Abstract

BackgroundNR2E3 (PNR) is an orphan nuclear receptor essential for proper photoreceptor determination and differentiation. In humans, mutations in NR2E3 have been associated with the recessively inherited enhanced short wavelength sensitive (S-) cone syndrome (ESCS) and, more recently, with autosomal dominant retinitis pigmentosa (adRP). NR2E3 acts as a suppressor of the cone generation program in late mitotic retinal progenitor cells. In adult rod photoreceptors, NR2E3 represses cone-specific gene expression and acts in concert with the transcription factors CRX and NRL to activate rod-specific genes. NR2E3 and CRX have been shown to physically interact in vitro through their respective DNA-binding domains (DBD). The DBD also contributes to homo- and heterodimerization of nuclear receptors.Methodology/Principal FindingsWe analyzed NR2E3 homodimerization and NR2E3/CRX complex formation in an in vivo situation by Bioluminescence Resonance Energy Transfer (BRET2). NR2E3 wild-type protein formed homodimers in transiently transfected HEK293T cells. NR2E3 homodimerization was impaired in presence of disease-causing mutations in the DBD, except for the p.R76Q and p.R104W mutant proteins. Strikingly, the adRP-linked p.G56R mutant protein interacted with CRX with a similar efficiency to that of NR2E3 wild-type and p.R311Q proteins. In contrast, all other NR2E3 DBD-mutant proteins did not interact with CRX. The p.G56R mutant protein was also more effective in abolishing the potentiation of rhodospin gene transactivation by the NR2E3 wild-type protein. In addition, the p.G56R mutant enhanced the transrepression of the M- and S-opsin promoter, while all other NR2E3 DBD-mutants did not.Conclusions/SignificanceThese results suggest different disease mechanisms in adRP- and ESCS-patients carrying NR2E3 mutations. Titration of CRX by the p.G56R mutant protein acting as a repressor in trans may account for the severe clinical phenotype in adRP patients.

Highlights

  • NR2E3 (MIM#604485) is a photoreceptor-specific nuclear receptor (PNR) that belongs to the nuclear hormone receptor superfamily of ligand-modulated transcription factors [1]

  • Because NR2E3 had been shown to interact with cone-rod homeobox (CRX) [4], we hypothesized that the transcriptional activity of CRX could be affected by structural changes present in NR2E3 mutant proteins

  • We evaluated by Bioluminescence Resonance Energy Transfer 2 (BRET2) analysis whether NR2E3 DNA-binding domains (DBD) mutations affected the interaction with CRX causing phenotypic variations among patients

Read more

Summary

Introduction

NR2E3 (MIM#604485) is a photoreceptor-specific nuclear receptor (PNR) that belongs to the nuclear hormone receptor superfamily of ligand-modulated transcription factors [1]. In late mitotic retinal progenitor cells, NR2E3 is thought to suppress the cone generation program [3], while in adult differentiated rods, NR2E3 exerts a dual function by repressing cone-specific genes [4,5] and by activating several rod-specific genes, including rhodopsin [4,6,7]. The most conserved C domain forms a 70-amino acid long DNA-binding domain (DBD) consisting of two Cys zinc fingers. The C-terminal E/F domain, or LBD, consists of a conserved secondary structure formed by 12 a-helixes, containing a strong dimerization function and the ligand-dependent AF-2 transactivation function. NR2E3 represses cone-specific gene expression and acts in concert with the transcription factors CRX and NRL to activate rod-specific genes. The DBD contributes to homo- and heterodimerization of nuclear receptors

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