Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 74
1.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Article En | MEDLINE | ID: mdl-38504136

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.


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
2.
Mol Ther ; 31(10): 2948-2961, 2023 10 04.
Article En | MEDLINE | ID: mdl-37580905

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.


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.
Pharmaceutics ; 14(8)2022 Jul 31.
Article En | MEDLINE | ID: mdl-36015231

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.

4.
PLoS Biol ; 20(6): e3001649, 2022 06.
Article En | MEDLINE | ID: mdl-35709082

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.


Retinal Degeneration , Retinitis Pigmentosa , Animals , Cilia , Eye Proteins , Mice , Microtubules
5.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article En | MEDLINE | ID: mdl-34502238

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.


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
6.
Int J Mol Sci ; 22(17)2021 Sep 05.
Article En | MEDLINE | ID: mdl-34502527

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.


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
7.
Sci Rep ; 11(1): 2030, 2021 01 21.
Article En | MEDLINE | ID: mdl-33479377

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.


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
8.
Front Neurosci ; 14: 571293, 2020.
Article En | MEDLINE | ID: mdl-33324144

PURPOSE: was to create an in vitro model of human retinal detachment (RD) to study the mechanisms of photoreceptor death. METHODS: Human retinas were obtained through eye globe donations for research purposes and cultivated as explants. Cell death was investigated in retinas with (control) and without retinal pigment epithelium (RPE) cells to mimic RD. Tissues were studied at different time points and immunohistological analyses for TUNEL, Cleaved caspase3, AIF, CDK4 and the epigenetic mark H3K27me3 were performed. Human and monkey eye globes with retinal detachment served as controls. RESULTS: The number of TUNEL-positive cells, compared between 1 and 7 days, increased with time in both retinas with RPE (from 1.2 ± 0.46 to 8 ± 0.89, n = 4) and without RPE (from 2.6 ± 0.73 to 16.3 ± 1.27, p < 0.014). In the group without RPE, cell death peaked at day 3 (p = 0.014) and was high until day 7. Almost no Cleaved-Caspase3 signal was observed, whereas a transient augmentation at day 3 of AIF-positive cells was observed to be about 10-fold in comparison to the control group (n = 2). Few CDK4-positive cells were found in both groups, but significantly more in the RD group at day 7 (1.8 ± 0.24 vs. 4.7 ± 0.58, p = 0.014). The H3K27me3 mark increased by 7-fold after 5 days in the RD group (p = 0.014) and slightly decreased at day 7 and was also observed to be markedly increased in human and monkey detached retina samples. CONCLUSION: AIF expression coincides with the first peak of cell death, whereas the H3K27me3 mark increases during the cell death plateau, suggesting that photoreceptor death is induced by different successive pathways after RD. This in vitro model should permit the identification of neuroprotective drugs with clinical relevance.

9.
Proc Natl Acad Sci U S A ; 117(28): 16292-16301, 2020 07 14.
Article En | MEDLINE | ID: mdl-32601208

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.


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
10.
Sci Rep ; 10(1): 8890, 2020 06 01.
Article En | MEDLINE | ID: mdl-32483256

The retinal pigment epithelium (RPE) is a monolayer of cobblestone-like epithelial cells that accomplishes critical functions for the retina. Several protocols have been published to differentiate pluripotent stem cells into RPE cells suitable for disease modelling and therapy development. In our study, the RPE identity of human induced pluripotent stem cell (hiPSC)-derived RPE (iRPE) was extensively characterized, and then used to test a lentiviral-mediated RPE65 gene augmentation therapy. A dose study of the lentiviral vector revealed a dose-dependent effect of the vector on RPE65 mRNA levels. A marked increase of the RPE65 mRNA was also observed in the iRPE (100-fold) as well as in an experimental set with RPE derived from another hiPSC source and from foetal human RPE. Although iRPE displayed features close to bona fide RPE, no or a modest increase of the RPE65 protein level was observed depending on the protein detection method. Similar results were observed with the two other cell lines. The mechanism of RPE65 protein regulation remains to be elucidated, but the current work suggests that high vector expression will not produce an excess of the normal RPE65 protein level.


Induced Pluripotent Stem Cells/cytology , Retinal Pigment Epithelium/cytology , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Gene Transfer Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Lentivirus/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Up-Regulation
11.
Sci Rep ; 9(1): 13873, 2019 09 25.
Article En | MEDLINE | ID: mdl-31554875

Age Related Macular Degeneration (AMD) is the first cause of social blindness in people aged over 65 leading to atrophy of retinal pigment epithelial cells (RPE), photoreceptors and choroids, eventually associated with choroidal neovascularization. Accumulation of undigested cellular debris within RPE cells or under the RPE (Drusen), oxidative stress and inflammatory mediators contribute to the RPE cell death. The major risk to develop AMD is the Y402H polymorphism of complement factor H (CFH). CFH interacting with oxidized phospholipids on the RPE membrane modulates the functions of these cells, but the exact role of CFH in RPE cell death and survival remain poorly understood. The aim of this study was to analyze the potential protective mechanism of CFH on RPE cells submitted to oxidative stress. Upon exposure to oxidized lipids 4-HNE (4-hydroxy-2-nonenal) derived from photoreceptors, both the human RPE cell line ARPE-19 and RPE cells derived from human induced pluripotent stem cells were protected from death only in the presence of the full length human recombinant CFH in the culture medium. This protective effect was independent from the membrane attack complex (MAC) formation. CFH maintained RPE cells tight junctions' structure and regulated the caspase dependent apoptosis process. These results demonstrated the CFH anti-oxidative stress functions independently of its capacity to inhibit MAC formation.


Complement Factor H/pharmacology , Complement Membrane Attack Complex/drug effects , Retinal Pigment Epithelium/drug effects , Aldehydes/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Death/drug effects , Cell Line , Complement Membrane Attack Complex/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Microscopy, Electron, Transmission , Oxidative Stress/drug effects , Real-Time Polymerase Chain Reaction , Recombinant Proteins , Retinal Pigment Epithelium/metabolism , Tight Junctions/drug effects
12.
Mol Ther Methods Clin Dev ; 13: 1-13, 2019 Jun 14.
Article En | MEDLINE | ID: mdl-30581889

Hereditary hearing loss often results from mutation of genes expressed by cochlear hair cells. Gene addition using AAV vectors has shown some efficacy in mouse models, but clinical application requires two additional advances. First, new AAV capsids must mediate efficient transgene expression in both inner and outer hair cells of the cochlea. Second, to have the best chance of clinical translation, these new vectors must also transduce hair cells in non-human primates. Here, we show that an AAV9 capsid variant, PHP.B, produces efficient transgene expression of a GFP reporter in both inner and outer hair cells of neonatal mice. We show also that AAV9-PHP.B mediates almost complete transduction of inner and outer HCs in a non-human primate. In a mouse model of Usher syndrome type 3A deafness (gene CLRN1), we use AAV9-PHP.B encoding Clrn1 to partially rescue hearing. Thus, we have identified a vector with promise for clinical treatment of hereditary hearing disorders, and we demonstrate, for the first time, viral transduction of the inner ear of a primate with an AAV vector.

13.
Adv Exp Med Biol ; 1074: 359-365, 2018.
Article En | MEDLINE | ID: mdl-29721964

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.


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
14.
Sci Rep ; 7(1): 8834, 2017 08 18.
Article En | MEDLINE | ID: mdl-28821742

In diabetic retinopathy, the exact mechanisms leading to retinal capillary closure and to retinal barriers breakdown remain imperfectly understood. Rho-associated kinase (ROCK), an effector of the small GTPase Rho, involved in cytoskeleton dynamic regulation and cell polarity is activated by hyperglycemia. In one year-old Goto Kakizaki (GK) type 2 diabetic rats retina, ROCK-1 activation was assessed by its cellular distribution and by phosphorylation of its substrates, MYPT1 and MLC. In both GK rat and in human type 2 diabetic retinas, ROCK-1 is activated and associated with non-apoptotic membrane blebbing in retinal vessels and in retinal pigment epithelium (RPE) that respectively form the inner and the outer barriers. Activation of ROCK-1 induces focal vascular constrictions, endoluminal blebbing and subsequent retinal hypoxia. In RPE cells, actin cytoskeleton remodeling and membrane blebs in RPE cells contributes to outer barrier breakdown. Intraocular injection of fasudil, significantly reduces both retinal hypoxia and RPE barrier breakdown. Diabetes-induced cell blebbing may contribute to ischemic maculopathy and represent an intervention target.


Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Retinal Pigment Epithelium/metabolism , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Aged , Animals , Biomarkers , Case-Control Studies , Cytoskeleton/metabolism , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Female , Fluorescent Antibody Technique , Humans , Hypoxia/metabolism , Immunohistochemistry , Male , Middle Aged , Rats , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Vessels/ultrastructure , rho-Associated Kinases/genetics
15.
Transl Res ; 188: 40-57.e4, 2017 10.
Article En | MEDLINE | ID: mdl-28754419

Several approaches have been developed for gene therapy in RPE65-related Leber congenital amaurosis. To date, strategies that have reached the clinical stages rely on adeno-associated viral vectors and two of them documented limited long-term effect. We have developed a lentiviral-based strategy of RPE65 gene transfer that efficiently restored protein expression and cone function in RPE65-deficient mice. In this study, we evaluated the ocular and systemic tolerances of this lentiviral-based therapy (LV-RPE65) on healthy nonhuman primates (NHPs), without adjuvant systemic anti-inflammatory prophylaxis. For the first time, we describe the early kinetics of retinal detachment at 2, 4, and 7 days after subretinal injection using multimodal imaging in 5 NHPs. We revealed prolonged reattachment times in LV-RPE65-injected eyes compared to vehicle-injected eyes. Low- (n = 2) and high-dose (n = 2) LV-RPE65-injected eyes presented a reduction of the outer nuclear and photoreceptor outer segment layer thickness in the macula, that was more pronounced than in vehicle-injected eyes (n = 4). All LV-RPE65-injected eyes showed an initial perivascular reaction that resolved spontaneously within 14 days. Despite foveal structural changes, full-field electroretinography indicated that the overall retinal function was preserved over time and immunohistochemistry identified no difference in glial, microglial, or leucocyte ocular activation between low-dose, high-dose, and vehicle-injected eyes. Moreover, LV-RPE65-injected animals did not show signs of vector shedding or extraocular targeting, confirming the safe ocular restriction of the vector. Our results evidence a limited ocular tolerance to LV-RPE65 after subretinal injection without adjuvant anti-inflammatory prophylaxis, with complications linked to this route of administration necessitating to block this transient inflammatory event.


Eye Proteins/administration & dosage , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Lentivirus/genetics , Receptors, G-Protein-Coupled/administration & dosage , Animals , Female , Macaca fascicularis , Retina
16.
Elife ; 62017 05 26.
Article En | MEDLINE | ID: mdl-28548639

Besides its role in vision, light impacts physiology and behavior through circadian and direct (aka 'masking') mechanisms. In Smith-Magenis syndrome (SMS), the dysregulation of both sleep-wake behavior and melatonin production strongly suggests impaired non-visual light perception. We discovered that mice haploinsufficient for the SMS causal gene, Retinoic acid induced-1 (Rai1), were hypersensitive to light such that light eliminated alert and active-wake behaviors, while leaving time-spent-awake unaffected. Moreover, variables pertaining to circadian rhythm entrainment were activated more strongly by light. At the input level, the activation of rod/cone and suprachiasmatic nuclei (SCN) by light was paradoxically greatly reduced, while the downstream activation of the ventral-subparaventricular zone (vSPVZ) was increased. The vSPVZ integrates retinal and SCN input and, when activated, suppresses locomotor activity, consistent with the behavioral hypersensitivity to light we observed. Our results implicate Rai1 as a novel and central player in processing non-visual light information, from input to behavioral output.


Circadian Rhythm/radiation effects , Hypothalamus/physiology , Light , Trans-Activators/metabolism , Wakefulness/radiation effects , Animals , Behavior, Animal , Mice
17.
Stem Cell Reports ; 8(6): 1659-1674, 2017 06 06.
Article En | MEDLINE | ID: mdl-28552606

The loss of cone photoreceptors that mediate daylight vision represents a leading cause of blindness, for which cell replacement by transplantation offers a promising treatment strategy. Here, we characterize cone differentiation in retinas derived from mouse embryonic stem cells (mESCs). Similar to in vivo development, a temporal pattern of progenitor marker expression is followed by the differentiation of early thyroid hormone receptor ß2-positive precursors and, subsequently, photoreceptors exhibiting cone-specific phototransduction-related proteins. We establish that stage-specific inhibition of the Notch pathway increases cone cell differentiation, while retinoic acid signaling regulates cone maturation, comparable with their actions in vivo. MESC-derived cones can be isolated in large numbers and transplanted into adult mouse eyes, showing capacity to survive and mature in the subretinal space of Aipl1-/- mice, a model of end-stage retinal degeneration. Together, this work identifies a robust, renewable cell source for cone replacement by purified cell suspension transplantation.


Mouse Embryonic Stem Cells/transplantation , Retinal Cone Photoreceptor Cells/cytology , Retinal Degeneration/therapy , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Eye Proteins/antagonists & inhibitors , Eye Proteins/genetics , Eye Proteins/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Oligodendrocyte Transcription Factor 2/metabolism , Opsins/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Otx Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/pathology , Signal Transduction , Tretinoin/metabolism , Tretinoin/pharmacology
18.
Sci Rep ; 7: 43093, 2017 03 07.
Article En | MEDLINE | ID: mdl-28266561

The homeodomain transcription factor Nkx2.1 (NK2 homeobox 1) controls cell differentiation of telencephalic GABAergic interneurons and oligodendrocytes. Here we show that Nkx2.1 also regulates astrogliogenesis of the telencephalon from embryonic day (E) 14.5 to E16.5. Moreover we identify the different mechanisms by which Nkx2.1 controls the telencephalic astrogliogenesis. In Nkx2.1 knockout (Nkx2.1-/-) mice a drastic loss of astrocytes is observed that is not related to cell death. Further, in vivo analysis using BrdU incorporation reveals that Nkx2.1 affects the proliferation of the ventral neural stem cells that generate early astrocytes. Also, in vitro neurosphere assays showed reduced generation of astroglia upon loss of Nkx2.1, which could be due to decreased precursor proliferation and possibly defects in glial specification/differentiation. Chromatin immunoprecipitation analysis and in vitro co-transfection studies with an Nkx2.1-expressing plasmid indicate that Nkx2.1 binds to the promoter of glial fibrillary acidic protein (GFAP), primarily expressed in astrocytes, to regulate its expression. Hence, Nkx2.1 controls astroglial production spatiotemporally in embryos by regulating proliferation of the contributing Nkx2.1-positive precursors.


Astrocytes/metabolism , Cell Differentiation , Embryonic Development , Telencephalon/metabolism , Thyroid Nuclear Factor 1/physiology , Animals , Astrocytes/physiology , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Mice , Mice, Knockout , Telencephalon/physiology , Thyroid Nuclear Factor 1/metabolism
19.
Mol Ther ; 25(3): 634-653, 2017 03 01.
Article En | MEDLINE | ID: mdl-28143742

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.


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
20.
PLoS One ; 12(2): e0171474, 2017.
Article En | MEDLINE | ID: mdl-28182703

Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-dependent transcription factor involved in many aspects of metabolism, immune response, and development. Total-body deletion of the two Pparg alleles provoked generalized lipoatrophy along with severe type 2 diabetes. Herein, we explore the appearance and development of structural and functional alterations of the kidney, comparing Pparg null-mice to their littermate controls (carrying Pparg floxed alleles). We show that renal hypertrophy and functional alterations with increased glucosuria and albuminuria are already present in 3 weeks-old Pparg null-mice. Renal insufficiency with decreased creatinine clearance progress at 7 weeks of age, with the advance of the type 2 diabetes. At 52 weeks of age, these alterations are accompanied by signs of fibrosis and mesangial expansion. More intriguingly, aged Pparg null-mice concomitantly present an anti-phospholipid syndrome (APS), characterized by the late appearance of microthrombi and a mesangioproliferative pattern of glomerular injury, associated with significant plasmatic levels of anti-ß2- glycoprotein1 antibodies and renal deposition of IgG, IgM, and C3. Thus, in line with the role of PPARγ in metabolic homeostasis, Pparg null-mice first represent a potent model for studying the initiation and the development of diabetic nephropathy. Second, and in relation with the important PPARγ activity in inflammation and in immune system, these mice also highlight a new role for PPARγ signaling in the promotion of APS, a syndrome whose pathogenesis is poorly known and whose current treatment is limited to prevention of thrombosis events.


Antiphospholipid Syndrome/genetics , Diabetic Nephropathies/metabolism , PPAR gamma/genetics , Animals , Antibodies/immunology , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/pathology , Complement C3/immunology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Homeostasis , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Mice , PPAR gamma/metabolism , Signal Transduction , beta 2-Glycoprotein I/immunology
...