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










Publication year range
1.
Sci Adv ; 9(31): eadg8163, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531424

ABSTRACT

The anatomical differences between the retinas of humans and most animal models pose a challenge for testing novel therapies. Nonhuman primate (NHP) retina is anatomically closest to the human retina. However, there is a lack of relevant NHP models of retinal degeneration (RD) suitable for preclinical studies. To address this unmet need, we generated three distinct inducible cynomolgus macaque models of RD. We developed two genetically targeted strategies using optogenetics and CRISPR-Cas9 to ablate rods and mimic rod-cone dystrophy. In addition, we created an acute model by physical separation of the photoreceptors and retinal pigment epithelium using a polymer patch. Among the three models, the CRISPR-Cas9-based approach was the most advantageous model in view of recapitulating disease-specific features and its ease of implementation. The acute model, however, resulted in the fastest degeneration, making it the most relevant model for testing end-stage vision restoration therapies such as stem cell transplantation.


Subject(s)
Retinal Degeneration , Animals , Humans , Retinal Degeneration/therapy , Retina , Retinal Rod Photoreceptor Cells , Retinal Pigment Epithelium , Primates
2.
Ophthalmol Sci ; 3(4): 100316, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37274010

ABSTRACT

Objective: To describe adaptive optics flood illumination ophthalmoscopy (AO-FIO) of the photoreceptor layer in normal nonhuman primates (NHPs) and in the case of a short-term induced retinal detachment (RD). Design: Longitudinal fundamental research study. Subjects: Four NHPs were used to image normal retinae with AO-FIO (in comparison with 4 healthy humans); 2 NHPs were used to assess the effects of RD. Intervention: The photoreceptor layer (cone mosaic metrics, including cone density, cone spacing, and cone regularity) was followed with AO-FIO imaging (rtx1, Imagine Eyes) during a surgically induced RD in 2 NHPs using a vehicle solution containing dimethyl sulfoxide, classically used as a chemical solvent. We also performed functional testing of the retina (full-field and multifocal electroretinogram [ERG]). Main Outcome Measures: Correlation of cone mosaic metrics (cone density, spacing, and regularity) between normal retinae of NHPs and humans, and cone metrics, power spectrum, and ERG wave amplitudes after RD. Results: Imaging features were very similar in terms of cone reflectivity, cell density, regularity, and spacing values, showing strong positive correlations between NHPs and humans. After RD, AO-FIO revealed several alterations of the cone mosaic slowly recovering during the 3 months after the reattachment, which were not detected functionally by ERG. Conclusions: These results demonstrate by in vivo AO-FIO imaging the transient structural changes of photoreceptors after an RD in the primate retina. They also provide an interesting illustration of the AO-FIO potential for investigating photoreceptor toxicity during preclinical studies in NHPs with a high translatability to human studies. Financial Disclosures: Proprietary or commercial disclosure may be found after the references.

4.
Mol Ther Methods Clin Dev ; 24: 306-316, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35229004

ABSTRACT

Positive clinical outcomes in adeno-associated virus (AAV)-mediated retinal gene therapy have often been attributed to the low immunogenicity of AAVs and immune privilege of the eye. However, several recent studies have shown potential for inflammatory responses. The current understanding of the factors contributing to inflammation, such as the pre-existence of serum antibodies against AAVs and their contribution to increases in antibody levels post-injection, is incomplete. The parameters that regulate the generation of new antibodies in response to the AAV capsid or transgene after intraocular injections are also insufficiently described. This study is a retrospective analysis of the pre-existing serum antibodies in correlation with changes in antibody levels after intraocular injections of AAV in non-human primates (NHPs) of the species Macaca fascicularis. In NHP serums, we analyzed the binding antibody (BAB) levels and a subset of these called neutralizing antibodies (NABs) that impede AAV transduction. We observed significantly higher pre-existing serum BABs against AAV8 compared with other serotypes and a dose-dependent increase in BABs and NABs in the serums collected post-injection, irrespective of the serotype or the mode of injection. Lastly, we were able to demonstrate a correlation between the serum BAB levels with clinical grading of inflammation and levels of transgene expression.

5.
Commun Biol ; 5(1): 89, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075261

ABSTRACT

Human cone phototropism is a key mechanism underlying the Stiles-Crawford effect, a psychophysiological phenomenon according to which photoreceptor outer/inner segments are aligned along with the direction of incoming light. However, such photomechanical movements of photoreceptors remain elusive in mammals. We first show here that primate cone photoreceptors have a planar polarity organized radially around the optical center of the eye. This planar polarity, based on the structure of the cilium and calyceal processes, is highly reminiscent of the planar polarity of the hair cells and their kinocilium and stereocilia. Secondly, we observe under super-high resolution expansion microscopy the cytoskeleton and Usher proteins architecture in the photoreceptors, which appears to establish a mechanical continuity between the outer and inner segments. Taken together, these results suggest a comprehensive cellular mechanism consistent with an active phototropism of cones toward the optical center of the eye, and thus with the Stiles-Crawford effect.


Subject(s)
Cell Polarity/physiology , Light , Retinal Cone Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/radiation effects , Animals , Biomechanical Phenomena , Cytoskeleton , Macaca fascicularis , Reproducibility of Results , Retinal Cone Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/physiology
6.
Redox Biol ; 48: 102198, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34856436

ABSTRACT

The nucleoredoxin gene NXNL2 encodes for two products through alternative splicing, rod-derived cone viability factor-2 (RdCVF2) that mediates neuronal survival and the thioredoxin-related protein (RdCVF2L), an enzyme that regulates the phosphorylation of TAU. To investigate the link between NXNL2 and tauopathies, we studied the Nxnl2 knockout mouse (Nxnl2-/-). We established the expression pattern of the Nxnl2 gene in the brain using a Nxnl2 reporter mouse line, and characterized the behavior of the Nxnl2-/- mouse at 2 months of age. Additionally, long term potentiation and metabolomic from hippocampal specimens were collected at 2 months of age. We studied TAU oligomerization, phosphorylation and aggregation in Nxnl2-/- brain at 18 months of age. Finally, newborn Nxnl2-/- mice were treated with adeno-associated viral vectors encoding for RdCVF2, RdCVF2L or both and measured the effect of this therapy on long-term potential, glucose metabolism and late-onset tauopathy. Nxnl2-/- mice at 2 months of age showed severe behavioral deficiency in fear, pain sensitivity, coordination, learning and memory. The Nxnl2-/- also showed deficits in long-term potentiation, demonstrating that the Nxnl2 gene is involved in regulating brain functions. Dual delivery of RdCVF2 and RdCVF2L in newborn Nxnl2-/- mice fully correct long-term potentiation through their synergistic action. The expression pattern of the Nxnl2 gene in the brain shows a predominant expression in circumventricular organs, such as the area postrema. Glucose metabolism of the hippocampus of Nxnl2-/- mice at 2 months of age was reduced, and was not corrected by gene therapy. At 18-month-old Nxnl2-/- mice showed brain stigmas of tauopathy, such as oligomerization, phosphorylation and aggregation of TAU. This late-onset tauopathy can be prevented, albeit with modest efficacy, by recombinant AAVs administrated to newborn mice. The Nxnl2-/- mice have memory dysfunction at 2-months that resembles mild-cognitive impairment and at 18-months exhibit tauopathy, resembling to the progression of Alzheimer's disease. We propose the Nxnl2-/- mouse is a model to study multistage aged related neurodegenerative diseases. The NXNL2 metabolic and redox signaling is a new area of therapeutic research in neurodegenerative diseases.

7.
Commun Biol ; 4(1): 125, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504896

ABSTRACT

Vision restoration is an ideal medical application for optogenetics, because the eye provides direct optical access to the retina for stimulation. Optogenetic therapy could be used for diseases involving photoreceptor degeneration, such as retinitis pigmentosa or age-related macular degeneration. We describe here the selection, in non-human primates, of a specific optogenetic construct currently tested in a clinical trial. We used the microbial opsin ChrimsonR, and showed that the AAV2.7m8 vector had a higher transfection efficiency than AAV2 in retinal ganglion cells (RGCs) and that ChrimsonR fused to tdTomato (ChR-tdT) was expressed more efficiently than ChrimsonR. Light at 600 nm activated RGCs transfected with AAV2.7m8 ChR-tdT, from an irradiance of 1015 photons.cm-2.s-1. Vector doses of 5 × 1010 and 5 × 1011 vg/eye transfected up to 7000 RGCs/mm2 in the perifovea, with no significant immune reaction. We recorded RGC responses from a stimulus duration of 1 ms upwards. When using the recorded activity to decode stimulus information, we obtained an estimated visual acuity of 20/249, above the level of legal blindness (20/400). These results lay the groundwork for the ongoing clinical trial with the AAV2.7m8 - ChR-tdT vector for vision restoration in patients with retinitis pigmentosa.


Subject(s)
Optogenetics , Photic Stimulation , Retinal Degeneration/therapy , Vision, Ocular/physiology , Animals , Equipment and Supplies , Female , Humans , Macaca fascicularis , Male , Optogenetics/instrumentation , Optogenetics/methods , Pattern Recognition, Visual/physiology , Photic Stimulation/instrumentation , Photic Stimulation/methods , Primates , Retinal Degeneration/physiopathology , Retinal Degeneration/rehabilitation , Therapies, Investigational/instrumentation , Therapies, Investigational/methods
8.
Methods Mol Biol ; 2173: 101-112, 2020.
Article in English | MEDLINE | ID: mdl-32651912

ABSTRACT

Adeno-associated virus (AAV) has emerged as the vector of choice for delivering genes to the mammalian retina. From the first gene therapy to receive FDA approval for the inherited retinal disease (Luxturna™) to more recent clinical trials using microbial opsins to regain light sensitivity, therapeutic transgenes rely on AAV vectors for safe and efficient gene delivery to retinal cells. Such vectors are administered to the retina via subretinal (SR) injection or intravitreal (IVT) injection routes depending on the targeted retinal cell type. An attractive target for gene therapy is the fovea, bearing the highest concentration of cone cells responsible for our high acuity daylight vision. However, previous clinical trials and large animal studies reported that SR administration of vector under the cone-exclusive fovea disrupts its fine structure and might impair visual acuity. Due to its technical difficulty and potential risks, alternatives to vector injection under this delicate region have been investigated by using novel AAV capsid variants identified via rational design or directed evolution. We recently established new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea. Our methods provide efficient foveal cone transduction without detaching this delicate region and rely on the use of engineered AAVs and optimal promoters compatible with optogenetic vision restoration. Here we describe in detail our AAV vectors, methods for intravitreal and subretinal injections as well as pre- and postoperative procedures as performed in cynomolgus macaques.


Subject(s)
Dependovirus/genetics , Animals , Female , Genetic Therapy , Genetic Vectors/genetics , Male , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Tomography, Optical Coherence/methods
9.
PLoS One ; 15(4): e0230713, 2020.
Article in English | MEDLINE | ID: mdl-32267845

ABSTRACT

PURPOSE: To evaluate the surgical technique for subretinal implantation of two sizes of PRIMA photovoltaic wireless microchip in two animal models, and refine these surgical procedures for human trials. METHODS: Cats and Macaca fascicularis primates with healthy retina underwent vitrectomy surgery and were implanted with subretinal wireless photovoltaic microchip at the macula/central retina. The 1.5mm PRIMA chip was initially studied in feline eyes. PRIMA implant (2mm,1.5mm sizes) arrays were studied in primates. Feasibility of subretinal chip implantation was evaluated with a newly-developed surgical technique, with surgical complications and adverse events recorded. RESULTS: The 1.5mm implant was placed in the central retina of 11 feline eyes, with implantation duration 43-106 days. The 1.5mm implant was correctly positioned into central macula of 11 primate eyes, with follow-up periods of minimum 6 weeks (n = 11), 2 years (n = 2), and one eye for 3 years. One primate eye underwent multi-chip 1.5mm implantation using two 1.5mm chips. The 2mm implant was delivered to 4 primate eyes. Optical coherence tomography confirmed correct surgical placement of photovoltaic arrays in the subretinal space in all 26 eyes. Intraoperative complications in primate eyes included retinal tear, macular hole, retinal detachment, and vitreous hemorrhage that resolved spontaneously. Postoperatively, there was no case of significant ocular inflammation in the 1.5mm implant group. CONCLUSIONS: We report subretinal implantation of 1.5mm and 2mm photovoltaic arrays in the central retina of feline and central macula of primate eyes with a low rate of device-related complications. The in vivo PRIMA implantation technique has been developed and refined for use for a 2mm PRIMA implant in ongoing human trials.


Subject(s)
Microtechnology/instrumentation , Prostheses and Implants , Retina/surgery , Wireless Technology , Animals , Cats , Macaca fascicularis , Safety
10.
Nat Biomed Eng ; 4(2): 172-180, 2020 02.
Article in English | MEDLINE | ID: mdl-31792423

ABSTRACT

Retinal dystrophies and age-related macular degeneration related to photoreceptor degeneration can cause blindness. In blind patients, although the electrical activation of the residual retinal circuit can provide useful artificial visual perception, the resolutions of current retinal prostheses have been limited either by large electrodes or small numbers of pixels. Here we report the evaluation, in three awake non-human primates, of a previously reported near-infrared-light-sensitive photovoltaic subretinal prosthesis. We show that multipixel stimulation of the prosthesis within radiation safety limits enabled eye tracking in the animals, that they responded to stimulations directed at the implant with repeated saccades and that the implant-induced responses were present two years after device implantation. Our findings pave the way for the clinical evaluation of the prosthesis in patients affected by dry atrophic age-related macular degeneration.


Subject(s)
Macular Degeneration/rehabilitation , Saccades , Vision, Ocular/physiology , Visual Perception , Visual Prosthesis , Animals , Disease Models, Animal , Eye Movement Measurements , Macaca fascicularis , Macular Degeneration/physiopathology , Male , Photic Stimulation , Retinal Ganglion Cells/physiology
11.
Biomaterials ; 230: 119603, 2020 02.
Article in English | MEDLINE | ID: mdl-31732225

ABSTRACT

Age-related macular degeneration as well as some forms of Retinitis Pigmentosa (RP) are characterized by a retinal degeneration involving the retinal pigment epithelium (RPE). Various strategies were proposed to cure these disorders including the replacement of RPE cells using human pluripotent stem cells (hPSCs), an unlimited source material to generate in vitro RPE cells. The formulation strategy of the cell therapy (either a reconstructed sheet or a cell suspension) is crucial to achieve an efficient and long lasting therapeutic effect. We previously developed a hPSC-RPE sheet disposed on human amniotic membrane that sustained the vision of rodents with retinal degeneration compared to the same cells injected as a suspension. However, the transplantation strategy was difficult to implement in large animals. Herein we developed two medical devices for the preparation, conservation and implantation of the hPSC-RPE sheet in nonhuman primates. The surgery was safe and well tolerated during the 7-week follow up. The graft integrity was preserved in primates. Moreover, the hPSC-RPE sheet did not induce teratoma or grafted cell dispersion to other organs in rodent models. This work clears the way for the first cell therapy for RP patients carrying RPE gene mutations (LRAT, RPE65 and MERTK).


Subject(s)
Pluripotent Stem Cells , Retinal Pigment Epithelium , Stem Cell Transplantation , Animals , Cell Differentiation , Humans , Primates , Rodentia
12.
JCI Insight ; 3(2)2018 01 25.
Article in English | MEDLINE | ID: mdl-29367457

ABSTRACT

Intraocular injection of adeno-associated viral (AAV) vectors has been an evident route for delivering gene drugs into the retina. However, gaps in our understanding of AAV transduction patterns within the anatomically unique environments of the subretinal and intravitreal space of the primate eye impeded the establishment of noninvasive and efficient gene delivery to foveal cones in the clinic. Here, we establish new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea with supporting studies in mouse models, human induced pluripotent stem cell-derived organoids, postmortem human retinal explants, and living macaques. We show that an AAV9 variant provides efficient foveal cone transduction when injected into the subretinal space several millimeters away from the fovea, without detaching this delicate region. An engineered AAV2 variant provides gene delivery to foveal cones with a well-tolerated dose administered intravitreally. Both delivery modalities rely on a cone-specific promoter and result in high-level transgene expression compatible with optogenetic vision restoration. The model systems described here provide insight into the behavior of AAV vectors across species to obtain safety and efficacy needed for gene therapy in neurodegenerative disorders.


Subject(s)
Fovea Centralis/pathology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Vision Disorders/therapy , Animals , Cell Line , Dependovirus/genetics , Female , Fovea Centralis/diagnostic imaging , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells , Injections, Intraocular , Intravital Microscopy , Macaca fascicularis , Male , Mice , Models, Animal , Optogenetics/methods , Patch-Clamp Techniques , Promoter Regions, Genetic/genetics , Transgenes/genetics , Vision Disorders/genetics , Vision Disorders/pathology
13.
Mol Ther ; 25(11): 2546-2560, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28807567

ABSTRACT

The majority of inherited retinal degenerations converge on the phenotype of photoreceptor cell death. Second- and third-order neurons are spared in these diseases, making it possible to restore retinal light responses using optogenetics. Viral expression of channelrhodopsin in the third-order neurons under ubiquitous promoters was previously shown to restore visual function, albeit at light intensities above illumination safety thresholds. Here, we report (to our knowledge, for the first time) activation of macaque retinas, up to 6 months post-injection, using channelrhodopsin-Ca2+-permeable channelrhodopsin (CatCh) at safe light intensities. High-level CatCh expression was achieved due to a new promoter based on the regulatory region of the gamma-synuclein gene (SNCG) allowing strong expression in ganglion cells across species. Our promoter, in combination with clinically proven adeno-associated virus 2 (AAV2), provides CatCh expression in peri-foveolar ganglion cells responding robustly to light under the illumination safety thresholds for the human eye. On the contrary, the threshold of activation and the proportion of unresponsive cells were much higher when a ubiquitous promoter (cytomegalovirus [CMV]) was used to express CatCh. The results of our study suggest that the inclusion of optimized promoters is key in the path to clinical translation of optogenetics.


Subject(s)
Channelrhodopsins/genetics , Genetic Vectors/administration & dosage , Promoter Regions, Genetic , Recovery of Function , Retinal Degeneration/therapy , Animals , Channelrhodopsins/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Expression , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Intravitreal Injections , Light , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Transduction, Genetic , Transgenes , Vision, Ocular/physiology
14.
Cell ; 161(4): 817-32, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957687

ABSTRACT

Rod-derived cone viability factor (RdCVF) is an inactive thioredoxin secreted by rod photoreceptors that protects cones from degeneration. Because the secondary loss of cones in retinitis pigmentosa (RP) leads to blindness, the administration of RdCVF is a promising therapy for this untreatable neurodegenerative disease. Here, we investigated the mechanism underlying the protective role of RdCVF in RP. We show that RdCVF acts through binding to Basigin-1 (BSG1), a transmembrane protein expressed specifically by photoreceptors. BSG1 binds to the glucose transporter GLUT1, resulting in increased glucose entry into cones. Increased glucose promotes cone survival by stimulation of aerobic glycolysis. Moreover, a missense mutation of RdCVF results in its inability to bind to BSG1, stimulate glucose uptake, and prevent secondary cone death in a model of RP. Our data uncover an entirely novel mechanism of neuroprotection through the stimulation of glucose metabolism.


Subject(s)
Eye Proteins/metabolism , Glycolysis , Thioredoxins/metabolism , Alkaline Phosphatase/metabolism , Animals , Basigin/genetics , Basigin/metabolism , Eye Proteins/genetics , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , Mice , Mutation, Missense , Retina/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Thioredoxins/genetics
15.
C R Biol ; 337(3): 207-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24702847

ABSTRACT

The most common hereditary retinal degeneration, retinitis pigmentosa (RP), leads to blindness by degeneration of cone photoreceptors. Meanwhile, genetic studies have shown that a significant proportion of RP genes is expressed only by rods, which raises the question of the mechanism leading to the degeneration of cones. Following the concept of sustainability factor cones, rods secrete survival factors that are necessary to maintain the cones, named Rod-derived Cone Viability Factors (RdCVFs). In patients suffering from RP, loss of rods results in the loss of RdCVFs expression and followed by cone degeneration. We have identified the bifunctional genes nucleoredoxin-like 1 and 2 that encode for, by differential splicing, a thioredoxin enzyme and a cone survival factor, respectively RdCVF and RdCVF2. The administration of these survival factors would maintain cones and central vision in most patients suffering from RP.


Subject(s)
Genetic Therapy/methods , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Thioredoxins/genetics , Thioredoxins/therapeutic use , Humans , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/pathology
16.
Hum Mol Genet ; 21(10): 2298-311, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22343139

ABSTRACT

The rod-derived cone viability factors, RdCVF and RdCVF2, have potential therapeutical interests for the treatment of inherited photoreceptor degenerations. In the mouse lacking Nxnl2, the gene encoding RdCVF2, the progressive decline of the visual performance of the cones in parallel with their degeneration, arises due to the loss of trophic support from RdCVF2. In contrary, the progressive loss of rod visual function of the Nxnl2-/- mouse results from a decrease in outer segment length, mediated by a cell autonomous mechanism involving the putative thioredoxin protein RdCVF2L, the second spliced product of the Nxnl2 gene. This novel signaling mechanism extends to olfaction as shown by the progressive impairment of olfaction in aged Nxnl2-/- mice and the protection of olfactory neurons by RdCVF2. This study shows that Nxnl2 is a bi-functional gene involved in the maintenance of both the function and the viability of sensory neurons.


Subject(s)
Cell Survival/genetics , Eye Proteins/genetics , RNA Splicing , Sensory Receptor Cells/cytology , Thioredoxins/genetics , Animals , Cells, Cultured , Eye Proteins/metabolism , Mice , Retinal Rod Photoreceptor Cells/metabolism , Sensory Receptor Cells/metabolism , Thioredoxins/metabolism
17.
Nat Genet ; 41(10): 1094-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19734903

ABSTRACT

The gene encoding apolipoprotein E (APOE) on chromosome 19 is the only confirmed susceptibility locus for late-onset Alzheimer's disease. To identify other risk loci, we conducted a large genome-wide association study of 2,032 individuals from France with Alzheimer's disease (cases) and 5,328 controls. Markers outside APOE with suggestive evidence of association (P < 10(-5)) were examined in collections from Belgium, Finland, Italy and Spain totaling 3,978 Alzheimer's disease cases and 3,297 controls. Two loci gave replicated evidence of association: one within CLU (also called APOJ), encoding clusterin or apolipoprotein J, on chromosome 8 (rs11136000, OR = 0.86, 95% CI 0.81-0.90, P = 7.5 x 10(-9) for combined data) and the other within CR1, encoding the complement component (3b/4b) receptor 1, on chromosome 1 (rs6656401, OR = 1.21, 95% CI 1.14-1.29, P = 3.7 x 10(-9) for combined data). Previous biological studies support roles of CLU and CR1 in the clearance of beta amyloid (Abeta) peptide, the principal constituent of amyloid plaques, which are one of the major brain lesions of individuals with Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Clusterin/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Receptors, Complement 3b/genetics , Haplotypes , Humans , Polymorphism, Single Nucleotide , White People/genetics
18.
Mol Cell Proteomics ; 8(6): 1206-18, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19279044

ABSTRACT

Rod-derived cone viability factor (RdCVF) is produced by the Nxnl1 gene that codes for a second polypeptide, RdCVFL, by alternative splicing. Although the role of RdCVF in promoting cone survival has been described, the implication of RdCVFL, a putative thioredoxin enzyme, in the protection of photoreceptors is presently unknown. Using a proteomics approach we identified 90 proteins interacting with RdCVFL including the microtubule-binding protein TAU. We demonstrate that the level of phosphorylation of TAU is increased in the retina of the Nxnl1(-/-) mice as it is hyperphosphorylated in the brain of patients suffering from Alzheimer disease, presumably in some cases through oxidative stress. Using a cell-based assay, we show that RdCVFL inhibits TAU phosphorylation. In vitro, RdCVFL protects TAU from oxidative damage. Photooxidative stress is implicated in retinal degeneration, particularly in retinitis pigmentosa, where it is considered to be a contributor to secondary cone death. The functional interaction between RdCVFL and TAU described here is the first characterization of the RdCVFL signaling pathway involved in neuronal cell death mediated by oxidative stress.


Subject(s)
Eye Proteins/metabolism , Retina/metabolism , Thioredoxins/metabolism , tau Proteins/metabolism , Animals , Cell Line , Chromatography, Liquid , Eye Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Oxidative Stress , Phosphorylation , Protein Binding , Protein Isoforms/metabolism , Tandem Mass Spectrometry , Thioredoxins/genetics
19.
BMC Mol Biol ; 8: 74, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17764561

ABSTRACT

BACKGROUND: Cone degeneration is the hallmark of the inherited retinal disease retinitis pigmentosa. We have previously identified a trophic factor "Rod-derived Cone Viability Factor (RdCVF) that is secreted by rods and promote cone viability in a mouse model of the disease. RESULTS: Here we report the bioinformatic identification and the experimental analysis of RdCVF2, a second trophic factor belonging to the Rod-derived Cone Viability Factor family. The mouse RdCVF gene is known to be bifunctional, encoding both a long thioredoxin-like isoform (RdCVF-L) and a short isoform with trophic cone photoreceptor viability activity (RdCVF-S). RdCVF2 shares many similarities with RdCVF in terms of gene structure, expression in a rod-dependent manner and protein 3D structure. Furthermore, like RdCVF, the RdCVF2 short isoform exhibits cone rescue activity that is independent of its putative thiol-oxydoreductase activity. CONCLUSION: Taken together, these findings define a new family of bifunctional genes which are: expressed in vertebrate retina, encode trophic cone viability factors, and have major therapeutic potential for human retinal neurodegenerative diseases such as retinitis pigmentosa.


Subject(s)
Eye Proteins/chemistry , Retinal Rod Photoreceptor Cells/metabolism , Thioredoxins/chemistry , Animals , Cell Survival , Cells, Cultured , Chick Embryo , Computer Simulation , Culture Media, Conditioned/chemistry , Eye Proteins/genetics , Eye Proteins/pharmacology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Models, Molecular , Molecular Sequence Data , Photoreceptor Cells, Vertebrate/metabolism , Phylogeny , Protein Isoforms/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Sequence Alignment , Sequence Analysis, DNA , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Thioredoxins/pharmacology
20.
J Neurosci ; 23(27): 9229-39, 2003 Oct 08.
Article in English | MEDLINE | ID: mdl-14534257

ABSTRACT

Semaphorins are a family of secreted and membrane-bound proteins, known to regulate axonal pathfinding. Sema4D, also called CD100, was first isolated in the immune system where it is involved in B and T cell activation. We found that in the mouse, Sema4D is expressed in cells throughout the CNS white matter, with a peak during the myelination period. Double-labeling experiments with different markers of oligodendrocyte lineage such as olig1, olig2, platelet-derived growth factor receptor alpha, and proteolipid protein showed that Sema4D was expressed selectively by oligodendrocytes and myelin. The presence of Sema4D in myelin was confirmed using Western blot. Sema4D expression in myelinating oligodendrocytes was further observed using neuron-oligodendrocyte cocultures. Moreover, using stripe assay, we found that Sema4D is strongly inhibitory for postnatal sensory and cerebellar granule cell axons. This prompted us to examine whether Sema4D expression is modified after CNS injury. At 8 d after spinal cord lesions, Sema4D expression was strongly upregulated in oligodendrocytes at the periphery of the lesion. Sema4D-positive cells were not colabeled with the astrocyte marker GFAP, with the microglial and macrophagic marker isolectin B4, or with NG2, a marker of oligodendrocyte precursors. This upregulation was transient because from 1 month after the lesion, Sema4D expression was back to its normal level. These results indicate that Sema4D is a novel inhibitory factor for axonal regeneration expressed in myelin.


Subject(s)
Antigens, CD , Axons/physiology , Central Nervous System/metabolism , Membrane Glycoproteins/metabolism , Oligodendroglia/metabolism , Semaphorins , Spinal Cord Injuries/metabolism , Animals , Axons/drug effects , Biomarkers/analysis , Cells, Cultured , Central Nervous System/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred Strains , Mice, Transgenic , Myelin Sheath/metabolism , Oligodendroglia/pathology , RNA, Messenger/metabolism , Spinal Cord Injuries/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...