Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 16(4): e1008721, 2020 04.
Article in English | MEDLINE | ID: mdl-32339198

ABSTRACT

Current estimates suggest 50% of glaucoma blindness worldwide is caused by primary angle-closure glaucoma (PACG) but the causative gene is not known. We used genetic linkage and whole genome sequencing to identify Spermatogenesis Associated Protein 13, SPATA13 (NM_001166271; NP_001159743, SPATA13 isoform I), also known as ASEF2 (Adenomatous polyposis coli-stimulated guanine nucleotide exchange factor 2), as the causal gene for PACG in a large seven-generation white British family showing variable expression and incomplete penetrance. The 9 bp deletion, c.1432_1440del; p.478_480del was present in all affected individuals with angle-closure disease. We show ubiquitous expression of this transcript in cell lines derived from human tissues and in iris, retina, retinal pigment and ciliary epithelia, cornea and lens. We also identified eight additional mutations in SPATA13 in a cohort of 189 unrelated PACS/PAC/PACG samples. This gene encodes a 1277 residue protein which localises to the nucleus with partial co-localisation with nuclear speckles. In cells undergoing mitosis SPATA13 isoform I becomes part of the kinetochore complex co-localising with two kinetochore markers, polo like kinase 1 (PLK-1) and centrosome-associated protein E (CENP-E). The 9 bp deletion reported in this study increases the RAC1-dependent guanine nucleotide exchange factors (GEF) activity. The increase in GEF activity was also observed in three other variants identified in this study. Taken together, our data suggest that SPATA13 is involved in the regulation of mitosis and the mutations dysregulate GEF activity affecting homeostasis in tissues where it is highly expressed, influencing PACG pathogenesis.


Subject(s)
Glaucoma, Open-Angle/genetics , Guanine Nucleotide Exchange Factors/genetics , Mutation , Adolescent , Adult , Aged , Cell Division , Cell Nucleus/metabolism , Eye/metabolism , Female , Glaucoma, Open-Angle/pathology , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Humans , Kinetochores/metabolism , Male , Middle Aged , Pedigree , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport
2.
Hum Mol Genet ; 28(23): 3867-3879, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31807779

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs), more commonly referred to as Batten disease, are a group of inherited lysosomal storage disorders that present with neurodegeneration, loss of vision and premature death. There are at least 13 genetically distinct forms of NCL. Enzyme replacement therapies and pre-clinical studies on gene supplementation have shown promising results for NCLs caused by lysosomal enzyme deficiencies. The development of gene therapies targeting the brain for NCLs caused by defects in transmembrane proteins has been more challenging and only limited therapeutic effects in animal models have been achieved so far. Here, we describe the development of an adeno-associated virus (AAV)-mediated gene therapy to treat the neurodegeneration in a mouse model of CLN6 disease, a form of NCL with a deficiency in the membrane-bound protein CLN6. We show that neonatal bilateral intracerebroventricular injections with AAV9 carrying CLN6 increase lifespan by more than 90%, maintain motor skills and motor coordination and reduce neuropathological hallmarks of Cln6-deficient mice up to 23 months post vector administration. These data demonstrate that brain-directed gene therapy is a valid strategy to treat the neurodegeneration of CLN6 disease and may be applied to other forms of NCL caused by transmembrane protein deficiencies in the future.


Subject(s)
Genetic Vectors/administration & dosage , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Animals , Animals, Newborn , Brain/growth & development , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Humans , Injections, Intraventricular , Membrane Proteins/metabolism , Mice , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Treatment Outcome
3.
PLoS One ; 11(11): e0166397, 2016.
Article in English | MEDLINE | ID: mdl-27846257

ABSTRACT

PURPOSE: Mutations in the EYS gene are a common cause of autosomal recessive retinitis pigmentosa (arRP), yet the role of the EYS protein in humans is presently unclear. The aim of this study was to investigate the isoform structure, expression and potential function of EYS in the mammalian retina in order to better understand its involvement in the pathogenesis of arRP. METHODS: To achieve the objective, we examined the expression of mRNA transcripts of EYS isoforms in human tissues and cell lines by RT-PCR. We also investigated the localisation of EYS in cultured cells and retinal cryo-sections by confocal fluorescence microscopy and Western blot analysis. RESULTS: RT-PCR analysis confirmed that EYS has at least four isoforms. In addition to the previously reported EYS isoforms 1 and 4, we present the experimental validation of two smaller variants referred to as EYS isoforms 2 and 3. All four isoforms are expressed in the human retina and Y79 cells and the short variants were additionally detected in the testis. Immunofluorescent confocal microscopy and Western blot analysis revealed that all EYS isoforms preferentially localise to the cytoplasm of Y79 and HeLa cells. Moreover, an enrichment of the endogenous protein was observed near the centrosomes in Y79 cells. Interestingly, EYS was observed at the ciliary axoneme in Y79 ciliated cells. In macaque retinal cryosections, EYS was found to localise in the region of the photoreceptor ciliary axoneme in both rods and cones as well as in the cytoplasm of the ganglion cells. CONCLUSION: The results obtained in this study lead us to speculate that, in photoreceptor cells, EYS could be a protein involved in maintaining the stability of the ciliary axoneme in both rods and cones. The variability of its isoform structure suggests that other roles are also possible and yet to be established.


Subject(s)
Eye Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Animals , Axoneme/genetics , Axoneme/metabolism , Axoneme/pathology , Cilia/genetics , Cilia/metabolism , Cilia/pathology , Cytoplasm/genetics , Cytoplasm/metabolism , DNA Mutational Analysis , Electroretinography , Eye Proteins/metabolism , HeLa Cells , Humans , Indoles , Macaca/genetics , Macaca/metabolism , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retina/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
4.
Exp Eye Res ; 148: 24-29, 2016 07.
Article in English | MEDLINE | ID: mdl-27179412

ABSTRACT

VAX2 is a transcription factor specifically expressed in the ventral region of the prospective neural retina in vertebrates and is required for ventral eye specification. Despite its extensive analysis in vertebrates, the biological role of VAX2 in the human is presently unclear. This study was undertaken to investigate VAX2 in humans aiming to gain new knowledge into its involvement in retinal function. Here, we report VAX2 gene expression and protein localization in cultured cells and adult retina. RT-PCR experiments indicated that VAX2 is enriched in neuronal tissues. Moreover, we identified a novel isoform most abundantly expressed in the retina. We termed the known transcript (NM_012476) isoform-1, and the newly identified transcript as isoform-2. Analysis of protein localization in cultured cells revealed that isoform-1 localizes to the nucleus and isoform-2 is widely expressed within the cell; partial co-localization of isoform-2 and actin filaments was also observed. In nonhuman primate retina VAX2 was seen either in the nuclear or in the cytoplasmic compartment depending on the retinal cell type. In addition, a noteworthy enrichment of the signal was observed in the outer segment of cone photoreceptors. Overall, this study provides the first insights into the expression of VAX2 in humans and its localization in the adult primate retina. Moreover, preliminary characterization of alternative variants suggests an involvement of VAX2 in multiple cellular pathways. Our findings raise the interesting possibility for further investigation of VAX2 in the retina in health and disease.


Subject(s)
Homeodomain Proteins/metabolism , Retina/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytoskeleton/metabolism , Humans , Macaca fascicularis , Mice , Prospective Studies , Protein Isoforms/metabolism , Retinal Cone Photoreceptor Cells/metabolism
5.
PLoS One ; 11(2): e0148678, 2016.
Article in English | MEDLINE | ID: mdl-26872363

ABSTRACT

The significance of the ubiquitin-proteasome system (UPS) for protein degradation has been highlighted in the context of neurodegenerative diseases, including retinal dystrophies. TOPORS, a dual E3 ubiquitin and SUMO1 ligase, forms a component of the UPS and selected substrates for its enzymatic activities, such as DJ-1/PARK7 and APOBEC2, are important for neuronal as well as retinal homeostasis, respectively. TOPORS is ubiquitously expressed, yet its mutations are only known to result in autosomal dominant retinitis pigmentosa. We performed a yeast two-hybrid (Y2H) screen of a human retinal cDNA library in order to identify interacting protein partners of TOPORS from the retina, and thus begin delineating the putative disease mechanism(s) associated with the retina-specific phenotype resulting from mutations in TOPORS. The screen led to isolation of the 26 S protease regulatory subunit 4 (P26s4/ PSMC1), an ATPase indispensable for correct functioning of UPS-mediated proteostasis. The interaction between endogenous TOPORS and P26s4 proteins was validated by co-immuno-precipitation from mammalian cell extracts and further characterised by immunofluorescent co-localisation studies in cell lines and retinal sections. Findings from hTERT-RPE1 and 661W cells demonstrated that TOPORS and P26s4 co-localise at the centrosome in cultured cells. Immunofluorescent staining of mouse retinae revealed a strong P26s4 reactivity at the interface between retinal pigmented epithelium (RPE) layer and the photoreceptors outer segments (OS). This finding leads us to speculate that P26s4, along with TOPORS, may have a role(s) in RPE phagocytosis, in addition to contributing to the overall photoreceptor and retinal homeostasis via the UPS.


Subject(s)
Adenosine Triphosphatases/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Interaction Maps , Retina/metabolism , Ubiquitin-Protein Ligases/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/genetics , Animals , Cell Line , DNA, Complementary/genetics , Humans , Mice , Mutation , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Retina/cytology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/genetics
6.
Sci Rep ; 6: 19450, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26781568

ABSTRACT

PRPF31-associated retinitis pigmentosa presents a fascinating enigma: some mutation carriers are blind, while others are asymptomatic. We identify the major molecular cause of this incomplete penetrance through three cardinal features: (1) there is population variation in the number (3 or 4) of a minisatellite repeat element (MSR1) adjacent to the PRPF31 core promoter; (2) in vitro, 3-copies of the MSR1 element can repress gene transcription by 50 to 115-fold; (3) the higher-expressing 4-copy allele is not observed among symptomatic PRPF31 mutation carriers and correlates with the rate of asymptomatic carriers in different populations. Thus, a linked transcriptional modifier decreases PRPF31 gene expression that leads to haploinsufficiency. This result, taken with other identified risk alleles, allows precise genetic counseling for the first time. We also demonstrate that across the human genome, the presence of MSR1 repeats in the promoters or first introns of genes is associated with greater population variability in gene expression indicating that copy number variation of MSR1s is a generic controller of gene expression and promises to provide new insights into our understanding of gene expression regulation.


Subject(s)
Eye Proteins/metabolism , Gene Expression Regulation , Penetrance , Repetitive Sequences, Nucleic Acid , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Scavenger Receptors, Class A/genetics , Alleles , Binding Sites , Conserved Sequence , DNA Copy Number Variations , Gene Expression , Gene Frequency , Genes, Reporter , Genetics, Population , Genotype , Humans , Nucleotide Motifs , Phenotype , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Transcription, Genetic
7.
Ann Hum Genet ; 78(1): 62-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24116917

ABSTRACT

Mutations in PRPF31 are responsible for autosomal dominant retinitis pigmentosa (adRP, RP11 form) and affected families show nonpenetrance. Differential expression of the wildtype PRPF31 allele is responsible for this phenomenon: coinheritance of a mutation and a higher expressing wildtype allele provide protection against development of disease. It has been suggested that a major modulating factor lies in close proximity to the wildtype PRPF31 gene on Chromosome 19, implying that a cis-acting factor directly alters PRPF31 expression. Variable expression of CNOT3 is one determinant of PRPF31 expression. This study explored the relationship between CNOT3 (a trans-acting factor) and its paradoxical cis-acting nature in relation to RP11. Linkage analysis on Chromosome 19 was performed in mutation-carrying families, and the inheritance of the wildtype PRPF31 allele in symptomatic-asymptomatic sibships was assessed-confirming that differential inheritance of wildtype chromosome 19q13 determines the clinical phenotype (P < 2.6 × 10(-7) ). A theoretical model was constructed that explains the apparent conflict between the linkage data and the recent demonstration that a trans-acting factor (CNOT3) is a major nonpenetrance factor: we propose that this apparently cis-acting effect arises due to the intimate linkage of CNOT3 and PRPF31 on Chromosome 19q13-a novel mechanism that we have termed "linked trans-acting epistasis."


Subject(s)
Epistasis, Genetic , Eye Proteins/genetics , Genes, Recessive , Polymorphism, Genetic , Retinitis Pigmentosa/genetics , Transcription Factors/genetics , Alleles , Chromosomes, Human, Pair 19/genetics , Computational Biology , Eye Proteins/metabolism , Female , Genes, Dominant , Genetic Linkage , Genetic Loci , Heterozygote , Humans , Male , Microsatellite Repeats , Mutation , Pedigree , Pneumonia, Aspiration/genetics , Transcription Factors/metabolism
8.
J Mol Genet Med ; 7(2)2013 Aug.
Article in English | MEDLINE | ID: mdl-25729402

ABSTRACT

Mutations in PRPF31 have been implicated in retinitis pigmentosa, a blinding disease caused by degeneration of rod photoreceptors. The disease mechanism in the majority of cases is haploinsufficiency. Crucially, attempts at generation of animal models of disease have proved unsuccessful, yielding animals with a visual phenotype that does not mirror human disease. This suggests that, in these animals, the transcriptional regulation of PRPF31 is different to humans and compared to other species. Study of the evolution of the PRPF31 core promoter has important implications for our understanding of human disease, as disease phenotype is modified by differentially expressed alleles in the population. PRPF31 lies in a head-to-head arrangement with TFPT, a gene involved in cellular apoptosis. The two genes were shown to share common regulatory elements in the human genome. In this study, the core promoters of PRPF31 and TFPT were characterised by dual-luciferase reporter assay using genomic DNA from the green monkey, domestic dog and house mouse. It was found that the core promoters were conserved between human and monkey. In dog, the TFPT core promoter was conserved, but different PRPF31 gene architecture meant the gene was controlled by a long-range promoter lying some 2000bp from the transcription start site. There was very low level of conservation (<20%) of the PRPF31 5' region between mouse and human. It was shown that mouse populations did not show variable Prpf31 expression levels, revealing a potential explanation for the lack of phenotype observed in the Prpf31 knock-out mouse model.

9.
PLoS Genet ; 8(11): e1003040, 2012.
Article in English | MEDLINE | ID: mdl-23144630

ABSTRACT

Heterozygous mutations in the PRPF31 gene cause autosomal dominant retinitis pigmentosa (adRP), a hereditary disorder leading to progressive blindness. In some cases, such mutations display incomplete penetrance, implying that certain carriers develop retinal degeneration while others have no symptoms at all. Asymptomatic carriers are protected from the disease by a higher than average expression of the PRPF31 allele that is not mutated, mainly through the action of an unknown modifier gene mapping to chromosome 19q13.4. We investigated a large family with adRP segregating an 11-bp deletion in PRPF31. The analysis of cell lines derived from asymptomatic and affected individuals revealed that the expression of only one gene among a number of candidates within the 19q13.4 interval significantly correlated with that of PRPF31, both at the mRNA and protein levels, and according to an inverse relationship. This gene was CNOT3, encoding a subunit of the Ccr4-not transcription complex. In cultured cells, siRNA-mediated silencing of CNOT3 provoked an increase in PRPF31 expression, confirming a repressive nature of CNOT3 on PRPF31. Furthermore, chromatin immunoprecipitation revealed that CNOT3 directly binds to a specific PRPF31 promoter sequence, while next-generation sequencing of the CNOT3 genomic region indicated that its variable expression is associated with a common intronic SNP. In conclusion, we identify CNOT3 as the main modifier gene determining penetrance of PRPF31 mutations, via a mechanism of transcriptional repression. In asymptomatic carriers CNOT3 is expressed at low levels, allowing higher amounts of wild-type PRPF31 transcripts to be produced and preventing manifestation of retinal degeneration.


Subject(s)
Eye Proteins/genetics , Penetrance , Retinitis Pigmentosa , Transcription Factors/genetics , Eye Proteins/metabolism , Gene Expression Regulation , Heterozygote , Humans , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , RNA, Small Interfering , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Sequence Deletion , Transcription Factors/metabolism
10.
Hum Mol Genet ; 21(18): 4126-37, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22723017

ABSTRACT

PRPF31, a gene located at chromosome 19q13.4, encodes the ubiquitous splicing factor PRPF31. The gene lies in a head-to-head arrangement with TFPT, a poorly characterized gene with a role in cellular apoptosis. Mutations in PRPF31 have been implicated in autosomal dominant retinitis pigmentosa (adRP), a frequent and important cause of blindness worldwide. Disease associated with PRPF31 mutations is unusual, in that there is often non-penetrance of the disease phenotype in affected families, caused by differential expression of PRPF31. This study aimed to characterize the basic promoter elements of PRPF31 and TFPT. Luciferase reporter constructs were made, using genomic DNA from an asymptomatic individual with a heterozygous deletion of the entire putative promoter region. Fragments were tested by the dual-luciferase reporter assay in HeLa and RPE-1 cell lines. A comparison was made between the promoter regions of symptomatic and asymptomatic mutation-carrying individuals. A patient (CAN493) with adRP was identified, harbouring a regulatory region mutation; both alleles were assayed by the dual-luciferase reporter assay. Luciferase assays led to the identification of core promoters for both PRPF31 and TFPT; despite their shared gene architecture, the two genes appear to be controlled by slightly different regulatory regions. One functional polymorphism was identified in the PRPF31 promoter that increased transcriptional activation. The change was not, however, consistent with the observed symptomatic-asymptomatic phenotypes in a family affected by PRPF31-adRP. Analysis of the mutant promoter fragment from CAN493 showed a >50% reduction in promoter activity, suggesting a disease mechanism of functional haploinsufficiency-the first report of this disease mechanism in adRP.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Eye Proteins/genetics , Gene Expression Regulation , Retinitis Pigmentosa/genetics , Transcription, Genetic , Aged , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Case-Control Studies , Cloning, Molecular , Conserved Sequence , DNA Mutational Analysis , Eye Proteins/metabolism , Female , Genes, Dominant , Genes, Reporter , Genetic Association Studies , HeLa Cells , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Male , Molecular Sequence Data , Phylogeny , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Sequence Deletion , Statistics, Nonparametric
11.
Hum Mol Genet ; 20(5): 975-87, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21159800

ABSTRACT

We recently reported that mutations in the widely expressed nuclear protein TOPORS (topoisomerase I-binding arginine/serine rich) are associated with autosomal dominant retinal degeneration. However, the precise localization and a functional role of TOPORS in the retina remain unknown. Here, we demonstrate that TOPORS is a novel component of the photoreceptor sensory cilium, which is a modified primary cilium involved with polarized trafficking of proteins. In photoreceptors, TOPORS localizes primarily to the basal bodies of connecting cilium and in the centrosomes of cultured cells. Morpholino-mediated silencing of topors in zebrafish embryos demonstrates in another species a comparable retinal problem as seen in humans, resulting in defective retinal development and failure to form outer segments. These defects can be rescued by mRNA encoding human TOPORS. Taken together, our data suggest that TOPORS may play a key role in regulating primary cilia-dependent photoreceptor development and function. Additionally, it is well known that mutations in other ciliary proteins cause retinal degeneration, which may explain why mutations in TOPORS result in the same phenotype.


Subject(s)
Centrosome/metabolism , Cilia/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Retinal Degeneration/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cells, Cultured , Cilia/genetics , Humans , Mice , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Photoreceptor Cells/metabolism , Protein Transport , Retina/metabolism , Retinal Degeneration/genetics , Ubiquitin-Protein Ligases/genetics , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...