6 research outputs found

    Data on the generation of two Nr2e3 mouse models by CRISPR / Cas9D10A nickase

    Get PDF
    NR2E3 encodes an orphan nuclear receptor that plays a dual function as both transcriptional activator and repressor in photoreceptors, being necessary for cone fate inhibition as well as rod differentiation and homeostasis. Mutations in this gene cause retinitis pigmentosa (RP), enhanced S cone syndrome (ESCS) and Goldmann-Favre syndrome (GFS). There is one reported Nr2e3 isoform that contains all 8 exons and a second -previously unreported- shorter isoform, which only spans the first 7 exons and whose function is still unknown. In this data article, we designed and generated two new mouse models by targeting exon 8 of Nr2e3 using the CRISPR/Cas9-D10A nickase in order to dissect the role of the two isoforms in Nr2e3 function and elucidate the different disease mechanisms caused by NR2E3 mutations. This strategy generated several modified alleles that altered the coding sequence of the last exon thereby affecting functional domains of the transcription factor. Allele Δ27 is an in-frame deletion of 27 bp that ablates the dimerization domain, whereas allele ΔE8 (full deletion of exon 8), produces only the short isoform that lacks the dimerization and repressor domains. Morphological and functional alterations of both Δ27 and ΔE8 mutants are reported in the associated research article "Nr2e3 functional domain ablation by CRISPR-Cas9D10A identifies a new isoform and generated Retinitis Pigmentosa and Enhanced S-cone Syndrome models" (Aísa-Marín et al., 2020)

    The Alter Retina: Alternative Splicing of Retinal Genes in Health and Disease.

    Get PDF
    Alternative splicing of mRNA is an essential mechanism to regulate and increase the diversity of the transcriptome and proteome. Alternative splicing frequently occurs in a tissueor time-specific manner, contributing to differential gene expression between cell types during development. Neural tissues present extremely complex splicing programs and display the highest number of alternative splicing events. As an extension of the central nervous system, the retina constitutes an excellent system to illustrate the high diversity of neural transcripts. The retina expresses retinal specific splicing factors and produces a large number of alternative transcripts, including exclusive tissue-specific exons, which require an exquisite regulation. In fact, a current challenge in the genetic diagnosis of inherited retinal diseases stems from the lack of information regarding alternative splicing of retinal genes, as a considerable percentage of mutations alter splicing or the relative production of alternative transcripts. Modulation of alternative splicing in the retina is also instrumental in the design of novel therapeutic approaches for retinal dystrophies, since it enables precision medicine for specific mutations

    Specific photoreceptor cell fate pathways are differentially altered in NR2E3-associated diseases

    Full text link
    Mutations in NR2E3, a gene encoding an orphan nuclear transcription factor, cause two retinal dystrophies with a distinct phenotype, but the precise role of NR2E3 in rod and cone transcriptional networks remains unclear. To dissect NR2E3 function, we performed scRNA-seq in the retinas of wildtype and two different Nr2e3 mouse models that show phenotypes similar to patients carrying NR2E3 mutations. Our results reveal that rod and cone populations are not homogeneous and can be separated into different sub-classes. We identify a previously unreported cone pathway that generates hybrid cones co-expressing both cone- and rod-related genes. In mutant retinas, this hybrid cone subpopulation is more abundant and includes a subpopulation of rods transitioning towards a cone cell fate. Hybrid photoreceptors with high misexpression of cone- and rod-related genes are prone to regulated necrosis. Overall, our results shed light on the role of NR2E3 in modulating photoreceptor differentiation towards cone and rod fates and explain how different mutations in NR2E3 lead to distinct visual disorders in humans

    Exploring the role of NR2E3 in photoreceptor cell fate determination

    Get PDF
    [eng] The transcription factor NR2E3 participates in photoreceptor cell fate determination as it functions as both transcriptional repressor of cone genes and activator of rod genes. Mutations in NR2E3 cause retinitis pigmentosa (RP) and enhanced S-cone syndrome (ESCS) in humans. This gene produces a large isoform encoded in 8 exons and a previously unreported shorter isoform of 7 exons, whose function is unknown. We generated two mouse models by targeting exon 8 of Nr2e3 using CRISPR/Cas9- D10A nickase. Allele Δ27 is an in-frame deletion of 27 bp that ablates the dimerization domain H10, whereas allele ΔE8 (full deletion of exon 8) produces only the short isoform, which lacks the C-terminal part of the ligand binding domain (LBD) that encodes both H10 and the AF2 domain involved in the Nr2e3 repressor activity. The Δ27 mutant shows developmental alterations and a non-progressive retinal dysfunction that resembles the ESCS patient’s phenotype. Early ∆27 affectation is caused by alterations in rod and cone differentiation, but expression of ∆27 long isoform in the adult retina slows down the degenerative process. On the contrary, the ΔE8 mutant exhibits progressive retinal degeneration, with a milder phenotype at early stages and severe dysfunction at old stages, as occurs in human RP patients. scRNA- seq of wildtype and Nr2e3 mutant retinas reveals that rod and cone populations are not homogeneous, and identifies a previously unreported pathway of cone differentiation, in which a subpopulation of cones displays traits of a hybrid intermediate stage of differentiation and co-expresses cone and rod genes. This subpopulation of mis-differentiated cones is highly increased in the Nr2e3 mutant retinas as well as a subpopulation of rods transitioning towards the cone fate. Hybrid photoreceptors with high mis-expression of both cone and rod genes are prone to regulated necrosis. Our findings suggest that the NR2E3 short isoform regulates the long isoform by competing for the binding to target sites and partners of the NR2E3 regulatory complex, as well as by reducing NR2E3 long isoform stability. Overall, our results (i) propose a role for NR2E3 as a cone-patterning and differentiation regulator, (ii) shed light on the role of NR2E3 in the modulation of the photoreceptor precursor differentiation towards cone and rod fates, and (iii) explain why mutations in NR2E3 lead to different visual disorders in humans, providing valuable models for studying mechanisms of NR2E3-associated retinal dystrophies and evaluating potential therapies

    Nr2e3 functional domain ablation by CRISPR-Cas9D10A identifies a new isoform and generates retinitis pigmentosa and enhanced S-cone syndrome models

    Get PDF
    Mutations in NR2E3 cause retinitis pigmentosa (RP) and enhanced S-cone syndrome (ESCS) in humans. This gene produces a large isoform encoded in 8 exons and a previously unreported shorter isoform of 7 exons, whose function is unknown. We generated two mouse models by targeting exon 8 of Nr2e3 using CRISPR/Cas9-D10A nickase. Allele Δ27 is an in-frame deletion of 27 bp that ablates the dimerization domain H10, whereas allele ΔE8 (full deletion of exon 8) produces only the short isoform, which lacks the C-terminal part of the ligand binding domain (LBD) that encodes both H10 and the AF2 domain involved in the Nr2e3 repressor activity. The Δ27 mutant shows developmental alterations and a non-progressive electrophysiological dysfunction that resembles the ESCS phenotype. The ΔE8 mutant exhibits progressive retinal degeneration, as occurs in human RP patients. Our mutants suggest a role for Nr2e3 as a cone-patterning regulator and provide valuable models for studying mechanisms of NR2E3-associated retinal dystrophies and evaluating potential therapies
    corecore