4.7 Article

Mechanisms of angiogenic incompetence in Hutchinson-Gilford progeria via downregulation of endothelial NOS

Journal

AGING CELL
Volume 20, Issue 7, Pages -

Publisher

WILEY
DOI: 10.1111/acel.13388

Keywords

ABE; aging; endothelial cells; eNOS; progeria

Funding

  1. NIH/NHLBI [R01HL126784, R01HL138252]
  2. NIH/NIA [R36AG070573]
  3. National Institute of Biomedical Imaging and Bioengineering/National Institutes of Health (NIBIB/NIH) Center for Engineering Complex Tissues [P41 EB023833]
  4. NIH [U01AI142756, UG3AI1505101, RM1HG009490, R01EB022376, R35GM118062]
  5. HHMI (DRL)

Ask authors/readers for more resources

HGPS is a rare genetic disorder caused by a mutation in the LMNA gene, leading to the production of toxic progerin protein that affects angiogenesis. Using iPSC-EC models, it was found that HGPS cells exhibit endothelial dysfunction and deficits in microvascular network formation.
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder with features of accelerated aging. The majority of HGPS cases are caused by a de novo point mutation in the LMNA gene (c.1824C>T; p.G608G) resulting in progerin, a toxic lamin A protein variant. Children with HGPS typically die from coronary artery diseases or strokes at an average age of 14.6 years. Endothelial dysfunction is a known driver of cardiovascular pathogenesis; however, it is currently unknown how progerin antagonizes normal angiogenic function in HGPS. Here, we use human iPSC-derived endothelial cell (iPSC-EC) models to study angiogenesis in HGPS. We cultured normal and HGPS iPSC-ECs under both static and fluidic culture conditions. HGPS iPSC-ECs show reduced endothelial nitric oxide synthase (eNOS) expression and activity compared with normal controls and concomitant decreases in intracellular nitric oxide (NO) level, which result in deficits in capillary-like microvascular network formation. Furthermore, the expression of matrix metalloproteinase 9 (MMP-9) was reduced in HGPS iPSC-ECs, while the expression of tissue inhibitor metalloproteinases 1 and 2 (TIMP1 and TIMP2) was upregulated relative to healthy controls. Finally, we used an adenine base editor (ABE7.10max-VRQR) to correct the pathogenic c.1824C>T allele in HGPS iPSC-ECs. Remarkably, ABE7.10max-VRQR correction of the HGPS mutation significantly reduced progerin expression to a basal level, rescued nuclear blebbing, increased intracellular NO level, normalized the misregulated TIMPs, and restored angiogenic competence in HGPS iPSC-ECs. Together, these results provide molecular insights of endothelial dysfunction in HGPS and suggest that ABE could be a promising therapeutic approach for correcting HGPS-related cardiovascular phenotypes.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available