4.6 Article

In vivo base editing rescues Hutchinson-Gilford progeria syndrome in mice

Journal

NATURE
Volume 589, Issue 7843, Pages 608-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-020-03086-7

Keywords

-

Funding

  1. NIH [U01AI142756, UG3AI150551, UG3TR002636, RM1HG009490, R01EB022376, R35GM118062, Z01HG200305, R01HL146654, R01HL126784]
  2. Howard Hughes Medical Institute
  3. NSF GRFP fellowship
  4. Damon Runyon Cancer Research Foundation [DRG-2343-18]
  5. Helen Hay Whitney postdoctoral fellowship
  6. Progeria Research Foundation

Ask authors/readers for more resources

Using adenine base editor (ABE), researchers successfully corrected the pathogenic mutation in fibroblasts from children with Hutchinson-Gilford progeria syndrome, mitigating RNA mis-splicing, reducing progerin levels, and correcting nuclear abnormalities. In vivo base editing with AAV9 encoding ABE in transgenic mice resulted in substantial, durable correction of the mutation, restoration of normal RNA splicing, and improvement of vascular pathology, ultimately extending the lifespan of the mice. These findings demonstrate the potential of in vivo base editing as a treatment for genetic diseases.
Hutchinson-Gilford progeria syndrome (HGPS or progeria) is typically caused by a dominant-negative C center dot G-to-T center dot A mutation (c.1824 C>T; p.G608G) in LMNA, the gene that encodes nuclear lamin A. This mutation causes RNA mis-splicing that produces progerin, a toxic protein that induces rapid ageing and shortens the lifespan of children with progeria to approximately 14 years1-4. Adenine base editors (ABEs) convert targeted A center dot T base pairs to G center dot C base pairs with minimal by-products and without requiring double-strand DNA breaks or donor DNA templates(5,6) . Here we describe the use of an ABE to directly correct the pathogenic HGPS mutation in cultured fibroblasts derived from children with progeria and in a mouse model of HGPS. Lentiviral delivery of the ABE to fibroblasts from children with HGPS resulted in 87-91% correction of the pathogenic allele, mitigation of RNA mis-splicing, reduced levels of progerin and correction of nuclear abnormalities. Unbiased off-target DNA and RNA editing analysis did not detect off-target editing in treated patient-derived fibroblasts. In transgenic mice that are homozygous for the human LMNA c.1824 C>T allele, a single retro-orbital injection of adeno-associated virus 9 (AAV9) encoding the ABE resulted in substantial, durable correction of the pathogenic mutation (around 20-60% across various organs six months after injection), restoration of normal RNA splicing and reduction of progerin protein levels. In vivo base editing rescued the vascular pathology of the mice, preserving vascular smooth muscle cell counts and preventing adventitial fibrosis. A single injection of ABE-expressing AAV9 at postnatal day 14 improved vitality and greatly extended the median lifespan of the mice from 215 to 510 days. These findings demonstrate the potential of in vivo base editing as a possible treatment for HGPS and other genetic diseases by directly correcting their root cause.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available