4.8 Article

Long telomeres protect against age-dependent cardiac disease caused by NOTCH1 haploinsufficiency

期刊

JOURNAL OF CLINICAL INVESTIGATION
卷 127, 期 5, 页码 1683-1688

出版社

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI90338

关键词

-

资金

  1. L.K. Whittier and Roddenberry Foundations
  2. Younger Family Fund
  3. California Institute for Regenerative Medicine (CIRM) [DISC2-08098]
  4. NIH/National Heart, Lung, and Blood Institute/National Center for Research Resources [U01-HL098179, U01-HL100406, C06-RR018928]
  5. Baxter Foundation
  6. NIH [AG044815, AG009521, NS089533, AR063963, AG020961]
  7. American Heart Association (AHA)
  8. Roddenberry Discovery Fellowship
  9. Winslow Discovery Fellowship
  10. UCSF Discovery Fellowship
  11. UCSF Program in Developmental and Stem Cell Biology [NIH-T32HD007470]
  12. Medical Scientist Training Program (MSTP) [NIH-T32GM007618]
  13. AHA [10SDG3510024]
  14. Perelman School of Medicine

向作者/读者索取更多资源

Diseases caused by gene haploinsufficiency in humans commonly lack a phenotype in mice that are heterozygous for the orthologous factor, impeding the study of complex phenotypes and critically limiting the discovery of therapeutics. Laboratory mice have longer telomeres relative to humans, potentially protecting against age-related disease caused by haploinsufficiency. Here, we demonstrate that telomere shortening in NOTCH1-haploinsufficient mice is sufficient to elicit age-dependent cardiovascular disease involving premature calcification of the aortic valve, a phenotype that closely mimics human disease caused by NOTCH1 haploinsufficiency. Furthermore, progressive telomere shortening correlated with severity of disease, causing cardiac valve and septal disease in the neonate that was similar to the range of valve disease observed within human families. Genes that were dysregulated due to NOTCH1 haploinsufficiency in mice with shortened telomeres were concordant with proosteoblast and proinflammatory gene network alterations in human NOTCH1 heterozygous endothelial cells. These dysregulated genes were enriched for telomere-contacting promoters, suggesting a potential mechanism for telomere-dependent regulation of homeostatic gene expression. These findings reveal a critical role for telomere length in a mouse model of age-dependent human disease and provide an in vivo model in which to test therapeutic candidates targeting the progression of aortic valve disease.

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