Efficient CRISPR/Cas9-mediated editing of trinucleotide repeat expansion in myotonic dystrophy patient-derived iPS and myogenic cells

S Dastidar, S Ardui, K Singh, D Majumdar… - Nucleic acids …, 2018 - academic.oup.com
S Dastidar, S Ardui, K Singh, D Majumdar, N Nair, Y Fu, D Reyon, E Samara, MFM Gerli
Nucleic acids research, 2018academic.oup.com
CRISPR/Cas9 is an attractive platform to potentially correct dominant genetic diseases by
gene editing with unprecedented precision. In the current proof-of-principle study, we
explored the use of CRISPR/Cas9 for gene-editing in myotonic dystrophy type-1 (DM1), an
autosomal-dominant muscle disorder, by excising the CTG-repeat expansion in the 3′-
untranslated-region (UTR) of the human myotonic dystrophy protein kinase (DMPK) gene in
DM1 patient-specific induced pluripotent stem cells (DM1-iPSC), DM1-iPSC-derived …
Abstract
CRISPR/Cas9 is an attractive platform to potentially correct dominant genetic diseases by gene editing with unprecedented precision. In the current proof-of-principle study, we explored the use of CRISPR/Cas9 for gene-editing in myotonic dystrophy type-1 (DM1), an autosomal-dominant muscle disorder, by excising the CTG-repeat expansion in the 3′-untranslated-region (UTR) of the human myotonic dystrophy protein kinase (DMPK) gene in DM1 patient-specific induced pluripotent stem cells (DM1-iPSC), DM1-iPSC-derived myogenic cells and DM1 patient-specific myoblasts. To eliminate the pathogenic gain-of-function mutant DMPK transcript, we designed a dual guide RNA based strategy that excises the CTG-repeat expansion with high efficiency, as confirmed by Southern blot and single molecule real-time (SMRT) sequencing. Correction efficiencies up to 90% could be attained in DM1-iPSC as confirmed at the clonal level, following ribonucleoprotein (RNP) transfection of CRISPR/Cas9 components without the need for selective enrichment. Expanded CTG repeat excision resulted in the disappearance of ribonuclear foci, a quintessential cellular phenotype of DM1, in the corrected DM1-iPSC, DM1-iPSC-derived myogenic cells and DM1 myoblasts. Consequently, the normal intracellular localization of the muscleblind-like splicing regulator 1 (MBNL1) was restored, resulting in the normalization of splicing pattern of SERCA1. This study validates the use of CRISPR/Cas9 for gene editing of repeat expansions.
Oxford University Press