Abstract

Dominant mutations occurring in the high-affinity Ca2+-binding sites (EF-hands) of the GUCA1A gene encoding guanylate cyclase-activating protein 1 (GCAP1) cause slowly progressing cone-rod dystrophy (CORD) in a dozen families worldwide. We developed a nonallele-specific adeno-associated virus (AAV)-based RNAi knockdown strategy to rescue the retina degeneration caused by GCAP1 mutations. We generated three genomic transgenic mouse lines expressing wildtype (WT) and L151F mutant mouse GCAP1 with or without a C-terminal GFP fusion. Under control of endogenous regulatory elements, the transgenes were expressed specifically in mouse photoreceptors. GCAP1(L151F) and GCAP1(L151F)-GFP transgenic mice presented with a late onset and slowly progressive photoreceptor degeneration, similar to that observed in human GCAP1-CORD patients. Transgenic expression of WT GCAP1-EGFP in photoreceptors had no adverse effect. Toward therapy development, a highly effective anti-mGCAP1 shRNA, mG1hp4, was selected from four candidate shRNAs using an in-vitro screening assay. Subsequently a self-complementary (sc) AAV serotype 2/8 expressing mG1hp4 was delivered subretinally to GCAP1(L151F)-GFP transgenic mice. Knockdown of the GCAP1(L151F)-GFP transgene product was visualized by fluorescence live imaging in the scAAV2/8-mG1hp4-treated retinas. Concomitant with the mutant GCAP1-GFP fusion protein, endogenous GCAP1 decreased as well in treated retinas. We propose nonallele-specific RNAi knockdown of GCAP1 as a general therapeutic strategy to rescue any GCAP1-based dominant cone-rod dystrophy in human patients.

Highlights

  • Cone-rod dystrophy (CORD, with a prevalence of 1/40,000) is a rare, highly heterogeneous class of hereditary retinal disease inherited in a dominant, recessive or X-linked fashion [1]

  • A 14,832 bp mouse guanylate cyclase-activating protein 1 (GCAP1) genomic sequence was modified to generate three transgenes which expressed either wildtype GCAP1 fused to EGFP (G1-GFP), or one of two mutant proteins, G1(L151F) and G1(L151F)-GFP

  • The L151F mutation was introduced in exon 4 to establish two GCAP1 transgenic mouse lines, G1(L151F) (Fig. 2A) and G1(L151F)-GFP (Fig. 2B)

Read more

Summary

Introduction

Cone-rod dystrophy (CORD, with a prevalence of 1/40,000) is a rare, highly heterogeneous class of hereditary retinal disease inherited in a dominant, recessive or X-linked fashion [1]. The disease manifests with photoaversion, reduced central visual acuity, achromatopsia at early stages, and eventually loss of peripheral vision attributed to progressive loss of first cone and rod photoreceptors. 27 genes have been linked to cone-rod dystrophy; of these, ten genes are associated with dominant CORD, 15 with recessive CORD, two are X-linked (RetNet, https://sph.uth.tmc.edu/retnet/). The protein products of these genes are involved in multiple aspects of photoreceptor structure and function [2]. One of the best characterized dominant CORD genes is GUCA1A encoding guanylate cyclase-activating protein 1 About one dozen families with more than 100 affected members have been identified to date [4]

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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