Abstract

Autosomal dominant optic atrophy (DOA) is the most common inherited optic neuropathy in the United Kingdom. DOA has an insidious onset in early childhood, typically presenting with bilateral, central visual loss caused by the preferential loss of retinal ganglion cells. 60%–70% of genetically confirmed DOA cases are associated with variants in OPA1, a ubiquitously expressed GTPase that regulates mitochondrial homeostasis through coordination of inner membrane fusion, maintenance of cristae structure, and regulation of bioenergetic output. Whether genetic correction of OPA1 pathogenic variants can alleviate disease-associated phenotypes remains unknown. Here, we demonstrate generation of patient-derived OPA1 c.1334G>A: p.R445H mutant induced pluripotent stem cells (iPSCs), followed by correction of OPA1 through CRISPR-Cas9-guided homology-directed repair (HDR) and evaluate the effect of OPA1 correction on mitochondrial homeostasis. CRISPR-Cas9 gene editing demonstrated an efficient method of OPA1 correction, with successful gene correction in 57% of isolated iPSCs. Correction of OPA1 restored mitochondrial homeostasis, re-establishing the mitochondrial network and basal respiration and ATP production levels. In addition, correction of OPA1 re-established the levels of wild-type (WT) mitochondrial DNA (mtDNA) and reduced susceptibility to apoptotic stimuli. These data demonstrate that nuclear gene correction can restore mitochondrial homeostasis and improve mtDNA integrity in DOA patient-derived cells carrying an OPA1 variant.

Highlights

  • Autosomal dominant optic atrophy (DOA) is the most prevalent inherited optic neuropathy within the United Kingdom, affecting approximately 1 in 25,000 people.[1]

  • Generation of OPA1 mutant induced pluripotent stem cell (iPSC) and isogenic controls via CRISPR-Cas[9] To establish and characterize a human DOA disease model, dermal fibroblasts from an affected individual with DOA harboring a missense variant c.1334G>A: p.R445H (DOA-iPSC), which is associated with DOA+, in the OPA1 GTPase domain, were re-programmed to iPSCs alongside wild-type (WT) control fibroblasts using non-integrating episomal plasmids and nucleofection.[30]

  • Despite our current knowledge surrounding the function of OPA1 in mitochondrial homeostasis, therapeutic options for DOA remain limited

Read more

Summary

Introduction

Autosomal dominant optic atrophy (DOA) is the most prevalent inherited optic neuropathy within the United Kingdom, affecting approximately 1 in 25,000 people.[1]. 432 Molecular Therapy: Nucleic Acids Vol 26 December 2021 a 2021 The Author(s). Www.moleculartherapy.org that all 8 splice isoforms of human OPA1 are capable of rescuing ATP synthesis within Opa[1] null mouse embryonic fibroblasts (MEFs), independent of their ability to rescue mitochondrial morphology, suggesting that functional fusion alone is not essential for OPA1mediated regulation of energy output.[13]. Reduced OPA1 function has been shown to increase cellular susceptibility to apoptotic stimuli,[12,20] predisposing cells to intrinsic and extrinsic toxic insults

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