Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Biol ; 20(6): e3001649, 2022 06.
Article in English | MEDLINE | ID: mdl-35709082

ABSTRACT

Inherited retinal degeneration due to loss of photoreceptor cells is a leading cause of human blindness. These cells possess a photosensitive outer segment linked to the cell body through the connecting cilium (CC). While structural defects of the CC have been associated with retinal degeneration, its nanoscale molecular composition, assembly, and function are barely known. Here, using expansion microscopy and electron microscopy, we reveal the molecular architecture of the CC and demonstrate that microtubules are linked together by a CC inner scaffold containing POC5, CENTRIN, and FAM161A. Dissecting CC inner scaffold assembly during photoreceptor development in mouse revealed that it acts as a structural zipper, progressively bridging microtubule doublets and straightening the CC. Furthermore, we show that Fam161a disruption in mouse leads to specific CC inner scaffold loss and triggers microtubule doublet spreading, prior to outer segment collapse and photoreceptor degeneration, suggesting a molecular mechanism for a subtype of retinitis pigmentosa.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Animals , Cilia , Eye Proteins , Mice , Microtubules
2.
Mol Ther ; 31(10): 2948-2961, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37580905

ABSTRACT

Photoreceptor cell degeneration and death is the major hallmark of a wide group of human blinding diseases including age-related macular degeneration and inherited retinal diseases such as retinitis pigmentosa. In recent years, inherited retinal diseases have become the "testing ground" for novel therapeutic modalities, including gene and cell-based therapies. Currently there is no available treatment for retinitis pigmentosa caused by FAM161A biallelic pathogenic variants. In this study, we injected an adeno-associated virus encoding for the longer transcript of mFam161a into the subretinal space of P24-P29 Fam161a knockout mice to characterize the safety and efficacy of gene augmentation therapy. Serial in vivo assessment of retinal function and structure at 3, 6, and 8 months of age using the optomotor response test, full-field electroretinography, fundus autofluorescence, and optical coherence tomography imaging as well as ex vivo quantitative histology and immunohistochemical studies revealed a significant structural and functional rescue effect in treated eyes accompanied by expression of the FAM161A protein in photoreceptors. The results of this study may serve as an important step toward future application of gene augmentation therapy in FAM161A-deficient patients by identifying a promising isoform to rescue photoreceptors and their function.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Humans , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Retinal Degeneration/pathology , Mice, Knockout , Eye Proteins/genetics , Eye Proteins/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Electroretinography
3.
Proc Natl Acad Sci U S A ; 117(28): 16292-16301, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601208

ABSTRACT

Notch pathway signaling is implicated in several human cancers. Aberrant activation and mutations of Notch signaling components are linked to tumor initiation, maintenance, and resistance to cancer therapy. Several strategies, such as monoclonal antibodies against Notch ligands and receptors, as well as small-molecule γ-secretase inhibitors (GSIs), have been developed to interfere with Notch receptor activation at proximal points in the pathway. However, the use of drug-like small molecules to target the downstream mediators of Notch signaling, the Notch transcription activation complex, remains largely unexplored. Here, we report the discovery of an orally active small-molecule inhibitor (termed CB-103) of the Notch transcription activation complex. We show that CB-103 inhibits Notch signaling in primary human T cell acute lymphoblastic leukemia and other Notch-dependent human tumor cell lines, and concomitantly induces cell cycle arrest and apoptosis, thereby impairing proliferation, including in GSI-resistant human tumor cell lines with chromosomal translocations and rearrangements in Notch genes. CB-103 produces Notch loss-of-function phenotypes in flies and mice and inhibits the growth of human breast cancer and leukemia xenografts, notably without causing the dose-limiting intestinal toxicity associated with other Notch inhibitors. Thus, we describe a pharmacological strategy that interferes with Notch signaling by disrupting the Notch transcription complex and shows therapeutic potential for treating Notch-driven cancers.


Subject(s)
Receptors, Notch/metabolism , Small Molecule Libraries/pharmacology , Transcriptional Activation/drug effects , Animals , Apoptosis/drug effects , Binding Sites , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drosophila , Drug Resistance, Neoplasm/drug effects , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice , Mutation , Phenotype , Protein Multimerization , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
4.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502238

ABSTRACT

Inherited retinal dystrophies (IRD) are due to various gene mutations. Each mutated gene instigates a specific cell homeostasis disruption, leading to a modification in gene expression and retinal degeneration. We previously demonstrated that the polycomb-repressive complex-1 (PRC1) markedly contributes to the cell death process. To better understand these mechanisms, we herein study the role of PRC2, specifically EZH2, which often initiates the gene inhibition by PRC1. We observed that the epigenetic mark H3K27me3 generated by EZH2 was progressively and strongly expressed in some individual photoreceptors and that the H3K27me3-positive cell number increased before cell death. H3K27me3 accumulation occurs between early (accumulation of cGMP) and late (CDK4 expression) events of retinal degeneration. EZH2 hyperactivity was observed in four recessive and two dominant mouse models of retinal degeneration, as well as two dog models and one IRD patient. Acute pharmacological EZH2 inhibition by intravitreal injection decreased the appearance of H3K27me3 marks and the number of TUNEL-positive cells revealing that EZH2 contributes to the cell death process. Finally, we observed that the absence of the H3K27me3 mark is a biomarker of gene therapy treatment efficacy in XLRPA2 dog model. PRC2 and PRC1 are therefore important actors in the degenerative process of multiple forms of IRD.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Eye Proteins/physiology , Polycomb Repressive Complex 1/physiology , Proto-Oncogene Proteins/physiology , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Animals , DNA Methylation , Dogs , Enhancer of Zeste Homolog 2 Protein/genetics , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/metabolism
5.
Int J Mol Sci ; 22(17)2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34502527

ABSTRACT

Glucocorticoids are amongst the most used drugs to treat retinal diseases of various origins. Yet, the transcriptional regulations induced by glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation in retinal pigment epithelium cells (RPE) that form the outer blood-retina barrier are unknown. Levels of endogenous corticoids, ligands for MR and GR, were measured in human ocular media. Human RPE cells derived from induced pluripotent stem cells (iRPE) were used to analyze the pan-transcriptional regulations induced by aldosterone-an MR-specific agonist, or cortisol or cortisol + RU486-a GR antagonist. The retinal phenotype of transgenic mice that overexpress the human MR (P1.hMR) was analyzed. In the human eye, the main ligand for GR and MR is cortisol. The iRPE cells express functional GR and MR. The subset of genes regulated by aldosterone and by cortisol + RU-486, and not by cortisol alone, mimics an imbalance toward MR activation. They are involved in extracellular matrix remodeling (CNN1, MGP, AMTN), epithelial-mesenchymal transition, RPE cell proliferation and migration (ITGB3, PLAUR and FOSL1) and immune balance (TNFSF18 and PTX3). The P1.hMR mice showed choroidal vasodilation, focal alteration of the RPE/choroid interface and migration of RPE cells together with RPE barrier function alteration, similar to human retinal diseases within the pachychoroid spectrum. RPE is a corticosteroid-sensitive epithelium. MR pathway activation in the RPE regulates genes involved in barrier function, extracellular matrix, neural regulation and epithelial differentiation, which could contribute to retinal pathology.


Subject(s)
Aldosterone/metabolism , Hydrocortisone/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Retinal Diseases/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Epithelial-Mesenchymal Transition , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/pathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology
6.
Am J Hum Genet ; 99(3): 770-776, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27588451

ABSTRACT

Cone-rod degeneration (CRD) belongs to the disease spectrum of retinal degenerations, a group of hereditary disorders characterized by an extreme clinical and genetic heterogeneity. It mainly differentiates from other retinal dystrophies, and in particular from the more frequent disease retinitis pigmentosa, because cone photoreceptors degenerate at a higher rate than rod photoreceptors, causing severe deficiency of central vision. After exome analysis of a cohort of individuals with CRD, we identified biallelic mutations in the orphan gene CEP78 in three subjects from two families: one from Greece and another from Sweden. The Greek subject, from the island of Crete, was homozygous for the c.499+1G>T (IVS3+1G>T) mutation in intron 3. The Swedish subjects, two siblings, were compound heterozygotes for the nearby mutation c.499+5G>A (IVS3+5G>A) and for the frameshift-causing variant c.633delC (p.Trp212Glyfs(∗)18). In addition to CRD, these three individuals had hearing loss or hearing deficit. Immunostaining highlighted the presence of CEP78 in the inner segments of retinal photoreceptors, predominantly of cones, and at the base of the primary cilium of fibroblasts. Interaction studies also showed that CEP78 binds to FAM161A, another ciliary protein associated with retinal degeneration. Finally, analysis of skin fibroblasts derived from affected individuals revealed abnormal ciliary morphology, as compared to that of control cells. Altogether, our data strongly suggest that mutations in CEP78 cause a previously undescribed clinical entity of a ciliary nature characterized by blindness and deafness but clearly distinct from Usher syndrome, a condition for which visual impairment is due to retinitis pigmentosa.


Subject(s)
Cell Cycle Proteins/genetics , Cilia/pathology , Cone-Rod Dystrophies/complications , Cone-Rod Dystrophies/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Mutation/genetics , Aged , Alleles , Animals , Cadaver , Cell Cycle Proteins/metabolism , Cohort Studies , Cone-Rod Dystrophies/pathology , Cone-Rod Dystrophies/physiopathology , Exome/genetics , Eye/embryology , Eye/metabolism , Eye Proteins/metabolism , Female , Fibroblasts/pathology , Greece , Hearing Loss, Sensorineural/complications , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Homozygote , Humans , Introns/genetics , Male , Mice , Middle Aged , Pedigree , Protein Binding , RNA, Messenger/analysis , Sweden , Transcriptome , Usher Syndromes/pathology
7.
Hum Mol Genet ; 25(20): 4462-4472, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28172811

ABSTRACT

Cone photoreceptor cell death as it occurs in certain hereditary retinal diseases is devastating, with the affected patients suffering from a loss of accurate and colour vision. Regrettably, these hereditary cone diseases are still untreatable to date. Thus, the identification of substances able to block or restrain cone cell death is of primary importance. We studied the neuroprotective effects of a histone deacetylase inhibitor, Trichostatin A (TSA), in a mouse model of inherited, primary cone degeneration (cpfl1). We show that HDAC inhibition protects cpfl1 cones in vitro, in retinal explant cultures. More importantly, in vivo, a single intravitreal TSA injection significantly increased cone survival for up to 16 days post-injection. In addition, the abnormal, incomplete cone migration pattern in the cpfl1 retina was significantly improved by HDAC inhibition. These findings suggest a crucial role for HDAC activity in primary cone degeneration and highlight a new avenue for future therapy developments for cone dystrophies and retinal diseases associated with impaired cone migration.


Subject(s)
Disease Models, Animal , Hydroxamic Acids/pharmacology , Neuroprotective Agents/pharmacology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/drug therapy , Animals , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Mice , Neuroprotective Agents/therapeutic use
8.
Mol Ther ; 25(3): 634-653, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28143742

ABSTRACT

The cone function is essential to mediate high visual acuity, color vision, and daylight vision. Inherited cone dystrophies and age-related macular degeneration affect a substantial percentage of the world population. To identify and isolate the most competent cells for transplantation and integration into the retina, cone tracing during development would be an important added value. To that aim, the Chrnb4-EGFP mouse line was characterized throughout retinogenesis. It revealed a sub-population of early retinal progenitors expressing the reporter gene that is progressively restricted to mature cones during retina development. The presence of the native CHRNB4 protein was confirmed in EGFP-positive cells, and it presents a similar pattern in the human retina. Sub-retinal transplantations of distinct subpopulations of Chrnb4-EGFP-expressing cells revealed the embryonic day 15.5 high-EGFP population the most efficient cells to interact with host retinas to provoke the appearance of EGFP-positive cones in the photoreceptor layer. Importantly, transplantations into the DsRed retinas revealed material exchanges between donor and host retinas, as >80% of transplanted EGFP-positive cones also were DsRed positive. Whether this cell material fusion is of significant therapeutic advantage requires further thorough investigations. The Chrnb4-EGFP mouse line definitely opens new research perspectives in cone genesis and retina repair.


Subject(s)
Cell Tracking/methods , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Recombinant Fusion Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Animals , Humans , Macular Degeneration , Mice , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Retina/embryology , Retina/metabolism , Retinoid X Receptor gamma/genetics , Retinoid X Receptor gamma/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism
9.
Adv Exp Med Biol ; 1074: 359-365, 2018.
Article in English | MEDLINE | ID: mdl-29721964

ABSTRACT

Retinitis Pigmentosa (RP) is a class of hereditary retinal dystrophy associated with gradual visual failure and a subsequent loss of light-sensitive cells in the retina, leading to blindness. Many mutated genes were found to be causative of this disease. Despite a number of compiling efforts, the process of cell death in photoreceptors remains to be clearly elucidated. We recently reported an abnormal cell cycle reentry in photoreceptors undergoing degeneration in Rd1 mice, a model of RP, and identified the polycomb repressive complex 1 (PRC1) core component BMI1 as a critical molecular factor orchestrating the cell death mechanism. As the cell death rescue in Rd1;Bmi-1 KO mice was independent on the conventional Ink4a/Arf pathways, we now explored the structural properties of BMI1 in order to examine the differential expression of its posttranslational modifications in Rd1 retina. Our results suggest that BMI1 cell death induction in Rd1 is not related to its phosphorylation status. We therefore propose the epigenetic activity of BMI1 as an alternative route for BMI1-mediated toxicity in Rd1.


Subject(s)
Eye Proteins/physiology , Photoreceptor Cells, Vertebrate/pathology , Polycomb Repressive Complex 1/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Retinitis Pigmentosa/pathology , Animals , Apoptosis , Chromatin/chemistry , Chromatin/ultrastructure , DNA Fragmentation , DNA, Superhelical/chemistry , Disease Models, Animal , Eye Proteins/chemistry , Mice , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Necrosis , Phosphorylation , Photoreceptor Cells, Vertebrate/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/deficiency , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/physiology , Protein Folding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
10.
Hum Mol Genet ; 24(12): 3359-71, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25749990

ABSTRACT

Defects in FAM161A, a protein of unknown function localized at the cilium of retinal photoreceptor cells, cause retinitis pigmentosa, a form of hereditary blindness. By using different fragments of this protein as baits to screen cDNA libraries of human and bovine retinas, we defined a yeast two-hybrid-based FAM161A interactome, identifying 53 bona fide partners. In addition to statistically significant enrichment in ciliary proteins, as expected, this interactome revealed a substantial bias towards proteins from the Golgi apparatus, the centrosome and the microtubule network. Validation of interaction with key partners by co-immunoprecipitation and proximity ligation assay confirmed that FAM161A is a member of the recently recognized Golgi-centrosomal interactome, a network of proteins interconnecting Golgi maintenance, intracellular transport and centrosome organization. Notable FAM161A interactors included AKAP9, FIP3, GOLGA3, KIFC3, KLC2, PDE4DIP, NIN and TRIP11. Furthermore, analysis of FAM161A localization during the cell cycle revealed that this protein followed the centrosome during all stages of mitosis, likely reflecting a specific compartmentalization related to its role at the ciliary basal body during the G0 phase. Altogether, these findings suggest that FAM161A's activities are probably not limited to ciliary tasks but also extend to more general cellular functions, highlighting possible novel mechanisms for the molecular pathology of retinal disease.


Subject(s)
Centrosome/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Genes, Recessive , Golgi Apparatus/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Animals , Carrier Proteins , Cattle , Cell Line , Cytoskeleton/metabolism , Humans , Intracellular Space/metabolism , Protein Binding , Protein Interaction Mapping/methods , Protein Interaction Maps , Protein Transport , Two-Hybrid System Techniques
11.
J Pathol ; 238(2): 300-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26387748

ABSTRACT

Disease-causing variants of a large number of genes trigger inherited retinal degeneration leading to photoreceptor loss. Because cones are essential for daylight and central vision such as reading, mobility, and face recognition, this review focuses on a variety of animal models for cone diseases. The pertinence of using these models to reveal genotype/phenotype correlations and to evaluate new therapeutic strategies is discussed. Interestingly, several large animal models recapitulate human diseases and can serve as a strong base from which to study the biology of disease and to assess the scale-up of new therapies. Examples of innovative approaches will be presented such as lentiviral-based transgenesis in pigs and adeno-associated virus (AAV)-gene transfer into the monkey eye to investigate the neural circuitry plasticity of the visual system. The models reported herein permit the exploration of common mechanisms that exist between different species and the identification and highlighting of pathways that may be specific to primates, including humans.


Subject(s)
Blindness/genetics , Disease Models, Animal , Animals , Animals, Genetically Modified , Blindness/therapy , Dogs , Genetic Therapy , Humans , Mutation/genetics , Retinal Cone Photoreceptor Cells/physiology , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Rodentia , Saimiri , Sheep , Swine
12.
Adv Exp Med Biol ; 854: 371-7, 2016.
Article in English | MEDLINE | ID: mdl-26427434

ABSTRACT

During different forms of neurodegenerative diseases, including the retinal degeneration, several cell cycle proteins are expressed in the dying neurons from Drosophila to human revealing that these proteins are a hallmark of neuronal degeneration. This is true for animal models of Alzheimer's, and Parkinson's diseases, Amyotrophic Lateral Sclerosis and for Retinitis Pigmentosa as well as for acute injuries such as stroke and light damage. Longitudinal investigation and loss-of-function studies attest that cell cycle proteins participate to the process of cell death although with different impacts, depending on the disease. In the retina, inhibition of cell cycle protein action can result to massive protection. Nonetheless, the dissection of the molecular mechanisms of neuronal cell death is necessary to develop adapted therapeutic tools to efficiently protect photoreceptors as well as other neuron types.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Therapy/methods , Molecular Targeted Therapy/methods , Retinal Degeneration/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Division/drug effects , Humans , Neurons/drug effects , Neurons/metabolism , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism
13.
Proc Natl Acad Sci U S A ; 110(7): E593-601, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23359713

ABSTRACT

The epigenetic regulator Bmi1 controls proliferation in many organs. Reexpression of cell cycle proteins such as cyclin-dependent kinases (CDKs) is a hallmark of neuronal apoptosis in neurodegenerative diseases. Here we address the potential role of Bmi1 as a key regulator of cell cycle proteins during neuronal apoptosis. We show that several cell cycle proteins are expressed in different models of retinal degeneration and required in the Rd1 photoreceptor death process. Deleting E2f1, a downstream target of CDKs, provided temporary protection in Rd1 mice. Most importantly, genetic ablation of Bmi1 provided extensive photoreceptor survival and improvement of retinal function in Rd1 mice, mediated by a decrease in cell cycle markers and regulators independent of p16(Ink4a) and p19(Arf). These data reveal that Bmi1 controls the cell cycle-related death process, highlighting this pathway as a promising therapeutic target for neuroprotection in retinal dystrophies.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Retinal Degeneration/metabolism , Analysis of Variance , Animals , E2F1 Transcription Factor/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Histological Techniques , Mice , Mice, Knockout , Microscopy, Fluorescence , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics
14.
Hum Mol Genet ; 21(23): 5174-84, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22940612

ABSTRACT

Retinitis pigmentosa (RP) is a retinal degenerative disease characterized by the progressive loss of photoreceptors. We have previously demonstrated that RP can be caused by recessive mutations in the human FAM161A gene, encoding a protein with unknown function that contains a conserved region shared only with a distant paralog, FAM161B. In this study, we show that FAM161A localizes at the base of the photoreceptor connecting cilium in human, mouse and rat. Furthermore, it is also present at the ciliary basal body in ciliated mammalian cells, both in native conditions and upon the expression of recombinant tagged proteins. Yeast two-hybrid analysis of binary interactions between FAM161A and an array of ciliary and ciliopathy-associated proteins reveals direct interaction with lebercilin, CEP290, OFD1 and SDCCAG8, all involved in hereditary retinal degeneration. These interactions are mediated by the C-terminal moiety of FAM161A, as demonstrated by pull-down experiments in cultured cell lines and in bovine retinal extracts. As other ciliary proteins, FAM161A can also interact with the microtubules and organize itself into microtubule-dependent intracellular networks. Moreover, small interfering RNA-mediated depletion of FAM161A transcripts in cultured cells causes the reduction in assembled primary cilia. Taken together, these data indicate that FAM161A-associated RP can be considered as a novel retinal ciliopathy and that its molecular pathogenesis may be related to other ciliopathies.


Subject(s)
Eye Proteins/genetics , Photoreceptor Connecting Cilium/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Cilia/metabolism , Eye Proteins/metabolism , Gene Expression , Humans , Mice , Photoreceptor Cells, Vertebrate/metabolism , Protein Binding , Protein Transport , RNA Interference , Rats , Retina/metabolism , Retina/pathology
15.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504136

ABSTRACT

For 15 years, gene therapy has been viewed as a beacon of hope for inherited retinal diseases. Many preclinical investigations have centered around vectors with maximal gene expression capabilities, yet despite efficient gene transfer, minimal physiological improvements have been observed in various ciliopathies. Retinitis pigmentosa-type 28 (RP28) is the consequence of bi-allelic null mutations in the FAM161A, an essential protein for the structure of the photoreceptor connecting cilium (CC). In its absence, cilia become disorganized, leading to outer segment collapses and vision impairment. Within the human retina, FAM161A has two isoforms: the long one with exon 4, and the short one without it. To restore CC in Fam161a-deficient mice shortly after the onset of cilium disorganization, we compared AAV vectors with varying promoter activities, doses, and human isoforms. While all vectors improved cell survival, only the combination of both isoforms using the weak FCBR1-F0.4 promoter enabled precise FAM161A expression in the CC and enhanced retinal function. Our investigation into FAM161A gene replacement for RP28 emphasizes the importance of precise therapeutic gene regulation, appropriate vector dosing, and delivery of both isoforms. This precision is pivotal for secure gene therapy involving structural proteins like FAM161A.


Subject(s)
Retinitis Pigmentosa , Animals , Mice , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Exons , Protein Isoforms/genetics , Protein Isoforms/metabolism , Genetic Therapy , Eye Proteins/genetics , Eye Proteins/chemistry , Eye Proteins/metabolism
16.
Am J Hum Genet ; 87(3): 376-81, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20705278

ABSTRACT

Retinitis pigmentosa (RP) is a degenerative disease of the retina leading to progressive loss of vision and, in many instances, to legal blindness at the end stage. The RP28 locus was assigned in 1999 to the short arm of chromosome 2 by homozygosity mapping in a large Indian family segregating autosomal-recessive RP (arRP). Following a combined approach of chromatin immunoprecipitation and parallel sequencing of genomic DNA, we identified a gene, FAM161A, which was shown to carry a homozygous nonsense mutation (p.Arg229X) in patients from the original RP28 pedigree. Another homozygous FAM161A stop mutation (p.Arg437X) was detected in three subjects from a cohort of 118 apparently unrelated German RP patients. Age at disease onset in these patients was in the second to third decade, with severe visual handicap in the fifth decade and legal blindness in the sixth to seventh decades. FAM161A is a phylogenetically conserved gene, expressed in the retina at relatively high levels and encoding a putative 76 kDa protein of unknown function. In the mouse retina, Fam161a mRNA is developmentally regulated and controlled by the transcription factor Crx, as demonstrated by chromatin immunoprecipitation and organotypic reporter assays on explanted retinas. Fam161a protein localizes to photoreceptor cells during development, and in adult animals it is present in the inner segment as well as the outer plexiform layer of the retina, the synaptic interface between photoreceptors and their efferent neurons. Taken together, our data indicate that null mutations in FAM161A are responsible for the RP28-associated arRP.


Subject(s)
Codon, Nonsense/genetics , Eye Proteins/genetics , Genes, Recessive/genetics , Genetic Loci/genetics , Retinitis Pigmentosa/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , DNA Mutational Analysis , Eye Proteins/chemistry , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology
17.
J Gene Med ; 14(11): 632-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23080553

ABSTRACT

BACKGROUND: Strategies leading to the long-term suppression of inappropriate ocular angiogenesis are required to avoid the need for repetitive monthly injections for treatment of diseases of the eye, such as age-related macular degeneration (AMD). The present study aimed to develop a strategy for the sustained repression of vascular endothelial growth factor (VEGF), which is identified as the key player in exudative AMD. METHODS: We have employed short hairpin (sh)RNAs combined with adeno-associated virus (AAV) delivery to obtain the targeted expression of potent gene-regulatory molecules. Anti-VEGF shRNAs were analyzed in human retinal pigment epithelial (RPE) cells using Renilla luciferase screening. For in vivo delivery of the most potent shRNA, self-complementary AAV vectors were packaged in serotype 8 capsids (scAAV2/8-hU6-sh9). In vivo efficacy was evaluated either by injection of scAAV2/8-hU6-sh9 into murine hind limb muscles or in a laser-induced murine model of choroidal neovascularization (CNV) following scAAV2/8-hU6-sh9 subretinal delivery. RESULTS: Plasmids encoding anti-VEGF shRNAs showed efficient knockdown of human VEGF in RPEs. Intramuscular administration led to localized expression and 91% knockdown of endogenous murine (m)VEGF. Subsequently, the ability of AAV2/8-encoded shRNAs to impair vessel formation was evaluated in the murine model of CNV. In this model, the sizes of the CNV were significantly reduced (up to 48%) following scAAV2/8-hU6-sh9 subretinal delivery. CONCLUSIONS: Using anti-VEGF vectors, we have demonstrated efficient silencing of endogenous mVEGF and showed that subretinal administration of scAAV2/8-hU6-sh9 has the ability to impair vessel formation in an AMD animal model. Thus, AAV-encoded shRNA can be used for the inhibition of neovascularization, leading to the development of sustained anti-VEGF therapy.


Subject(s)
Choroidal Neovascularization/genetics , Dependovirus/genetics , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line , Choroidal Neovascularization/metabolism , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/metabolism
18.
Pharmaceutics ; 14(8)2022 Jul 31.
Article in English | MEDLINE | ID: mdl-36015231

ABSTRACT

This review offers the basics of lentiviral vector technologies, their advantages and pitfalls, and an overview of their use in the field of ophthalmology. First, the description of the global challenges encountered to develop safe and efficient lentiviral recombinant vectors for clinical application is provided. The risks and the measures taken to minimize secondary effects as well as new strategies using these vectors are also discussed. This review then focuses on lentiviral vectors specifically designed for ocular therapy and goes over preclinical and clinical studies describing their safety and efficacy. A therapeutic approach using lentiviral vector-mediated gene therapy is currently being developed for many ocular diseases, e.g., aged-related macular degeneration, retinopathy of prematurity, inherited retinal dystrophies (Leber congenital amaurosis type 2, Stargardt disease, Usher syndrome), glaucoma, and corneal fibrosis or engraftment rejection. In summary, this review shows how lentiviral vectors offer an interesting alternative for gene therapy in all ocular compartments.

19.
Hum Mol Genet ; 18(7): 1266-75, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19147682

ABSTRACT

RPE65 is a retinoid isomerase required for the production of 11-cis-retinal, the chromophore of both cone and rod visual pigments. We recently established an R91W knock-in mouse strain as homologous animal model for patients afflicted by this mutation in RPE65. These mice have impaired vision and can only synthesize minute amounts of 11-cis-retinal. Here, we investigated the consequences of this chromophore insufficiency on cone function and pathophysiology. We found that the R91W mutation caused cone opsin mislocalization and progressive geographic cone atrophy. Remnant visual function was mostly mediated by rods. Ablation of rod opsin corrected the localization of cone opsin and improved cone retinal function. Thus, our analyses indicate that under conditions of limited chromophore supply rods and cones compete for 11-cis-retinal that derives from regeneration pathway(s) which are reliant on RPE65. Due to their higher number and the instability of cone opsin, rods are privileged under this condition while cones suffer chromophore deficiency and degenerate. These findings reinforce the notion that in patients any effective gene therapy with RPE65 needs to target the cone-rich macula directly to locally restore the cones' chromophore supply outside the reach of rods.


Subject(s)
Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Pigments/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinaldehyde/metabolism , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Cell Death , Cone Opsins/metabolism , Eye Proteins/genetics , Humans , Mice , Mutation/genetics , Protein Transport , cis-trans-Isomerases
20.
Sci Rep ; 11(1): 2030, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479377

ABSTRACT

FAM161A mutations are the most common cause of inherited retinal degenerations in Israel. We generated a knockout (KO) mouse model, Fam161atm1b/tm1b, lacking the major exon #3 which was replaced by a construct that include LacZ under the expression of the Fam161a promoter. LacZ staining was evident in ganglion cells, inner and outer nuclear layers and inner and outer-segments of photoreceptors in KO mice. No immunofluorescence staining of Fam161a was evident in the KO retina. Visual acuity and electroretinographic (ERG) responses showed a gradual decrease between the ages of 1 and 8 months. Optical coherence tomography (OCT) showed thinning of the whole retina. Hypoautofluorescence and hyperautofluorescence pigments was observed in retinas of older mice. Histological analysis revealed a progressive degeneration of photoreceptors along time and high-resolution transmission electron microscopy (TEM) analysis showed that photoreceptor outer segment disks were disorganized in a perpendicular orientation and outer segment base was wider and shorter than in WT mice. Molecular degenerative markers, such as microglia and CALPAIN-2, appear already in a 1-month old KO retina. These results indicate that a homozygous Fam161a frameshift mutation affects retinal function and causes retinal degeneration. This model will be used for gene therapy treatment in the future.


Subject(s)
Calpain/genetics , Eye Proteins/genetics , Retina/diagnostic imaging , Retinal Degeneration/genetics , Animals , Disease Models, Animal , Electroretinography , Frameshift Mutation/genetics , Humans , Lac Operon/genetics , Mice , Mice, Knockout , Retina/pathology , Retinal Degeneration/diagnostic imaging , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Tomography, Optical Coherence , Visual Acuity/genetics
SELECTION OF CITATIONS
SEARCH DETAIL