Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Klin Monbl Augenheilkd ; 234(3): 320-328, 2017 Mar.
Article in German | MEDLINE | ID: mdl-28355661

ABSTRACT

Characteristics of inherited retinal dystrophies include deficiencies in light perception and nervous conduction within the retina, leading to reduced vision or even blindness. In this context, the loss of function of photoreceptor-specific genes causes a variety of clinically and aetiologically distinct syndromes - each of them belonging to the group of rare diseases. With a prevalence of 1 in 2500, however, inherited retinal diseases are clinically significant and important - especially since these diseases lead to restrictions of a patient's fitness for work and overall quality of life. More than 250 genetic mutations causing the various types of inherited retinal dystrophies have been identified by now (https://sph.uth.tmc.edu/Retnet). In recent years, preclinical research on suitable animal models has yielded important progress in the understanding of the mutations underlying the pathological and molecular biological processes of these diseases. These findings have led to the development of novel and innovative therapeutic strategies for the treatment of inherited retinal dysfunctions, which are still incurable. Meanwhile, many of the successful preclinical studies have led to translational research projects aiming to find treatment options for human patients. However, some preliminary results of these human translational studies indicate the need to optimise and refine the underlying therapeutic concepts.


Subject(s)
Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Retinal Dystrophies/genetics , Retinal Dystrophies/therapy , Animals , Evidence-Based Medicine , Genetic Vectors/genetics , Humans , Treatment Outcome
2.
Cell Death Dis ; 5: e1234, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24853412

ABSTRACT

Hereditary retinal degeneration (RD) relates to a heterogeneous group of blinding human diseases in which the light sensitive neurons of the retina, the photoreceptors, die. RD is currently untreatable and the underlying cellular mechanisms remain poorly understood. However, the activity of the enzyme poly-ADP-ribose polymerase-1 (PARP1) and excessive generation of poly-ADP-ribose (PAR) polymers in photoreceptor nuclei have been shown to be causally involved in RD. The activity of PARP1 is to a large extent governed by its functional antagonist, poly-ADP-glycohydrolase (PARG), which thus also may have a role in RD. To investigate this, we analyzed PARG expression in the retina of wild-type (wt) mice and in the rd1 mouse model for human RD, and detected increased PARG protein in a subset of degenerating rd1 photoreceptors. Knockout (KO) animals lacking the 110 kDa nuclear PARG isoform were furthermore analyzed, and their retinal morphology and function were indistinguishable from wild-type animals. Organotypic wt retinal explants can be experimentally treated to induce rd1-like photoreceptor death, but PARG110 KO retinal explants were unexpectedly highly resistant to such treatment. The resistance was associated with decreased PAR accumulation and low PARP activity, indicating that PARG110 may positively regulate PARP1, an event that therefore is absent in PARG110 KO tissue. Our study demonstrates a causal involvement of PARG110 in the process of photoreceptor degeneration. Contrasting its anticipated role as a functional antagonist, absence of PARG110 correlated with low PARP activity, suggesting that PARG110 and PARP1 act in a positive feedback loop, which is especially active under pathologic conditions. This in turn highlights both PARG110 and PARP1 as potential targets for neuroprotective treatments for RD.


Subject(s)
Cyclic AMP/metabolism , Glycoside Hydrolases/deficiency , Nerve Degeneration , Photoreceptor Cells, Vertebrate/enzymology , Retinal Degeneration/enzymology , Animals , Cell Death , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Disease Models, Animal , Enzyme Activation , Genetic Predisposition to Disease , Glycoside Hydrolases/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , Phenotype , Phosphodiesterase Inhibitors/pharmacology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Protein Isoforms , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Time Factors , Tissue Culture Techniques
3.
Klin Monbl Augenheilkd ; 231(3): 232-40, 2014 Mar.
Article in German | MEDLINE | ID: mdl-24658860

ABSTRACT

Achromatopsia is an autosomal recessive inherited retinal disease caused by a complete loss of cone photoreceptor function. About 80 % of achromatopsia patients show mutations in the alpha or beta subunit (A3 and B3) of the cGMP controlled cation channel CNG (cyclic nucleotide-gated channel) of cone photoreceptors. Homologous to the human disease, CNGA3 deficient mice reveal a loss of cone specific functionality leading to degeneration of affected cone photoreceptors. The Institute for Ophthalmic Research in Tübingen has now succeeded in curing achromatopsia ACHM2 in an animal model. In this article, we explain the recombinant adeno-associated virus-based approach in detail. Furthermore, applied non-invasive diagnostic techniques for quality and success control, ERG, SLO and OCT, are described. The success of the therapy is indicated by a restored cone photoreceptor function as well as the neuronal processing of retinal signals resulting in a specific, cone-mediated behaviour. The outstanding results derived from the animal model are the starting point for the first human translation of a gene therapy for achromatopsia in Germany.


Subject(s)
Color Vision Defects/genetics , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Genetic Therapy/methods , Transfection/methods , Animals , Color Vision Defects/diagnosis , Evidence-Based Medicine , Humans , Mice , Treatment Outcome
4.
Mol Vis ; 17: 2934-46, 2011.
Article in English | MEDLINE | ID: mdl-22128240

ABSTRACT

PURPOSE: Rho GTPases such as RAS-related C3 botulinum substrate 1 (RAC1) and cell division cycle 42 homolog (S. cerevisiae; CDC42) have been linked to cellular processes including movement, development, and apoptosis. Recently, RAC1 has been shown to be a pro-apoptotic factor in the retina during light-induced photoreceptor degeneration. Here, we analyzed the role of CDC42 in the degenerating retina. METHODS: Photoreceptor degeneration was studied in a mouse model for autosomal dominant retinitis pigmentosa (VPP) with or without a rod-specific knockdown of Cdc42, as well as in wild-type and Cdc42 knockdown mice after light exposure. Gene and protein expression were analyzed by real-time PCR, western blotting, and immunofluorescence. Retinal morphology and function were assessed by light microscopy and electroretinography, respectively. RESULTS: CDC42 accumulated in the perinuclear region of terminal deoxynucleotidyl transferase dUTP nick end labeling-negative photoreceptors during retinal degeneration induced by excessive light exposure and in the rd1, rd10, and VPP mouse models of retinitis pigmentosa. The knockdown of Cdc42 did not affect retinal morphology or function in the adult mice and did not influence photoreceptor apoptosis or molecular signaling during induced and inherited retinal degeneration. CONCLUSIONS: Retinal degeneration induces the accumulation of CDC42 in the perinuclear region of photoreceptors. In contrast to RAC1, however, lack of CDC42 does not affect the progression of degeneration. CDC42 is also dispensable for normal morphology and function of adult rod photoreceptor cells. RECEIVED: May 25, 2011 ACCEPTED: November 10, 2011.


Subject(s)
GTPase-Activating Proteins , Gene Expression/radiation effects , Retina/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Animals , Apoptosis , Blotting, Western , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Disease Models, Animal , Electroretinography , Fluorescent Antibody Technique , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , In Situ Nick-End Labeling , Light/adverse effects , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Retina/pathology , Retina/radiation effects , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/radiation effects , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/genetics , Rhodopsin/metabolism
5.
Neuroscience ; 169(4): 1815-30, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20600653

ABSTRACT

Cone photoreceptor breakdown underlies functional vision loss in many blinding diseases. Cone loss is often secondary to that of rods, but little experimental data are available on the relationship between the two populations. Because of its high cone numbers, we used the diurnal rodent Arvicanthis ansorgei to explore changes in rod and cone survival and function during chemically-induced retinal degeneration. Adult animals received intraperitoneal injections of N-methyl-N-nitrosourea (MNU), and changes in retinal fundus appearance, histology, phenotype, apoptosis (TUNEL staining) and functionality (scotopic and photopic electroretinography) were monitored as a function of post-treatment time and retinal topography. Relative to control animals injected with vehicle only, MNU-injected animals showed time-, region- and population-specific changes as measured by morphological and immunochemical criteria. Histological (gradual thinning of photoreceptor layer) and phenotypical (reduced immunostaining of rhodopsin and rod transducin, and mid wavelength cone opsin and cone arrestin) modifications were first observed in superior central retina at 11 days post-injection. These degenerative changes spread into the superior peripheral and inferior hemisphere during the following 10 days. Rod loss preceded that of cones as visualized by differential immunolabelling and presence of apoptotic cells in rod but not cone cells. By 3 months post-injection, degeneration of the photoreceptor layer was complete in the superior hemisphere, but only partial in the inferior hemisphere. Despite the persistence of cone photoreceptors, scotopic and photopic electroretinography performed at 90 days post-treatment showed that both rod and cone function were severely compromised. In conclusion, MNU-induced retinal degeneration in Arvicanthis follows a predictable spatial and temporal pattern allowing clear separation of rod- and cone-specific pathogenic mechanisms. Compared to other rodents in which MNU has been used, Arvicanthis ansorgei demonstrates pronounced resistance to photoreceptor cell loss.


Subject(s)
Muridae , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/pathology , Animals , Circadian Rhythm/physiology , Disease Models, Animal , Male
6.
Cell Mol Life Sci ; 67(18): 3173-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20467778

ABSTRACT

Cone dystrophies are genetic diseases characterized by loss of cone photoreceptor function and severe impairment of daylight vision. Loss of function is accompanied by a progressive degeneration of cones limiting potential therapeutic interventions. In this study we combined microarray-based gene-expression analysis with electroretinography and immunohistochemistry to characterize the pathological processes in the cone photoreceptor function loss 1 (cpfl1) mouse model. The cpfl1-mouse is a naturally arising mouse mutant with a loss-of-function mutation in the cone-specific Pde6c gene. Cpfl1-mice displayed normal rod-specific light responses while cone-specific responses were strongly diminished. Despite the lack of a general retinal degeneration, the cone-specific functional defect resulted in a marked activation of GFAP, a hallmark of Müller-cell gliosis. Microarray-based network-analysis confirmed activation of Müller-glia-specific transcripts. Unexpectedly, we found up-regulation of the cytokine LIF and the anti-apoptotic transcription factor STAT3 in cpfl1 cone photoreceptors. We postulate that STAT3-related pathways are induced in cpfl1 cone photoreceptors to counteract degeneration.


Subject(s)
Gene Expression Regulation , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , STAT3 Transcription Factor/metabolism , Animals , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology
7.
Graefes Arch Clin Exp Ophthalmol ; 248(8): 1063-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20012642

ABSTRACT

BACKGROUND: Bone spicule pigments (BSP) are a hallmark of retinitis pigmentosa (RP). In this study, we examined the process of BSP formation in the rhodopsin knockout (rho (-/-)) mouse, a murine model for human RP. METHODS: In rho (-/-) mice from 2 to 16 months of age, representing the range from early to late stages of degeneration, retinal sections and whole mounts were examined morphologically by light and electron microscopy. The results were compared to scanning laser ophthalmoscopy of BSP degeneration in human RP. RESULTS: After the loss of all photoreceptor cells in rho-/- mice, the outer retina successively degenerated, leading to approximation and finally a direct contact of inner retinal vessels and the retinal pigment epithelium (RPE). We could show that it was the event of proximity of retinal vessel and RPE that triggered migration of RPE cells along the contacting vessels towards the inner retina. Ultrastructurally, these mislocalized RPE cells partially sealed the vessels by tight junction linkage and deposited extracellular matrix perivascularly. Also, the vascular endothelium developed fenestrations similar to the RPE-choroid interface. In whole mounts, the pigmented cell clusters outlining retinal capillaries correlated well with BSPs in human RP. The structure of the inner retina remained well preserved, even in late stages. CONCLUSIONS: The Rho (-/-) mouse is the first animal model that depicts all major pathological changes, even in the late stages of RP. Using the rho (-/-) mouse model we were able to analyze the complete dynamic process of BSP formation. Therefore we conclude that: (1) In rho (-/-) retinas, BSPs only form in areas devoid of photoreceptors; (2) Direct contact between inner retinal vessels and RPE appears to be a major trigger for migration of RPE cells; (3) The distribution of the RPE cells in BSPs reflects the vascular network at the time of formation. The similarity of the disease process between mouse and human and the possibility to study all consecutive steps of the course of the disease makes the rho (-/-) mouse valuable for further insights in the dynamics of BSP formation in human RP.


Subject(s)
Disease Models, Animal , Photoreceptor Cells, Vertebrate/ultrastructure , Retinitis Pigmentosa/pathology , Animals , Cell Movement , Fluorescent Antibody Technique, Indirect , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Retinal Vessels/metabolism , Retinal Vessels/ultrastructure , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Tomography, Optical Coherence
8.
Ophthalmologe ; 106(6): 505-11, 2009 Jun.
Article in German | MEDLINE | ID: mdl-19517115

ABSTRACT

Usher syndrome denotes a clinically and genetically heterogeneous combination of retinitis pigmentosa and sensorineural deafness. The division into subtypes I, II, and III is based on the degree of hearing loss: Type I is characterized by deafness from birth together with ataxia and retarded motor development, type II by a stationary deafness of a moderate degree, and type III by a progressive deafness with adult onset. In Germany, Usher syndrome currently bears particular relevance because in January 2009 a new compulsory screening of auditory function in newborn infants was introduced. Consequently, it can be expected that a higher number of patients with Usher syndrome will be identified in early childhood and referred to ophthalmologists. The focus of this work is to introduce the typical clinical picture of Usher syndrome, summarize diagnostic options, and give an overview of therapeutic strategies.


Subject(s)
Usher Syndromes/diagnosis , Usher Syndromes/therapy , Humans
9.
Br J Ophthalmol ; 92(2): 272-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18227206

ABSTRACT

AIMS: To provide evidence for a novel route of gene administration to normal and diseased retinas, we performed systemic transplantation of genetically engineered bone marrow-derived cells (BMDCs) to wild-type mice and to mutant mice with retinal degeneration. METHODS: Lethally irradiated recipient mice-C57BL/6 (wild-type), SCA7 (spinocerebellar ataxia type 7) and FVB/N (rd1 mutant)-were transplanted intravenously with 5x106 BMDCs, which were transduced with a retroviral vector to express the enhanced green fluorescent protein (GFP). Chimeras were killed at 1, 3, 8, 11, 12 and 15 months (wild-type) or at 8 and 12 months (mutants) after transplantation. Eyes were enucleated, and the retinas were analysed using immunohistochemistry. RESULTS: In wild-type retinas, BMDCs preferentially engrafted in the inner and outer plexiform layers, the ganglion cell layer and the optic nerve. No BMDCs were found in the photoreceptor layer. BMDCs were more common in the degenerating retinas of the mutant mice. The majority of BMDCs in the retina were identified as microglia based on morphology and immunophenotype. Approximately 8-16% of all CD11b(+) cells in the retina expressed GFP. None of the BMDCs expressed neuronal cell markers. GFP-expressing BMDCs were found to persist for more than 1 year after transplantation. CONCLUSIONS: We demonstrate that gene-modified BMDCs show long-term engraftment and stable expression of GFP from a retrovirus in both wild-type and mutant mouse retinas. Thus, BMDCs may be used as vehicles for gene delivery to the retina.


Subject(s)
Bone Marrow Transplantation/methods , Gene Transfer Techniques , Genetic Engineering/methods , Retinal Degeneration/therapy , Animals , Graft Survival , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Retina/metabolism , Retinal Degeneration/metabolism , Transduction, Genetic
11.
Neuroscience ; 107(1): 127-42, 2001.
Article in English | MEDLINE | ID: mdl-11744253

ABSTRACT

The abundance of the integral membrane protein synaptophysin in synaptic vesicles and its multiple possible functional contributions to transmitter exocytosis and synaptic vesicle formation stand in sharp contrast to the observed lack of defects in synaptophysin knockout mice. Assuming that deficiencies are compensated by the often coexpressed synaptophysin isoform synaptoporin, we now show that retinal rod photoreceptors, which do not synthesize synaptoporin either in wild-type or in knockout mice, are affected by the loss of synaptophysin. Multiple pale-appearing photoreceptors, as seen by electron microscopy, possess reduced cytoplasmic electron density, swollen mitochondria, an enlarged cell surface area, and, most importantly, a significantly reduced number of synaptic vesicles with an unusually bright interior. Quantification of the number of synaptic vesicles per unit area, not only in these, but also in all other rod terminals of knockout animals, reveals a considerable reduction in vesicles that is even more pronounced during the dark period, i.e., at times of highest synaptic activity. Moreover, activity-dependent reduction in synaptic vesicle diameter, typically occurring in wild-type mice, is not detected in knockout animals. The large number of clathrin-coated pits and vesicles in dark-adapted synaptophysin knockout mice is taken as an indication of compensatory usage of synaptophysin-independent pathway(s), and, conversely, in view of the overall reduction in the number of synaptic vesicles, as an indication for the presence of another synaptophysin-dependent synaptic vesicle recycling pathway. Our results provide in vivo evidence for the importance of the integral membrane protein synaptophysin for synaptic vesicle recycling and formation.


Subject(s)
Action Potentials/genetics , Exocytosis/genetics , Protein Transport/genetics , Retinal Rod Photoreceptor Cells/abnormalities , Synaptic Transmission/genetics , Synaptic Vesicles/pathology , Synaptophysin/deficiency , Animals , Clathrin-Coated Vesicles/pathology , Clathrin-Coated Vesicles/ultrastructure , Dark Adaptation/genetics , Electroretinography , Female , Fluorescent Antibody Technique , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Photic Stimulation , Presynaptic Terminals/pathology , Presynaptic Terminals/ultrastructure , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/ultrastructure , Sex Characteristics , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptophysin/genetics
12.
Invest Ophthalmol Vis Sci ; 42(12): 3066-71, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11687556

ABSTRACT

PURPOSE: Usher syndrome (US) is a recessively inherited disorder combining retinitis pigmentosa (RP) and a sensorineural hearing loss. The classification in subtypes is based mainly on auditory tests. The purpose of this study was to analyze implicit time (IT) differences in the electroretinogram (ERG) between RP alone, US I, and US II. METHODS: The data of 15 control subjects and of 15 patients with US I, 15 with US II, and 15 with RP with nonzero 33-Hz flicker ERG responses were analyzed. The ITs of three signal peaks (P1-P3) were evaluated. Sensitivity and specificity of a test to distinguish between US I and II based on timing differences were determined. Multifocal (mf)ERGs were used to assess differences in disease topography. RESULTS: Despite the similar amplitude loss with retinal eccentricity in the mfERG in all three groups, the peak delay in US I was negligible compared with that in US II and RP. In the flicker ERG data, US I and control subjects had almost identical peak times, and the same was true for subjects with US II and RP. Because of the slight overlap between US I and II, the diagnostic test achieved a sensitivity of 100% and a specificity of 93.3%. CONCLUSIONS: Substantial timing differences between US I and II and their usefulness for a diagnostic test were demonstrated. This finding may also be the basis for further investigations regarding the structural differences of retinal impairment between US I and II on a cellular level.


Subject(s)
Electroretinography/methods , Hearing Loss, Sensorineural/diagnosis , Retinitis Pigmentosa/diagnosis , Adolescent , Adult , Child , Hearing Loss, Sensorineural/classification , Hearing Loss, Sensorineural/physiopathology , Humans , Middle Aged , Retina/physiology , Retinitis Pigmentosa/classification , Retinitis Pigmentosa/physiopathology , Retrospective Studies , Sensitivity and Specificity , Syndrome , Time Factors
13.
Nat Genet ; 29(1): 70-4, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528395

ABSTRACT

Leber congenital amaurosis (LCA) is the most serious form of the autosomal recessive childhood-onset retinal dystrophies. Mutations in the gene encoding RPE65, a protein vital for regeneration of the visual pigment rhodopsin in the retinal pigment epithelium, account for 10-15% of LCA cases. Whereas previous studies of RPE65 deficiency in both animal models and patients attributed remaining visual function to cones, we show here that light-evoked retinal responses in fact originate from rods. For this purpose, we selectively impaired either rod or cone function in Rpe65-/- mice by generating double- mutant mice with models of pure cone function (rhodopsin-deficient mice; Rho-/-) and pure rod function (cyclic nucleotide-gated channel alpha3-deficient mice; Cnga3-/-). The electroretinograms (ERGs) of Rpe65-/- and Rpe65-/-Cnga3-/- mice were almost identical, whereas there was no assessable response in Rpe65-/-Rho-/- mice. Thus, we conclude that the rod system is the source of vision in RPE65 deficiency. Furthermore, we found that lack of RPE65 enables rods to mimic cone function by responding under normally cone-isolating lighting conditions. We propose as a mechanism decreased rod sensitivity due to a reduction in rhodopsin content to less than 1%. In general, the dissection of pathophysiological processes in animal models through the introduction of additional, selective mutations is a promising concept in functional genetics.


Subject(s)
Optic Atrophies, Hereditary/genetics , Pigment Epithelium of Eye/physiology , Proteins/genetics , Retinal Rod Photoreceptor Cells/physiopathology , Vision, Ocular/physiology , Animals , Carrier Proteins , Disease Models, Animal , Electroretinography , Eye Proteins , Mice , Mice, Mutant Strains , Optic Atrophies, Hereditary/physiopathology , cis-trans-Isomerases
14.
Invest Ophthalmol Vis Sci ; 42(7): 1653-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11381074

ABSTRACT

PURPOSE: Evidence has accumulated that excessive light exposure may promote age-related and inherited retinal degeneration, in which photoreceptor death by apoptosis leads to loss of vision. In the current study, the effect of elevated corticosteroid levels on light-induced apoptosis of photoreceptors was determined. METHODS: Photoreceptor apoptosis was induced in retinas of BALB/c mice by exposure to diffuse white light. High levels of corticosteroids were induced, either endogenously (fasting-mediated stress) or by a single intraperitoneal injection of dexamethasone (DEX). Photoreceptor damage was assessed morphologically and by electroretinography. Glucocorticoid receptor (GR) and activator protein (AP)-1 activities were shown by Western blot analysis and electrophoretic mobility shift assay (EMSA) of retinal nuclear extracts. RESULTS: Fasting and injection of DEX led to an activation of GR in the retina, as judged by its translocation to the nucleus of retinal cells. On induction of GR activity before light exposure, AP-1 activity, normally induced by damaging doses of light, remained at basal levels. Both treatments completely prevented photoreceptor apoptosis and preserved retinal function. CONCLUSIONS: Activity of the transcription factor AP-1 is associated with light-induced apoptosis. In the current study, pharmacologic suppression of AP-1 activity protected against light damage. Inhibition of AP-1 activity may have occurred by the protein-protein interaction of GR and AP-1.


Subject(s)
Apoptosis , Light/adverse effects , Photoreceptor Cells, Vertebrate/radiation effects , Radiation Injuries, Experimental/prevention & control , Receptors, Glucocorticoid/metabolism , Retinal Degeneration/prevention & control , Animals , Apoptosis/radiation effects , Blotting, Western , Corticosterone/blood , Dexamethasone/pharmacology , Electroretinography , Food Deprivation , Glucocorticoids/pharmacology , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism
15.
Invest Ophthalmol Vis Sci ; 42(2): 506-13, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157890

ABSTRACT

PURPOSE: To determine a time window in the rhodopsin knockout (Rho(-/-)) mouse during which retinal function is already sufficiently developed but cone degeneration is not yet substantial, thus representing an all-cone retina. METHODS: Electroretinograms (ERGs) were obtained from 14 homozygous Rho(-/-) mice and eight C57Bl/6 control mice. The same individuals were tested every 7 days, beginning as early as postnatal day (P)14. The ERG protocols included flash and flicker stimuli, both under photopic and scotopic conditions. Retinal and choroidal morphology was observed in animals of comparable age. RESULTS: Functionally, the developmental phase lasted until postnatal week (PW)3 in both the Rho(-/-) mice and the control animals. During PW4 to 6, the Rho(-/-) mice showed a plateau in ERG parameters with normal or even supernormal cone responses and complete absence of rod contributions. At PW7, there was a marked onset of degeneration, which progressed so that no ERG signals were left at PW13, when the control eyes still had normal ERG responses. Microscopically, cone degeneration paralleled the functional changes, beginning at approximately PW6 and almost complete at PW13, whereas retinal pigment epithelium (RPE) and choroid did not show any abnormalities. CONCLUSIONS: From PW4 to 6, Rho(-/-) mice appear to have normal cone and no rod function. Despite the missing rod outer segment (OS), the structure of retina, RPE, and choroid remained unchanged. Therefore, the Rho(-/-) mice can serve during this age period as a model for pure cone function. Such a model is particularly useful to evaluate rod-cone interaction and to dissect rod- from cone-mediated signaling pathways in vivo.


Subject(s)
Electroretinography , Models, Animal , Retinal Cone Photoreceptor Cells/physiology , Rhodopsin/physiology , Animals , Animals, Newborn , Evaluation Studies as Topic , Fundus Oculi , Mice , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation , Photography , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Time Factors
16.
Invest Ophthalmol Vis Sci ; 41(7): 1998-2005, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10845627

ABSTRACT

PURPOSE: To establish a method for the recording of multifocal electroretinograms (MF-ERGs) in animals under fundus control using a scanning-laser ophthalmoscope (SLO) and to analyze the spatial distribution of disease in a strain of Abyssinian cats with a recessively inherited rod-cone degeneration (ARCD). METHODS: Four normal and 12 Abyssinian cats at four different clinical stages of ARCD were examined with the RETIscan MF-ERG system using 61 hexagonal elements within a visual field of approximately 30 degrees radius. The stimulus pattern was generated by the green laser beam (515 nm) of a Heidelberg Engineering HRA SLO, whose power was reduced with a Schott long-pass filter allowing for simultaneous infrared fundus imaging. RESULTS: Topographical recordings could be obtained in all animals except one in stage 4. Amplitudes were minimal at the optic disc and had a slight maximum at the area centralis. Implicit times had a tendency to lower values in the central region, most pronounced in progressed stages of ARCD. The clinical stages of ARCD correlated with a successive generalized loss of amplitude and a rise in implicit time. Without a decrease in retinal illuminance, topographical landmarks like the optic disc were no longer detectable, pointing to stray light as a possible cause. CONCLUSIONS: It was demonstrated that topographical MF-ERG recordings can be obtained in an animal model under fundus control using SLO stimulation. The appearance of retinal landmarks was found to be dependent on sufficient attenuation of laser power. Because the changes in ARCD are more patchy than in human retinitis pigmentosa (RP), a generalized loss of function was detected. However, like in RP, the central area was found to retain a better function than the periphery, especially in later stages of the disease. In summary, fundus controlled methods like the one presented will greatly improve the reliability of MF-ERG in future research on glaucoma, transplantation studies, and evaluation of gene therapy.


Subject(s)
Cat Diseases/physiopathology , Disease Models, Animal , Electroretinography/veterinary , Eye Diseases, Hereditary/veterinary , Photoreceptor Cells, Vertebrate/physiology , Retinal Degeneration/veterinary , Animals , Cat Diseases/diagnosis , Cats , Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/physiopathology , Lasers , Ophthalmoscopes/veterinary , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/diagnosis , Retinal Degeneration/physiopathology
17.
Doc Ophthalmol ; 100(2-3): 167-84, 2000.
Article in English | MEDLINE | ID: mdl-11142745

ABSTRACT

INTRODUCTION: Multifocal electroretinography (MF-ERG) is widely used in the detection of local retinal dysfunction. However, the position of the stimulus on the retina and the stability of fixation are usually not directly accessible. Thus, devices have been designed for a continuous fundus visualization during recording. METHODS: MF-ERGs were recorded with a RetiScan system connected to two different Scanning-laser ophthalmoscopes (SLOs) that use either a red (633 nm) or green (415 nm) laser for stimulation, and a VERIS 4 system connected to a piggyback stimulator prototype that added the stimulus to the optical pathway of the SLO by means of a wavelength-sensitive mirror. Fundus visualization was achieved with the infrared lasers of the SLOs (780 and 835 nm). RESULTS: The most extensive study so far with a green laser stimulus in a cat model of retinal degeneration demonstrated the capability of the device to detect retinal landmarks and the different stages of degeneration. Also, the advantages of exactly reproducible stimulus positioning for averaging within and comparison between disease groups became apparent. The results with the same setup in transgenic mice suggest a pure cone origin of the responses. In humans, recordings show that fixation is sufficiently good in most subjects. It is not clear yet whether red or green laser stimulation (or both) is preferable. The results with the prototype were very similar to the MF-ERGs obtained with a standard CRT screen. CONCLUSIONS: All three devices allowed us to record MF-ERGs with continuous fundus monitoring. Although further refinements are necessary, it is obvious that fundus controlled methods will improve the reliability of MF-ERG in future research on glaucoma, transplantation studies, and evaluation of gene therapy.


Subject(s)
Electroretinography/methods , Ophthalmoscopy/methods , Retina/pathology , Retinal Degeneration/diagnosis , Animals , Cats , Diagnostic Imaging/methods , Fundus Oculi , Humans , Mice , Mice, Transgenic , Photic Stimulation , Retina/physiopathology , Retinal Degeneration/physiopathology , Visual Field Tests/methods , Visual Fields
18.
J Neurosci ; 19(19): 8603-15, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10493760

ABSTRACT

We examined optokinetic and optomotor responses of 450 zebrafish mutants, which were isolated previously based on defects in organ formation, tissue patterning, pigmentation, axon guidance, or other visible phenotypes. These strains carry single point mutations in >400 essential loci. We asked which fraction of the mutants develop blindness or other types of impairments specific to the visual system. Twelve mutants failed to respond in either one or both of our assays. Subsequent histological and electroretinographic analysis revealed unique deficits at various stages of the visual pathway, including lens degeneration (bumper), melanin deficiency (sandy), lack of ganglion cells (lakritz), ipsilateral misrouting of axons (belladonna), optic-nerve disorganization (grumpy and sleepy), inner nuclear layer or outer plexiform layer malfunction (noir, dropje, and possibly steifftier), and disruption of retinotectal impulse activity (macho and blumenkohl). Surprisingly, mutants with abnormally large or small eyes or severe wiring defects frequently exhibit no discernible behavioral deficits. In addition, we identified 13 blind mutants that display outer-retina dystrophy, making this syndrome the single-most common cause of inherited blindness in zebrafish. Our screen showed that a significant fraction (approximately 5%) of the essential loci also participate in visual functions but did not reveal any systematic genetic linkage to particular morphological traits. The mutations uncovered by our behavioral assays provide distinct entry points for the study of visual pathways and set the stage for a genetic dissection of vertebrate vision.


Subject(s)
Fish Diseases/genetics , Mutation , Vision Disorders/veterinary , Visual Pathways/physiopathology , Zebrafish/genetics , Albinism/genetics , Albinism/veterinary , Alleles , Animals , Axons/physiology , Blindness/genetics , Blindness/veterinary , Fish Diseases/physiopathology , Lens, Crystalline/pathology , Lens, Crystalline/physiopathology , Melanins/deficiency , Nystagmus, Optokinetic , Vision Disorders/genetics , Vision Disorders/physiopathology , Visual Pathways/pathology
19.
Am J Clin Nutr ; 69(5): 931-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10232633

ABSTRACT

BACKGROUND: Two German sisters aged 14 and 17 y were admitted to the Tübingen eye hospital with a history of night blindness. In both siblings, plasma retinol binding protein (RBP) concentrations were below the limit of detection (<0.6 micromol/L) and plasma retinol concentrations were extremely low (0.19 micromol/L). Interestingly, intestinal absorption of retinyl esters was normal. In addition, other factors associated with low retinol concentrations (eg, low plasma transthyretin or zinc concentrations or mutations in the transthyretin gene) were not present. Neither sibling had a history of systemic disease. OBJECTIVE: Our aim was to investigate the cause of the retinol deficiency in these 2 siblings. DESIGN: The 2 siblings and their mother were examined clinically, including administration of the relative-dose-response test, DNA sequencing of the RBP gene, and routine laboratory testing. RESULTS: Genomic DNA sequence analysis revealed 2 point mutations in the RBP gene: a T-to-A substitution at nucleotide 1282 of exon 3 and a G-to-A substitution at nucleotide 1549 of exon 4. These mutations resulted in amino acid substitutions of asparagine for isoleucine at position 41 (Ile41-->Asn) and of aspartate for glycine at position 74 (Gly74-->Asp). Sequence analysis of cloned polymerase chain reaction products spanning exons 3 and 4 showed that these mutations were localized on different alleles. The genetic defect induced severe biochemical vitamin A deficiency but only mild clinical symptoms (night blindness and a modest retinal dystrophy without effects on growth). CONCLUSIONS: We conclude that the cellular supply of vitamin A to target tissues might be bypassed in these siblings via circulating retinyl esters, beta-carotene, or retinoic acid, thereby maintaining the health of peripheral tissues.


Subject(s)
Retinol-Binding Proteins/genetics , Vitamin A Deficiency/genetics , Adolescent , Base Sequence , Diterpenes , Exons , Female , Humans , Molecular Sequence Data , Nuclear Family , Point Mutation , Prealbumin/chemistry , Prealbumin/genetics , Retinol-Binding Proteins/metabolism , Retinol-Binding Proteins, Plasma , Retinyl Esters , Vitamin A/analogs & derivatives , Vitamin A/blood , Vitamin A/pharmacology , Vitamin A Deficiency/blood , Vitamin A Deficiency/metabolism , Zinc/blood
20.
Invest Ophthalmol Vis Sci ; 40(1): 3-11, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888420

ABSTRACT

PURPOSE: To describe the phenotype caused by a retinol deficiency in a family with compound heterozygous missense mutations (Ile41Asn and Gly75Asp) in the gene for serum retinol binding protein (RBP). METHODS: The two affected sisters, 17 (BR) and 13 (MR) years old, were examined clinically and with perimetry, color vision tests, dark adaptometry, rod- and cone-isolated electroretinograms (ERGs), multifocal ERGs, electrooculograms (EOGs), and laboratory tests. RESULTS: There were no complaints besides night vision problems and no history of systemic disease. Visual acuity was reduced to 20/40 (BR) and 20/25 (MR). Anterior segments were normal except for a discrete iris coloboma. Both patients showed a typical "fundus xerophthalmicus," featuring a progressed atrophy of the retinal pigment epithelium. Dark adaptation thresholds were elevated. In the scotopic ERG, only reduced mixed responses were recordable. The photopic ERG was reduced in BR and normal in MR; implicit times were highly (BR) to slightly (MR) elevated. There was no (BR) to little (MR) light reaction in the EOG. All-trans retinol levels were 0.19 microM and 0.18 microM (normal range, 0.7-1.5 microM) for BR and MR, respectively, and did not increase in a dose-response test. RBP was below detection threshold, and retinyl esters were normal. CONCLUSIONS: Both affected siblings had no detectable serum RBP, one sixth of normal retinol levels, and normal retinyl esters. The retinal pigment epithelium was severely affected, but besides acne there were no changes to other organs. This gives evidence for an alternative tissue source of vitamin A, presumably retinyl esters from chylomicron remnants. The normal retinol levels in the tear fluid explain the lack of xerophthalmia. However, considering the role of RBP in the tear fluid and, during development, in the yolk sac there is also evidence that there are organ-specific RBP forms not affected by the genetic defect.


Subject(s)
Avitaminosis/genetics , Mutation, Missense , Retinol-Binding Proteins/genetics , Adolescent , Atrophy , Avitaminosis/pathology , Avitaminosis/physiopathology , Coloboma/pathology , Color Perception Tests , Dark Adaptation , Electrooculography , Electroretinography , Female , Humans , Iris/abnormalities , Night Blindness/genetics , Night Blindness/pathology , Night Blindness/physiopathology , Nuclear Family , Phenotype , Photoreceptor Cells, Vertebrate/physiology , Pigment Epithelium of Eye/pathology , Retinol-Binding Proteins/biosynthesis , Visual Acuity , Visual Field Tests , Vitamin A/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...