
  3. Medical Research Council [MR/J000930/1]
  4. Fondation Contre le Cancer [IAP P7/43 BeMGI, ARC 10/15-027]
  5. St. John's College
  6. European Union Neurobid Consortium
  7. National Institutes of Health [R01 NS-42253]
  8. Cancer Research-United Kingdom Oxford Cancer Centre Development Fund
  9. Oxford National Institute for Health Research Biomedical Research Centre
  10. Biotechnology and Biological Sciences Research Council [BB/I021833/1] Funding Source: researchfish
  11. Medical Research Council [G0900901, MR/J000930/1, MR/L022656/1] Funding Source: researchfish
  12. BBSRC [BB/I021833/1] Funding Source: UKRI
  13. MRC [MR/J000930/1, G0900901, MR/L022656/1] Funding Source: UKRI

向作者/读者索取更多资源

Inflammation and loss of cell polarity play pivotal roles in neurodegeneration and cancer. A central question in both diseases is how the loss of cell polarity is sensed by cell death machinery. Here, we identify apoptosis-stimulating protein of p53 with signature sequences of ankyrin repeat-, SH3 domain-, and proline-rich region-containing protein 2 (ASPP2), a haploinsufficient tumor suppressor, activator of p53, and regulator of cell polarity, as a transcriptional target of signal transducer and activator of transcription 1 (STAT1). LPS induces ASPP2 expression in murine macrophage and microglial cell lines, a human monocyte cell line, and primary human astrocytes in vitro. LPS and IFNs induce ASPP2 transcription through an NF-kappa B RELA/p65-independent but STAT1-dependent pathway. In an LPS-induced maternal inflammation mouse model, LPS induces nuclear ASPP2 in vivo at the blood-cerebral spinal fluid barrier (the brain's barrier to inflammation), and ASPP2 mediates LPS-induced apoptosis. Consistent with the role of ASPP2 as a gatekeeper to inflammation, ASPP2-deficient brains possess enhanced neuroinflammation. Elevated ASPP2 expression is also observed in mouse models and human neuroinflammatory disease tissue, where ASPP2 was detected in GFAP-expressing reactive astrocytes that coexpress STAT1. Because the ability of ASPP2 to maintain cellular polarity is vital to CNS development, our findings suggest that the identified STAT1/ASPP2 pathway may connect tumor suppression and cell polarity to neuroinflammation.

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