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

CV Theodoris, F Mourkioti, Y Huang… - The Journal of …, 2017 - Am Soc Clin Investig
CV Theodoris, F Mourkioti, Y Huang, SS Ranade, L Liu, HM Blau, D Srivastava
The Journal of clinical investigation, 2017Am Soc Clin Investig
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 …
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.
The Journal of Clinical Investigation