Helmholtz Gemeinschaft

Search
Browse
Statistics
Feeds

Striated muscle-specific base editing enables correction of mutations causing dilated cardiomyopathy

[img]
Preview
PDF (Original Article) - Requires a PDF viewer such as GSview, Xpdf or Adobe Acrobat Reader
8MB
[img] Other (Supplementary Information)
52MB

Item Type:Article
Title:Striated muscle-specific base editing enables correction of mutations causing dilated cardiomyopathy
Creators Name:Grosch, M. and Schraft, L. and Chan, A. and Küchenhoff, L. and Rapti, K. and Ferreira, A.M. and Kornienko, J. and Li, S. and Radke, M.H. and Krämer, C. and Clauder-Münster, S. and Perlas, E. and Backs, J. and Gotthardt, M. and Dieterich, C. and van den Hoogenhof, M.M.G. and Grimm, D. and Steinmetz, L.M.
Abstract:Dilated cardiomyopathy is the second most common cause for heart failure with no cure except a high-risk heart transplantation. Approximately 30% of patients harbor heritable mutations which are amenable to CRISPR-based gene therapy. However, challenges related to delivery of the editing complex and off-target concerns hamper the broad applicability of CRISPR agents in the heart. We employ a combination of the viral vector AAVMYO with superior targeting specificity of heart muscle tissue and CRISPR base editors to repair patient mutations in the cardiac splice factor Rbm20, which cause aggressive dilated cardiomyopathy. Using optimized conditions, we repair >70% of cardiomyocytes in two Rbm20 knock-in mouse models that we have generated to serve as an in vivo platform of our editing strategy. Treatment of juvenile mice restores the localization defect of RBM20 in 75% of cells and splicing of RBM20 targets including TTN. Three months after injection, cardiac dilation and ejection fraction reach wild-type levels. Single-nuclei RNA sequencing uncovers restoration of the transcriptional profile across all major cardiac cell types and whole-genome sequencing reveals no evidence for aberrant off-target editing. Our study highlights the potential of base editors combined with AAVMYO to achieve gene repair for treatment of hereditary cardiac diseases.
Keywords:Cardiac Myocytes, Dilated Cardiomyopathy, Gene Editing, Mutation, Myocardium, RNA-Binding Proteins, Animals, Mice
Source:Nature Communications
ISSN:2041-1723
Publisher:Nature Publishing Group
Volume:14
Number:1
Page Range:3714
Date:22 June 2023
Official Publication:https://doi.org/10.1038/s41467-023-39352-1
PubMed:View item in PubMed

Repository Staff Only: item control page

Downloads

Downloads per month over past year

Open Access
MDC Library