Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article En | MEDLINE | ID: mdl-34502128

Age-related macular degeneration (AMD), one of the leading causes of blindness worldwide, causes personal suffering and high socioeconomic costs. While there has been progress in the treatments for the neovascular form of AMD, no therapy is yet available for the more common dry form, also known as geographic atrophy. We analysed the retinal tissue in a mouse model of retinal degeneration caused by sodium iodate (NaIO3)-induced retinal pigment epithelium (RPE) atrophy to understand the underlying pathology. RNA sequencing (RNA-seq), qRT-PCR, Western blot, immunohistochemistry of the retinas and multiplex ELISA of the mouse serum were applied to find the pathways involved in the degeneration. NaIO3 caused patchy RPE loss and thinning of the photoreceptor layer. This was accompanied by the increased retinal expression of complement components c1s, c3, c4, cfb and cfh. C1s, C3, CFH and CFB were complement proteins, with enhanced deposition at day 3. C4 was upregulated in retinal degeneration at day 10. Consistently, the transcript levels of proinflammatory ccl-2, -3, -5, il-1ß, il-33 and tgf-ß were increased in the retinas of NaIO3 mice, but vegf-a mRNA was reduced. Macrophages, microglia and gliotic Müller cells could be a cellular source for local retinal inflammatory changes in the NaIO3 retina. Systemic complement and cytokines/chemokines remained unaltered in this model of NaIO3-dependent retinal degeneration. In conclusion, systemically administered NaIO3 promotes degenerative and inflammatory processes in the retina, which can mimic the hallmarks of geographic atrophy.


Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Susceptibility , Iodates/adverse effects , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Complement System Proteins/genetics , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Immunity, Innate , Immunohistochemistry , Mice , Retinal Degeneration/pathology
2.
Hum Mol Genet ; 28(3): 459-475, 2019 02 01.
Article En | MEDLINE | ID: mdl-30307502

The retinal disease gene peripherin 2 (PRPH2) is essential for the formation of photoreceptor outer segments (OSs), where it functions in oligomers with and without its homologue ROM1. However, the precise role of these proteins in OS morphogenesis is not understood. By utilizing a knock-in mouse expressing a chimeric protein comprised of the body of Rom1 and the C-terminus of Prph2 (termed RRCT), we find that the Prph2 C-terminus is necessary and sufficient for the initiation of OSs, while OS maturation requires the body of Prph2 and associated large oligomers. Importantly, dominant-negative physiological and biochemical defects in RRCT heterozygous rods are rescued by removing Rom1, suggesting Rom1 is a regulator for OS formation. Our experiments evaluating Prph2 trafficking show that Rom1 is a key determinant of whether Prph2 complexes utilize conventional versus unconventional (Golgi bypass) secretory pathways to reach the OS. These findings significantly advance our understanding of the molecular underpinnings of OS morphogenesis and particularly the role of Rom1.


Eye Proteins/physiology , Membrane Proteins/physiology , Peripherins/physiology , Photoreceptor Cells, Vertebrate/physiology , Animals , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Knock-In Techniques/methods , Heterozygote , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Mutation , Nerve Tissue Proteins/genetics , Peripherins/genetics , Peripherins/metabolism , Phenotype , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/physiology , Tetraspanins
3.
Hum Mol Genet ; 27(20): 3507-3518, 2018 10 15.
Article En | MEDLINE | ID: mdl-29961824

Mutations in peripherin 2 (PRPH2, also known as Rds), a tetraspanin protein found in photoreceptor outer segments (OSs), cause retinal degeneration ranging from rod-dominant retinitis pigmentosa (RP) to cone-dominant macular dystrophy (MD). Understanding why some Prph2 mutants affect rods while others affect cones remains a critical unanswered question. Prph2 is essential for OS structure and function and exhibits a very specific pattern of oligomerization with its homolog Rom1. Non-covalent Prph2/Rom1 homo- and hetero-tetramers assemble into higher-order covalently linked complexes held together by an intermolecular disulfide bond at Prph2-C150/Rom1-C153. Here we disrupt this crucial bond using a C150S-Prph2 knockin mouse line to study the role of Prph2 higher-order complex formation. We find that C150S-Prph2 traffics to the OS, interacts with Rom1 and forms non-covalent tetramers, but alone cannot support normal OS structure and function. However, C150S-Prph2 supports the initiation or elaboration of OS disc structures, and improves rod OS ultrastructure in the presence of wild-type (WT) Prph2 (i.e. Prph2C150S/+ versus Prph2+/-). Prph2C150S/+ animals exhibit haploinsufficiency in rods, but a dominant-negative phenotype in cones, suggesting cones have a different requirement for large Prph2 complexes than rods. Importantly, cone but not rod function can be improved by the addition of one Prph2Y141C allele, a mutation responsible for pattern dystrophy owing to the extra cysteine. Combined these findings show that covalently linked Prph2 complexes are essential for OS formation, but not for Prph2 targeting to the OS, and that cones are especially sensitive to having a broad distribution of Prph2 complex types (i.e. tetramers and large complexes).


Eye Proteins/metabolism , Membrane Proteins/metabolism , Mutation , Peripherins/metabolism , Protein Multimerization , Retinal Degeneration/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Animals , Cell Line , Disease Models, Animal , Female , Male , Mice , Peripherins/genetics , Protein Interaction Domains and Motifs , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Retinal Photoreceptor Cell Outer Segment/physiology , Tetraspanins
4.
Adv Exp Med Biol ; 1074: 109-115, 2018.
Article En | MEDLINE | ID: mdl-29721934

Considerable progress has been made in the design and delivery of non-viral gene therapy vectors, but, like their viral counterparts, therapeutic levels of transgenes have not met the requirements for successful clinical applications so far. The biggest advantage of polymer-based nanoparticle vectors is the ease with which they can be modified to increase their ability to penetrate the cell membrane and target specific cells by simply changing the formulation of the nanoparticle compaction. We took advantage of this characteristic to improve transfection rates of our particles to meet the transgene levels which will be needed for future treatment of patients. For this study, we successfully investigated the possibility of our established pegylated polylysine particles to be administered via intravitreal rather than subretinal route to ease the damage during injection. We also demonstrated that our particles are flexible enough to sustain changes in the formulation to accommodate additional targeting sequences without losing their efficiency in transfecting neuronal cells in the retina. Together, these results give us the opportunity to even further improve our particles.


Gene Transfer Techniques , Genetic Vectors/administration & dosage , Injections, Intraocular/methods , Nanoparticles/administration & dosage , Polylysine/administration & dosage , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , DNA, Recombinant/administration & dosage , Intravitreal Injections , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Particle Size , Polyethylene Glycols/administration & dosage , Retinal Pigment Epithelium/cytology , Transgenes
5.
Hum Mol Genet ; 25(16): 3500-3514, 2016 08 15.
Article En | MEDLINE | ID: mdl-27365499

Peripherin 2 (Prph2) is a photoreceptor tetraspanin, and deletion of codon 153 (K153Δ) leads to retinitis pigmentosa, pattern dystrophy, and fundus flavimaculatus in the same family. To study this variability, we generated a K153Δ-Prph2 knockin mouse. K153Δ-Prph2 cannot form the complexes required for outer segment formation, and in cones cannot interact with its binding partner rod outer segment membrane protein 1. K153Δ causes dominant defects in rod and cone function; however, rod but not cone ultrastructure is improved by the presence of K153Δ-Prph2. Likewise, supplementation of K153Δ heterozygotes with WT-Prph2 results in structural but not functional improvements. These results support the idea that mutations may differentially affect Prph2's role as a structural component, and its role as a functional protein key for organizing membrane domains for cellular signalling. These roles may be different in rods and cones, thus contributing to the phenotypic heterogeneity that characterizes diseases associated with Prph2 mutations.


Peripherins/genetics , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/metabolism , Animals , Codon/genetics , Gene Knock-In Techniques , Heterozygote , Humans , Mice , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/ultrastructure , Sequence Deletion
6.
J Control Release ; 219: 471-487, 2015 Dec 10.
Article En | MEDLINE | ID: mdl-26439665

Currently there are no viable treatment options for patients with debilitating inherited retinal degeneration. The vast variability in disease-inducing mutations and resulting phenotypes has hampered the development of therapeutic interventions. Gene therapy is a logical approach, and recent work has focused on ways to optimize vector design and packaging to promote optimized expression and phenotypic rescue after intraocular delivery. In this review, we discuss ongoing ocular clinical trials, which currently use viral gene delivery, but focus primarily on new advancements in optimizing the efficacy of non-viral gene delivery for ocular diseases. Non-viral delivery systems are highly customizable, allowing functionalization to improve cellular and nuclear uptake, bypassing cellular degradative machinery, and improving gene expression in the nucleus. Non-viral vectors often yield transgene expression levels lower than viral counterparts, however their favorable safety/immune profiles and large DNA capacity (critical for the delivery of large ocular disease genes) make their further development a research priority. Recent work on particle coating and vector engineering presents exciting ways to overcome limitations of transient/low gene expression levels, but also highlights the fact that further refinements are needed before use in the clinic.


DNA/administration & dosage , Eye Diseases/therapy , Gene Transfer Techniques , Genetic Therapy , Animals , Biological Transport , Cell Nucleus/metabolism , Eye Diseases/metabolism , Humans , Nanoparticles/administration & dosage , Retina/metabolism
7.
PLoS One ; 10(9): e0138508, 2015.
Article En | MEDLINE | ID: mdl-26406599

Mutations in the photoreceptor protein peripherin-2 (also known as RDS) cause severe retinal degeneration. RDS and its homolog ROM-1 (rod outer segment protein 1) are synthesized in the inner segment and then trafficked into the outer segment where they function in tetramers and covalently linked larger complexes. Our goal is to identify binding partners of RDS and ROM-1 that may be involved in their biosynthetic pathway or in their function in the photoreceptor outer segment (OS). Here we utilize several methods including mass spectrometry after affinity purification, in vitro co-expression followed by pull-down, in vivo pull-down from mouse retinas, and proximity ligation assay to identify and confirm the SNARE proteins Syntaxin 3B and SNAP-25 as novel binding partners of RDS and ROM-1. We show that both covalently linked and non-covalently linked RDS complexes interact with Syntaxin 3B. RDS in the mouse is trafficked from the inner segment to the outer segment by both conventional (i.e., Golgi dependent) and unconventional secretory pathways, and RDS from both pathways interacts with Syntaxin3B. Syntaxin 3B and SNAP-25 are enriched in the inner segment (compared to the outer segment) suggesting that the interaction with RDS/ROM-1 occurs in the inner segment. Syntaxin 3B and SNAP-25 are involved in mediating fusion of vesicles carrying other outer segment proteins during outer segment targeting, so could be involved in the trafficking of RDS/ROM-1.


Eye Proteins/metabolism , Membrane Proteins/metabolism , Peripherins/metabolism , Qa-SNARE Proteins/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Golgi Apparatus/metabolism , Mass Spectrometry/methods , Mice , Qa-SNARE Proteins/isolation & purification , Signal Transduction , Synaptosomal-Associated Protein 25/isolation & purification , Tetraspanins
8.
Int J Mol Sci ; 16(7): 15086-103, 2015 Jul 03.
Article En | MEDLINE | ID: mdl-26151844

Herein, we have investigated retinal cell-death pathways in response to the retina toxin sodium iodate (NaIO3) both in vivo and in vitro. C57/BL6 mice were treated with a single intravenous injection of NaIO3 (35 mg/kg). Morphological changes in the retina post NaIO3 injection in comparison to untreated controls were assessed using electron microscopy. Cell death was determined by TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining. The activation of caspases and calpain was measured using immunohistochemistry. Additionally, cytotoxicity and apoptosis in retinal pigment epithelial (RPE) cells, primary retinal cells, and the cone photoreceptor (PRC) cell line 661W were assessed in vitro after NaIO3 treatment using the ApoToxGlo™ assay. The 7-AAD/Annexin-V staining was performed and necrostatin (Nec-1) was administered to the NaIO3-treated cells to confirm the results. In vivo, degenerating RPE cells displayed a rounded shape and retracted microvilli, whereas PRCs featured apoptotic nuclei. Caspase and calpain activity was significantly upregulated in retinal sections and protein samples from NaIO3-treated animals. In vitro, NaIO3 induced necrosis in RPE cells and apoptosis in PRCs. Furthermore, Nec-1 significantly decreased NaIO3-induced RPE cell death, but had no rescue effect on treated PRCs. In summary, several different cell-death pathways are activated in retinal cells as a result of NaIO3.


Apoptosis , Caspases/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Caspases/genetics , Iodates/toxicity , Mice , Mice, Inbred C57BL , Necrosis , Retinal Degeneration/etiology , Retinal Pigment Epithelium/drug effects
9.
Graefes Arch Clin Exp Ophthalmol ; 253(5): 721-31, 2015 May.
Article En | MEDLINE | ID: mdl-25875043

PURPOSE: To identify programmed cell death (PCD) pathways involved in N-methyl-N-nitrosourea (MNU)-induced photoreceptor (PR) degeneration. METHODS: Adult C57BL/6 mice received a single MNU i.p. injection (60 mg/kg bodyweight), and were observed over a period of 7 days. Degeneration was visualized by H&E overview staining and electron microscopy. PR cell death was measured by quantifying TUNEL-positive cells in the outer nuclear layer (ONL). Activity measurements of key PCD enzymes (calpain, caspases) were used to identify the involved cell death pathways. Furthermore, the expression level of C/EBP homologous protein (CHOP) and glucose-regulated protein 78 (GRP78), key players in endoplasmic reticulum (ER) stress-induced apoptosis, was analyzed using quantitative real-time PCR. RESULTS: A decrease in ONL thickness and the appearance of apoptotic PR nuclei could be detected beginning 3 days post-injection (PI). This was accompanied by an increase of TUNEL-positive cells. Significant upregulation of activated caspases (3, 9, 12) was found at different time periods after MNU injection. Additionally, several other players of nonconventional PCD pathways were also upregulated. Consequently, calpain activity increased in the ONL, with a maximum on day 7 PI and an upregulation of CHOP and GRP78 expression beginning on day 1 PI was found. CONCLUSIONS: The data indicate that regular apoptosis is the major cause of MNU-induced PR cell death. However, alternative PCD pathways, including ER stress and calpain activation, are also involved. Knowledge about the mechanisms involved in this mouse model of PR degeneration could facilitate the design of putative combinatory therapeutic approaches.


Apoptosis , Disease Models, Animal , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Degeneration/pathology , Alkylating Agents , Animals , Calpain/metabolism , Caspases/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Humans , In Situ Nick-End Labeling , Injections, Intraperitoneal , Methylnitrosourea , Mice , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Degeneration/chemically induced , Retinal Degeneration/genetics , Transcription Factor CHOP/genetics
10.
J Biol Chem ; 290(6): 3488-99, 2015 Feb 06.
Article En | MEDLINE | ID: mdl-25477517

One-fifth of all cases of Leber congenital amaurosis are type 1 (LCA1). LCA1 is a severe form of retinal dystrophy caused by loss-of-function mutations in guanylate cyclase 1 (GC1), a key member of the phototransduction cascade involved in modulating the photocurrents. Although GC1 has been studied for some time, the mechanisms responsible for its regulation and membrane targeting are not fully understood. We reported earlier that retinal degeneration 3 (RD3) protein interacts with GC1 and promotes its targeting to the photoreceptor outer segments (POS). Here, we extend our studies to show a direct association between RD3 and guanylate cyclase activating protein 1 (GCAP1). Furthermore, we demonstrate that this functional interaction is important for GC1 targeting to POS. We also show that most LCA1-causing mutations in GC1 result in lost GC1 interaction with RD3 or GC1 being targeted to the plasma membrane. Our data suggest that GC1, GCAP1, and RD3 form a complex in the endoplasmic reticulum that targets GC1 to POS. Interruption of this assembly is likely the underlying mechanism for a subset of LCA1. This study offers insights for the development of therapeutic strategies to treat this severe form of blindness.


Guanylate Cyclase-Activating Proteins/metabolism , Guanylate Cyclase/metabolism , Leber Congenital Amaurosis/metabolism , Nuclear Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase-Activating Proteins/genetics , Humans , Leber Congenital Amaurosis/genetics , Mice , Nuclear Proteins/genetics , Protein Binding , Protein Transport , Receptors, Cell Surface/genetics , Retinal Photoreceptor Cell Outer Segment/metabolism
11.
Retina ; 32(3): 617-28, 2012 Mar.
Article En | MEDLINE | ID: mdl-21878836

PURPOSE: Rasagiline (N-propargyl-1(R)-aminoindan) is a second-generation propargylamine with neuroprotective effects. We used the Prph2/rds mouse to assess the effect of rasagiline on photoreceptor cell death and to examine the possible modulation of different pathways of programmed cell death. METHODS: The animals were orally treated with various doses of rasagiline from Postnatal Day 1 to 56. Methodological approaches consisted of morphometric analyses of the outer nuclear layer thickness and investigation of apoptotic events using TUNEL (TdT-mediated dUTP-biotin nick end labeling) assay, immunohistochemistry, and immunoblot staining. The expression of programmed cell death marker genes involved in photoreceptor degeneration was studied by quantitative real-time polymerase chain reaction. RESULTS: In the Prph2/rds mouse, treatment resulted in a significant dose-dependent neuroprotection at Postnatal Day 56 and a delay in the induction of apoptotic events at Postnatal Day 14. Programmed cell death marker gene expression showed that several mechanisms were involved in photoreceptor degeneration. Furthermore, rasagiline did not only target apoptosis but also other pathways such as autophagy and inflammation. CONCLUSION: This study showed for the first time significant neuroprotective effects of rasagiline in the retina of Prph2/rds mice through caspase-dependent pathways. However, the activation of caspase-independent programmed cell death pathways that are not affected by rasagiline eventually led to retinal degeneration, but in a delayed manner.


Indans/pharmacology , Neuroprotective Agents/pharmacology , Retinal Degeneration/drug therapy , Administration, Oral , Animals , Apoptosis/drug effects , Blotting, Western , Cell Death/drug effects , Disease Models, Animal , Immunohistochemistry , Indans/administration & dosage , Mice , Mice, Transgenic , Neuroprotective Agents/administration & dosage , Real-Time Polymerase Chain Reaction , Retinal Photoreceptor Cell Outer Segment/drug effects
12.
Graefes Arch Clin Exp Ophthalmol ; 249(6): 859-69, 2011 Jun.
Article En | MEDLINE | ID: mdl-21240523

BACKGROUND: Retinal degeneration is followed by significant changes in the structure and function of photoreceptors in humans and several genetic animal models. However, it is not clear whether similar changes occur when the degeneration is induced pharmacologically. Therefore, our aim was to investigate the influence of retinotoxic N-methyl-N-nitrosourea (MNU) on the function, morphology and underlying molecular pathways of programmed cell death. METHODS: C57/BL6 mice were injected with different doses of MNU, and function was determined by analysing optokinetic reflex measurements and cued water maze results at several time points post-injection. Morphometric measurements were also taken from H&E-stained paraffin eye sections. TUNEL-positive cells and caspase-3 and -6 were detected by immunohistochemistry. To assess the molecular changes leading to cell death, qRT-PCR from neurosensory retina mRNA was performed. RESULTS: The application of MNU led to an instant decrease in function and a delayed decrease in the thickness of the retinal outer nuclear layer. These responses were observed in the absence of any structural changes in the retinal pigment epithelium. The degeneration of the photoreceptor cell layer was highest with 60 mg/kg MNU. The assessment of TUNEL-positive cells visualised cell death after treatment, but no detectable caspase-3 activity was observed concomitant with these changes. qRT-PCR revealed the possible involvement of the inflammatory mediator caspase-1 and endoplasmic reticulum stress-mediated apoptosis by caspase-12. CONCLUSION: MNU leads to the dose-dependent degeneration of photoreceptor cells in mice by caspase-3-independent pathways and is, therefore, a suitable model to study retinal degeneration in an animal model.


Caspase 3/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/physiopathology , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Immunohistochemistry , In Situ Nick-End Labeling , Male , Methylnitrosourea/toxicity , Mice , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/enzymology , Psychomotor Performance/physiology , Retinal Degeneration/chemically induced , Retinal Degeneration/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Visual Acuity/physiology
13.
Invest Ophthalmol Vis Sci ; 50(8): 4004-10, 2009 Aug.
Article En | MEDLINE | ID: mdl-19339739

PURPOSE: To correlate damage to the retinal pigment epithelium (RPE) with decreased visual function after the systemic administration of sodium iodate (NaIO(3)). METHODS: Damage was produced in mice by injection of 15, 25, or 35 mg/kg NaIO(3). Visual function was assessed with the cued water maze (WM) behavioral test and the optokinetic reflex (OKR) measurement at different times after injection. Autofluorescence in whole eye flatmounts was quantified, and hematoxylin and eosin staining of paraffin sections was performed to assess changes in the outer retina. RESULTS: After 15 mg/kg NaIO(3), cued WM test results were normal, whereas OKR measurements were significantly decreased at all times. Focal RPE loss began on day 21, but no significant damage to the outer nuclear layer was observed. After 25 mg/kg NaIO(3), the cued WM test was transitionally reduced and the OKR measurement again decreased at all times. Large areas of RPE loss occurred on day 14 with a reduced outer nuclear layer on the same day. With 35 mg/kg NaIO(3), the cued WM test was reduced beginning on day 14 with complete obliteration of the OKR beginning on day 3, large areas of RPE loss on the same day, and a reduced outer nuclear layer on day 7. CONCLUSIONS: Stable, patchy RPE loss was observed with a low concentration of NaIO(3). The OKR measurement showed changes in visual function earlier than the cued WM test and before histologic findings were observed.


Iodates/toxicity , Retinal Diseases/chemically induced , Retinal Pigment Epithelium/drug effects , Vision Disorders/chemically induced , Visual Acuity/drug effects , Animals , Behavior, Animal , Injections, Intravenous , Iodates/administration & dosage , Male , Maze Learning , Mice , Mice, Inbred C57BL , Nystagmus, Optokinetic , Psychomotor Performance/drug effects , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/pathology , Vision Disorders/physiopathology , Visual Acuity/physiology
...