Abstract

Gene editing methods are an attractive therapeutic option for Duchenne muscular dystrophy, and they have an immediate application in the generation of research models. To generate myoblast cultures that could be useful in in vitro drug screening, we have optimised a CRISPR/Cas9 gene edition protocol. We have successfully used it in wild type immortalised myoblasts to delete exon 52 of the dystrophin gene, modelling a common Duchenne muscular dystrophy mutation; and in patient’s immortalised cultures we have deleted an inhibitory microRNA target region of the utrophin UTR, leading to utrophin upregulation. We have characterised these cultures by demonstrating, respectively, inhibition of dystrophin expression and overexpression of utrophin, and evaluating the expression of myogenic factors (Myf5 and MyH3) and components of the dystrophin associated glycoprotein complex (α-sarcoglycan and β-dystroglycan). To demonstrate their use in the assessment of DMD treatments, we have performed exon skipping on the DMDΔ52-Model and have used the unedited DMD cultures/ DMD-UTRN-Model combo to assess utrophin overexpression after drug treatment. While the practical use of DMDΔ52-Model is limited to the validation to our gene editing protocol, DMD-UTRN-Model presents a possible therapeutic gene edition target as well as a useful positive control in the screening of utrophin overexpression drugs.

Highlights

  • Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive disease affecting 1 out of 5.000 newborn males

  • The most efficient combination of sgRNAs in HEK293 cells for each objective was selected to be used in the transfection of human immortalized myoblasts (Supplementary figure 1C and D)

  • After fluorescence activated cell sorting (FACS) sorting of individual GFP-positive cells, clones were expanded for DNA extraction (Supplementary figure 2)

Read more

Summary

Introduction

Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive disease affecting 1 out of 5.000 newborn males It is commonly caused by deletions disrupting the open reading frame of the DMD gene causing a lack of dystrophin p­ rotein[1]. It is important to assess exon-skipping strategies targeting other DMD ­exons[9] and therapies that may benefit all DMD and BMD patients, independent of their mutations. One such potential therapy is gene transfer: several trials are ongoing testing different drugs (SGT-001, SRP-9001 or PF-06939926). Several new compounds that aim to overexpress utrophin are currently being d­ eveloped[27,28,29], and this preclinical development could benefit from a gold standard or an adequate positive control to use in these assays

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call