Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Exp Eye Res ; 187: 107755, 2019 10.
Article in English | MEDLINE | ID: mdl-31408630

ABSTRACT

Dark-rearing has been found to slow the rate of retinal degeneration in albino P23H but not S334ter mutant rhodopsin transgenic (Tg) rats. Since eye pigmentation has the same protective slowing effect as dark-rearing in RCS rats, we examined whether eye pigmentation has a comparable slowing effect in the different mutant rhodopsin Tg rats. Different lines of albino P23H and S334ter Tg rats on the Sprague-Dawley (SD) background were bred to Long-Evans (LE) rats to produce pigmented Tg rats. These were compared to albino Tg rats at postnatal days of different ages using the outer nuclear layer (ONL) as a morphological measure of photoreceptor number and electroretinogram (ERG) a- and b-wave amplitudes as a measure of retinal function. When compared to albino P23H rats, pigmented P23H rats had a slower rate of degeneration as measured by greater ONL thicknesses and greater ERG a- and b-wave amplitudes. By contrast, pigmented S334ter rats showed no difference in ONL thicknesses or ERG a- and b-wave amplitudes when compared to their albino equivalents. Thus, degeneration of photoreceptors in P23H Tg rats is slowed by eye pigmentation as measured by ONL thickness, while it is not in the S334ter Tg rats. Eye pigmentation also protects functional changes in ERG a- and b-waves for the P23H lines, but not for the S334ter lines.


Subject(s)
Eye Color/genetics , Retina/physiopathology , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Rhodopsin/genetics , Animals , Electroretinography , Mutation , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Transgenic
2.
Exp Eye Res ; 167: 56-90, 2018 02.
Article in English | MEDLINE | ID: mdl-29122605

ABSTRACT

We produced 8 lines of transgenic (Tg) rats expressing one of two different rhodopsin mutations in albino Sprague-Dawley (SD) rats. Three lines were generated with a proline to histidine substitution at codon 23 (P23H), the most common autosomal dominant form of retinitis pigmentosa in the United States. Five lines were generated with a termination codon at position 334 (S334ter), resulting in a C-terminal truncated opsin protein lacking the last 15 amino acid residues and containing all of the phosphorylation sites involved in rhodopsin deactivation, as well as the terminal QVAPA residues important for rhodopsin deactivation and trafficking. The rates of photoreceptor (PR) degeneration in these models vary in proportion to the ratio of mutant to wild-type rhodopsin. The models have been widely studied, but many aspects of their phenotypes have not been described. Here we present a comprehensive study of the 8 Tg lines, including the time course of PR degeneration from the onset to one year of age, retinal structure by light and electron microscopy (EM), hemispheric asymmetry and gradients of rod and cone degeneration, rhodopsin content, gene dosage effect, rapid activation and invasion of the outer retina by presumptive microglia, rod outer segment disc shedding and phagocytosis by the retinal pigmented epithelium (RPE), and retinal function by the electroretinogram (ERG). The biphasic nature of PR cell death was noted, as was the lack of an injury-induced protective response in the rat models. EM analysis revealed the accumulation of submicron vesicular structures in the interphotoreceptor space during the peak period of PR outer segment degeneration in the S334ter lines. This is likely due to the elimination of the trafficking consensus domain as seen before as with other rhodopsin mutants lacking the C-terminal QVAPA. The 8 rhodopsin Tg lines have been, and will continue to be, extremely useful models for the experimental study of inherited retinal degenerations.


Subject(s)
Disease Models, Animal , Photoreceptor Cells, Vertebrate/pathology , Point Mutation , Retina/physiology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/genetics , Animals , Electroretinography , Microscopy , Microscopy, Electron , Phenotype , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Degeneration/physiopathology
3.
Exp Eye Res ; 165: 175-181, 2017 12.
Article in English | MEDLINE | ID: mdl-28974356

ABSTRACT

Retinal degenerations, including age-related macular degeneration and the retinitis pigmentosa family of diseases, are among the leading causes of legal blindness in the United States. We previously found that Stanniocalcin-1 (STC-1) reduced photoreceptor loss in the S334ter-3 and Royal College of Surgeons rat models of retinal degeneration. The results were attributed in part to a reduction in oxidative stress. Herein, we tested the hypothesis that long-term delivery of STC-1 would provide therapeutic rescue in more chronic models of retinal degeneration. To achieve sustained delivery, we produced an adeno-associated virus (AAV) construct to express STC-1 (AAV-STC-1) under the control of a retinal ganglion cell targeting promoter human synapsin 1 (hSYN1). AAV-STC-1 was injected intravitreally into the P23H-1 and S334ter-4 rhodopsin transgenic rats at postnatal day 10. Tissues were collected at postnatal day 120 for confirmation of STC-1 overexpression and histologic and molecular analysis. Electroretinography (ERG) was performed in a cohort of animals at that time. Overexpression of STC-1 resulted in a significant preservation of photoreceptors as assessed by outer nuclear thickness in the P23H-1 (P < 0.05) and the S334ter-4 (P < 0.005) models compared to controls. Additionally, retinal function was significantly improved in the P23H-1 model with overexpressed STC-1 as assessed by ERG analysis (scotopic b-wave P < 0.005 and photopic b-wave P < 0.05). Microarray analysis identified common downstream gene expression changes that occurred in both models. Genes of interest based on their function were selected for validation by quantitative real-time PCR and were significantly increased in the S334ter-4 model.


Subject(s)
Dependovirus , Glycoproteins/therapeutic use , Neuroprotective Agents/therapeutic use , Retinitis Pigmentosa/drug therapy , Animals , Disease Models, Animal , Electroretinography , Glycoproteins/administration & dosage , Neuroprotective Agents/administration & dosage , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Transgenic , Retinitis Pigmentosa/pathology
4.
J Lipid Res ; 57(5): 818-31, 2016 05.
Article in English | MEDLINE | ID: mdl-26947037

ABSTRACT

Retinal degeneration (RD) affects millions of people and is a major cause of ocular impairment and blindness. With a wide range of mutations and conditions leading to degeneration, targeting downstream processes is necessary for developing effective treatments. Ceramide and sphingosine-1-phosphate, a pair of bioactive sphingolipids, are involved in apoptosis and its prevention, respectively. Apoptotic cell death is a potential driver of RD, and in order to understand the mechanism of degeneration and potential treatments, we studied rhodopsin mutant RD model, P23H-1 rats. Investigating this genetic model of human RD allows us to investigate the association of sphingolipid metabolites with the degeneration of the retina in P23H-1 rats and the effects of a specific modulator of sphingolipid metabolism, FTY720. We found that P23H-1 rat retinas had altered sphingolipid profiles that, when treated with FTY720, were rebalanced closer to normal levels. FTY720-treated rats also showed protection from RD compared with their vehicle-treated littermates. Based on these data, we conclude that sphingolipid dysregulation plays a secondary role in retinal cell death, which may be common to many forms of RDs, and that the U.S. Food and Drug Administration-approved drug FTY720 or related compounds that modulate sphingolipid metabolism could potentially delay the cell death.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Retinal Dystrophies/metabolism , Sphingolipids/metabolism , Animals , Biosynthetic Pathways , Disease Progression , Drug Evaluation, Preclinical , Fingolimod Hydrochloride/therapeutic use , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Rats, Sprague-Dawley , Retinal Dystrophies/drug therapy , Sphingomyelin Phosphodiesterase/metabolism
5.
Adv Exp Med Biol ; 854: 185-91, 2016.
Article in English | MEDLINE | ID: mdl-26427410

ABSTRACT

RHO (Rod opsin) encodes a G-protein coupled receptor that is expressed exclusively by rod photoreceptors of the retina and forms the essential photopigment, rhodopsin, when coupled with 11-cis-retinal. Many rod opsin disease -mutations cause rod opsin protein misfolding and trigger endoplasmic reticulum (ER) stress, leading to activation of the Unfolded Protein Response (UPR) signal transduction network. Chop is a transcriptional activator that is induced by ER stress and promotes cell death in response to chronic ER stress. Here, we examined the role of Chop in transgenic mice expressing human P23H rhodopsin (hP23H Rho Tg) that undergo retinal degeneration. With the exception of one time point, we found no significant induction of Chop in these animals and no significant change in retinal degeneration by histology and electrophysiology when hP23H Rho Tg animals were bred into a Chop (-/-) background. Our results indicate that Chop does not play a significant causal role during retinal degeneration in these animals. We suggest that other modules of the ER stress-induced UPR signaling network may be involved photoreceptor disease induced by P23H rhodopsin.


Subject(s)
Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/genetics , Transcription Factor CHOP/genetics , Animals , Cell Survival/genetics , Electroretinography , Gene Expression , Humans , Mice, Knockout , Mice, Transgenic , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/metabolism , Transcription Factor CHOP/deficiency , Transgenes/genetics
6.
Adv Exp Med Biol ; 854: 487-93, 2016.
Article in English | MEDLINE | ID: mdl-26427450

ABSTRACT

MERTK-associated retinal degenerations are thought to have defects in phagocytosis of shed outer segment membranes by the retinal pigment epithelium (RPE), as do the rodent models of these diseases. We have subretinally injected an RPE-specific AAV2 vector, AAV2-VMD2-hMERTK, to determine whether this would provide long-term photoreceptor rescue in the RCS rat, which it did for up to 6.5 months, the longest time point examined. Moreover, we found phagosomes in the RPE in the rescued regions of RCS retinas soon after the onset of light. The same vector also had a major protective effect in Mertk-null mice, with a concomitant increase in ERG response amplitudes in the vector-injected eyes. These findings suggest that planned clinical trials with this vector will have a favorable outcome.


Subject(s)
Genetic Therapy/methods , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Animals , Bestrophins , Chloride Channels/genetics , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Eye Proteins/genetics , Genetic Vectors/genetics , Humans , Mice, Knockout , Phagocytosis/genetics , Phagocytosis/physiology , Phagosomes/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , Rats, Mutant Strains , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/metabolism , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiopathology , Treatment Outcome , c-Mer Tyrosine Kinase
7.
PLoS Genet ; 11(12): e1005723, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26656104

ABSTRACT

Inherited photoreceptor degenerations (IPDs) are the most genetically heterogeneous of Mendelian diseases. Many IPDs exhibit substantial phenotypic variability, but the basis is usually unknown. Mutations in MERTK cause recessive IPD phenotypes associated with the RP38 locus. We have identified a murine genetic modifier of Mertk-associated photoreceptor degeneration, the C57BL/6 (B6) allele of which acts as a suppressor. Photoreceptors degenerate rapidly in Mertk-deficient animals homozygous for the 129P2/Ola (129) modifier allele, whereas animals heterozygous for B6 and 129 modifier alleles exhibit an unusual intermixing of degenerating and preserved retinal regions, with females more severely affected than males. Mertk-deficient mice homozygous for the B6 modifier allele display degeneration only in the far periphery, even at 8 months of age, and have improved retinal function compared to animals homozygous for the 129 allele. We genetically mapped the modifier to an approximately 2-megabase critical interval that includes Tyro3, a paralog of Mertk. Tyro3 expression in the outer retina varies with modifier genotype in a manner characteristic of a cis-acting expression quantitative trait locus (eQTL), with the B6 allele conferring an approximately three-fold higher expression level. Loss of Tyro3 function accelerates the pace of photoreceptor degeneration in Mertk knockout mice, and TYRO3 protein is more abundant in the retinal pigment epithelium (RPE) adjacent to preserved central retinal regions of Mertk knockout mice homozygous for the B6 modifier allele. Endogenous human TYRO3 protein co-localizes with nascent photoreceptor outer segment (POS) phagosomes in a primary RPE cell culture assay, and expression of murine Tyro3 in cultured cells stimulates phagocytic ingestion of POS. Our findings demonstrate that Tyro3 gene dosage modulates Mertk-associated retinal degeneration, provide strong evidence for a direct role for TYRO3 in RPE phagocytosis, and suggest that an eQTL can modify a recessive IPD.


Subject(s)
Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Retinal Degeneration/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Phagocytosis , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Retina/metabolism , Retina/pathology , Retinal Degeneration/pathology , Retinal Pigment Epithelium/pathology , c-Mer Tyrosine Kinase
8.
Invest Ophthalmol Vis Sci ; 56(11): 6961-70, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26513501

ABSTRACT

PURPOSE: Endoplasmic reticulum (ER) stress activates inositol requiring enzyme 1 (IRE1), a key regulator of the unfolded protein response. The ER stress activated indicator (ERAI) transgenic mouse expresses a yellow fluorescent GFP variant (Venus) when IRE1 is activated by ER stress. We tested whether ERAI mice would allow for real-time longitudinal studies of ER stress in living mouse eyes. METHODS: We chemically and genetically induced ER stress, and qualitatively and quantitatively studied the Venus signal by fluorescence ophthalmoscopy. We determined retinal cell types that contribute to the signal by immunohistology, and we performed molecular and biochemical assays using whole retinal lysates to assess activity of the IRE1 pathway. RESULTS: We found qualitative increase in vivo in fluorescence signal at sites of intravitreal tunicamycin injection in ERAI eyes, and quantitative increase in ERAI mice mated to RhoP23H mice expressing ER stress-inducing misfolded rhodopsin protein. As expected, we found that increased Venus signal arose primarily from photoreceptors in RhoP23H/+;ERAI mice. We found increased Xbp1S and XBP1s transcriptional target mRNA levels in RhoP23H/+;ERAI retinas compared to Rho+/+;ERAI retinas, and that Venus signal increased in ERAI retinas as a function of age. CONCLUSIONS: Fluorescence ophthalmoscopy of ERAI mice enables in vivo visualization of retinas undergoing ER stress. ER stress activated indicator mice enable identification of individual retinal cells undergoing ER stress by immunohistochemistry. ER stress activated indicator mice show higher Venus signal at older ages, likely arising from amplification of basal retinal ER stress levels by GFP's inherent stability.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Retina/diagnostic imaging , Animals , Endoplasmic Reticulum/diagnostic imaging , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/drug effects , Longitudinal Studies , Membrane Proteins/analysis , Membrane Proteins/physiology , Mice , Mice, Transgenic , Ophthalmoscopy , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/physiology , Retina/chemistry , Retina/drug effects , Retina/pathology , Retina/physiology , Signal Transduction/physiology , Tomography, Optical Coherence , Tunicamycin/pharmacology , Ultrasonography
9.
Invest Ophthalmol Vis Sci ; 56(11): 6362-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26436889

ABSTRACT

PURPOSE: To preserve photoreceptor cell structure and function in a rodent model of retinitis pigmentosa with P23H rhodopsin by selective inhibition of the mutant rhodopsin allele using a second generation antisense oligonucleotide (ASO). METHODS: Wild-type mice and rats were treated with ASO by intravitreal (IVT) injection and rhodopsin mRNA and protein expression were measured. Transgenic rats expressing the murine P23H rhodopsin gene (P23H transgenic rat Line 1) were administered either a mouse-specific P23H ASO or a control ASO. The contralateral eye was injected with PBS and used as a comparator control. Electroretinography (ERG) measurements and analyses of the retinal outer nuclear layer were conducted and correlated with rhodopsin mRNA levels. RESULTS: Rhodopsin mRNA and protein expression was reduced after a single ASO injection in wild-type mice with a rhodopsin-specific ASO. Transgenic rat eyes that express a murine P23H rhodopsin gene injected with a murine P23H ASO had a 181 ± 39% better maximum amplitude response (scotopic a-wave) as compared with contralateral PBS-injected eyes; the response in control ASO eyes was not significantly different from comparator contralateral eyes. Morphometric analysis of the outer nuclear layer showed a significantly thicker nuclear layer in eyes injected with murine P23H ASO (18%) versus contralateral PBS-injected eyes. CONCLUSIONS: Allele-specific ASO-mediated knockdown of mutant P23H rhodopsin expression slowed the rate of photoreceptor degeneration and preserved the function of photoreceptor cells in eyes of the P23H rhodopsin transgenic rat. Our data indicate that ASO treatment is a potentially effective therapy for the treatment of retinitis pigmentosa.


Subject(s)
Gene Expression Regulation , Macular Degeneration/prevention & control , Oligonucleotides, Antisense/genetics , RNA, Messenger/genetics , Rhodopsin/genetics , Alleles , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Macular Degeneration/genetics , Macular Degeneration/metabolism , Male , Mice , Rats , Rats, Transgenic , Real-Time Polymerase Chain Reaction , Rhodopsin/biosynthesis
10.
Mol Neurobiol ; 52(1): 679-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25270370

ABSTRACT

Rhodopsin is a G protein-coupled receptor essential for vision and rod photoreceptor viability. Disease-associated rhodopsin mutations, such as P23H rhodopsin, cause rhodopsin protein misfolding and trigger endoplasmic reticulum (ER) stress, activating the unfolded protein response (UPR). The pathophysiologic effects of ER stress and UPR activation on photoreceptors are unclear. Here, by examining P23H rhodopsin knock-in mice, we found that the UPR inositol-requiring enzyme 1 (IRE1) signaling pathway is strongly activated in misfolded rhodopsin-expressing photoreceptors. IRE1 significantly upregulated ER-associated protein degradation (ERAD), triggering pronounced P23H rhodopsin degradation. Rhodopsin protein loss occurred as soon as photoreceptors developed, preceding photoreceptor cell death. By contrast, IRE1 activation did not affect JNK signaling or rhodopsin mRNA levels. Interestingly, pro-apoptotic signaling from the PERK UPR pathway was also not induced. Our findings reveal that an early and significant pathophysiologic effect of ER stress in photoreceptors is the highly efficient elimination of misfolded rhodopsin protein. We propose that early disruption of rhodopsin protein homeostasis in photoreceptors could contribute to retinal degeneration.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/metabolism , Animals , Animals, Newborn , Apoptosis , Endoplasmic Reticulum Stress , Gene Knock-In Techniques , Immunoprecipitation , Membrane Proteins/metabolism , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Retina/ultrastructure , Retinal Photoreceptor Cell Inner Segment/metabolism , Retinal Photoreceptor Cell Inner Segment/pathology , Retinal Photoreceptor Cell Inner Segment/ultrastructure , Rhodopsin/genetics , Signal Transduction , Transcription Factor CHOP/metabolism , Ubiquitination
11.
PLoS One ; 8(2): e56026, 2013.
Article in English | MEDLINE | ID: mdl-23409115

ABSTRACT

Huntington disease (HD) is an inherited neurodegenerative disease that affects multiple brain regions. It is caused by an expanded polyglutamine tract in huntingtin (Htt). The development of therapies for HD and other neurodegenerative diseases has been hampered by multiple factors, including the lack of clear therapeutic targets, and the cost and complexity of testing lead compounds in vivo. The R6/2 HD mouse model is widely used for pre-clinical trials because of its progressive and robust neural dysfunction, which includes retinal degeneration. Profilin-1 is a Htt binding protein that inhibits Htt aggregation. Its binding to Htt is regulated by the rho-associated kinase (ROCK), which phosphorylates profilin at Ser-137. ROCK is thus a therapeutic target in HD. The ROCK inhibitor Y-27632 reduces Htt toxicity in fly and mouse models. Here we characterized the progressive retinopathy of R6/2 mice between 6-19 weeks of age to determine an optimal treatment window. We then tested a clinically approved ROCK inhibitor, HA-1077, administered intravitreally via liposome-mediated drug delivery. HA-1077 increased photopic and flicker ERG response amplitudes in R6/2 mice, but not in wild-type littermate controls. By targeting ROCK with a new inhibitor, and testing its effects in a novel in vivo model, these results validate the in vivo efficacy of a therapeutic candidate, and establish the feasibility of using the retina as a readout for CNS function in models of neurodegenerative disease.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Huntington Disease/metabolism , Protein Kinase Inhibitors/pharmacology , Retina/drug effects , Retina/metabolism , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Disease Models, Animal , Electroretinography , Female , Gene Expression , Huntingtin Protein , Huntington Disease/genetics , Liposomes , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation/drug effects , Profilins/metabolism , Protein Kinase Inhibitors/administration & dosage , Retina/pathology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/metabolism
12.
Invest Ophthalmol Vis Sci ; 53(12): 7590-9, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23074209

ABSTRACT

PURPOSE: Endoplasmic reticulum (ER) stress has been observed in animal models of retinitis pigmentosa expressing P23H rhodopsin. We compared levels of tightly induced ER stress genes, Binding of immunoglobulin protein (BiP) and CCAAT/enhancer-binding protein homologous protein (Chop), in seven additional models of retinal degeneration arising from genetic or environmental causes. METHODS: Retinas from transgenic S334ter rhodopsin (lines 3, 4, and 5) and Royal College of Surgeons (RCS and RCS-p+) rats from postnatal (P) days 10 to 120 were analyzed. In a constant light (CL) model of retinal degeneration, BALB/c mice were exposed to 15,000 lux of CL for 0 to 8 hours. Retinal tissues from three to eight animals per experimental condition were collected for histologic and molecular analyses. RESULTS: S334ter animals revealed significant increases in BiP, S334ter-3 (3.3× at P15), S334ter-4 (4× at P60), and S334ter-5 (2.2× at P90), and Chop, S334ter-3 (1.3× at P15), S334ter-4 (1.5× at P30), and S334ter-5 (no change), compared with controls. P23H-3 rats showed significant increase of BiP at P60 (2.3×) and Chop (1.6×). RCS and RCS-p+ rats showed significant increases in BiP at P60 (2.4×) and P20 (1.8×), respectively, but no statistically significant changes in Chop. BALB/c mice showed increases in BiP (1.5×) and Chop (1.3×) after 4 hours of CL. Increased levels of these ER stress markers correlated with photoreceptor cell loss. CONCLUSIONS: Our study reveals surprising increases in BiP and to a lesser degree Chop in retinal degenerations arising from diverse causes. We propose that manipulation of ER stress responses may be helpful in treating many environmental and heritable forms of retinal degeneration.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Oligopeptides/genetics , RNA/genetics , Retinal Degeneration/genetics , Transcription Factor CHOP/genetics , Animals , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Environmental Exposure/adverse effects , Mice , Mice, Inbred BALB C , Oligopeptides/biosynthesis , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/genetics , Rhodopsin/metabolism , Transcription Factor CHOP/biosynthesis , bcl-2-Associated X Protein
13.
Invest Ophthalmol Vis Sci ; 53(10): 6232-44, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22899760

ABSTRACT

PURPOSE: To assess structural, functional, and visual behavioral relationships in mutant rhodopsin transgenic (Tg) rats and to determine whether early optokinetic tracking (OKT) visual experience, known to permanently elevate visual thresholds in normal rats, can enhance vision in rats with photoreceptor degeneration. METHODS: Eight lines of pigmented Tg rats and RCS rats were used in this study. OKT thresholds were tested at single ages (1, 2, 3, 4, and 6 months) in naïve groups of rats, or daily in groups that began at eye-opening (P15) or 10 days later (P25). Electroretinogram (ERG) response amplitudes were recorded after OKT testing, and outer nuclear layer (ONL) thickness measurements were then obtained. RESULTS: OKT thresholds, when measured at a single time point in naïve Tg lines beginning at P30, did not decline until months after significant photoreceptor loss. Daily testing of Tg lines resulted mostly with OKT thresholds inversely related to photoreceptor degeneration, with rapid degenerations resulting in sustained OKT thresholds for long periods despite the rapid photoreceptor loss. Slower degenerations resulted in rapid decline of thresholds, long before the loss of most photoreceptors, which was even more pronounced when daily testing began at eye opening. This amplified loss of function was not a result of testing-induced damage to the rod or cone photoreceptors, as ERG amplitudes and ONL thicknesses were the same as untested controls. CONCLUSIONS: The unexpected lack of correlation of OKT testing with photoreceptor degeneration in the Tg rats emphasizes the need in behavioral therapeutic studies for careful analysis of visual thresholds of experimental animals prior to therapeutic intervention.


Subject(s)
Retinal Degeneration/physiopathology , Visual Perception/physiology , Animals , Disease Models, Animal , Electroretinography , Mutation , Rats , Rats, Transgenic , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/genetics , Sensory Thresholds/physiology
14.
Invest Ophthalmol Vis Sci ; 53(4): 1895-904, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22408006

ABSTRACT

PURPOSE: The absence of Mertk in RCS rats results in defective RPE phagocytosis, accumulation of outer segment (OS) debris in the subretinal space, and subsequent death of photoreceptors. Previous research utilizing Mertk gene replacement therapy in RCS rats provided proof of concept for treatment of this form of recessive retinitis pigmentosa (RP); however, the beneficial effects on retinal function were transient. In the present study, we evaluated whether delivery of a MERTK transgene using a tyrosine-mutant AAV8 capsid could lead to more robust and longer-term therapeutic outcomes than previously reported. METHODS: An AAV8 Y733F vector expressing a human MERTK cDNA driven by a RPE-selective promoter was administrated subretinally at postnatal day 2. Functional and morphological analyses were performed at 4 months and 8 months post-treatment. Retinal vasculature and Müller cell activation were analyzed by quantifying acellular capillaries and glial fibrillary acidic protein immunostaining, respectively. RESULTS: Electroretinographic responses from treated eyes were more than one-third of wild-type levels and OS were well preserved in the injection area even at 8 months. Rescue of RPE phagocytosis, prevention of retinal vasculature degeneration, and inhibition of Müller cell activation were demonstrated in the treated eyes for at least 8 months. CONCLUSIONS: This research describes a longer and much more robust functional and morphological rescue than previous studies. We also demonstrate for the first time that an AAV8 mutant capsid serotype vector has a substantial therapeutic potential for RPE-specific gene delivery. These results suggest that tyrosine-mutant AAV8 vectors hold promise for the treatment of individuals with MERTK-associated RP.


Subject(s)
Genetic Therapy/methods , Proto-Oncogene Proteins/administration & dosage , Receptor Protein-Tyrosine Kinases/administration & dosage , Retinitis Pigmentosa/therapy , Animals , Animals, Newborn , Blotting, Western , Disease Models, Animal , Electroretinography , Follow-Up Studies , Genetic Vectors , Humans , Immunohistochemistry , Injections , Microscopy, Electron, Transmission , Mutation , Plasmids , Proto-Oncogene Proteins/therapeutic use , RNA/genetics , Rats , Rats, Mutant Strains , Receptor Protein-Tyrosine Kinases/therapeutic use , Retina , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tomography, Optical Coherence , Transgenes , Tyrosine/genetics , c-Mer Tyrosine Kinase
15.
Mol Ther ; 20(4): 788-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22294148

ABSTRACT

Oxidative stress and photoreceptor apoptosis are prominent features of many forms of retinal degeneration (RD) for which there are currently no effective therapies. We previously observed that mesenchymal stem/stromal cells reduce apoptosis by being activated to secrete stanniocalcin-1 (STC-1), a multifunctional protein that reduces oxidative stress by upregulating mitochondrial uncoupling protein-2 (UCP-2). Therefore, we tested the hypothesis that intravitreal injection of STC-1 can rescue photoreceptors. We first tested STC-1 in the rhodopsin transgenic rat characterized by rapid photoreceptor loss. Intravitreal STC-1 decreased the loss of photoreceptor nuclei and transcripts and resulted in measurable retinal function when none is otherwise present in this rapid degeneration. We then tested STC-1 in the Royal College of Surgeons (RCS) rat characterized by a slower photoreceptor degeneration. Intravitreal STC-1 reduced the number of pyknotic nuclei in photoreceptors, delayed the loss of photoreceptor transcripts, and improved function of rod photoreceptors. Additionally, STC-1 upregulated UCP-2 and decreased levels of two protein adducts generated by reactive oxygen species (ROS). Microarrays from the two models demonstrated that STC-1 upregulated expression of a similar profile of genes for retinal development and function. The results suggested that intravitreal STC-1 is a promising therapy for various forms of RD including retinitis pigmentosa and atrophic age-related macular degeneration (AMD).


Subject(s)
Glycoproteins/pharmacology , Retinal Degeneration/drug therapy , Animals , Electroretinography , Enzyme-Linked Immunosorbent Assay , Humans , Ion Channels/genetics , Ion Channels/metabolism , Macular Degeneration/drug therapy , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Rats , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/drug therapy , Uncoupling Protein 2
18.
Exp Eye Res ; 87(6): 561-70, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18848932

ABSTRACT

The early loss of photoreceptors in some retinal degenerations in mice has been shown to have a profound effect on vascular development of the retina. To better characterize this relationship, we have examined the formation of retinal blood vessels during the first month of life in 8 lines of transgenic rats with different ages of onset and rates of photoreceptor cell loss mediated by the expression of mutant rhodopsin (P23H and S334ter). The number of capillary profiles in the superficial plexus (SP) and deep capillary plexus (DCP) of the retina were quantified in retinal sections taken at postnatal day (P) 8, 10, 12, 15 and 30. In normal wild-type rats, the SP and DCP had mostly established mature, adult patterns by P15, as previously shown. In the transgenic rats, the loss of photoreceptors had relatively little effect on the SP. By contrast, the loss of photoreceptors during vascular development had a major impact on the DCP. In the two lines with early and most rapid photoreceptor loss, S334ter-7 and S334ter-3, where about 90% and 65%, respectively, of the photoreceptors were already lost by P15, the DCP either failed to form (S334ter-7) or the number of capillary profiles was less than 7% of controls (S334ter-3). In lines where almost all photoreceptors were still present at P15 (S334ter-4, S334ter-9, P23H-2 and P23H-3), the number of profiles in the DCP were the same as in wild-type controls at P30. In two lines with an intermediate rate of degeneration (S334ter-5 and P23H-1), where only about 25% of the photoreceptors were lost by P15, there was an intermediate number of vascular profiles in the DCP at P30. Thus, a very close relationship between the number of photoreceptors and vessel profiles in the DCP during its development exists in the transgenic rats, and the loss of photoreceptors results in the failure or inhibition of the DCP to develop. Several mechanisms may explain this relationship including changes in the level of physiological oxygen tension or alteration in the release of angiogenic factors that normally drive vessel development. Analysis of older transgenic retinas up to 1 year of age revealed that (1) vascular profiles are lost from the DCP in essentially all lines once fewer than about 30-33% of photoreceptors remain; (2) in those lines where the DCP essentially did not develop (S334ter-7 and S334ter-3), the effect of photoreceptor absence was permanent, and there was no late vascularization of the DCP; (3) the number of capillary profiles in the SP remained no different from controls in any of the lines, despite long-standing loss of photoreceptors; and (4) neovascularization of the RPE by retinal capillaries occurred with a latency of 60-180 days after the loss of photoreceptors, except in S334ter-7 rats, where neovascularization essentially did not occur. Analysis of RCS rats was carried out for comparison.


Subject(s)
Mutation , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/pathology , Retinal Vessels/growth & development , Rhodopsin/genetics , Aging/pathology , Animals , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/blood supply , Retinal Pigment Epithelium/pathology , Rod Cell Outer Segment/pathology
19.
J Comp Neurol ; 511(6): 724-35, 2008 Dec 20.
Article in English | MEDLINE | ID: mdl-18925574

ABSTRACT

Several neurotrophic factors (NTFs) are effective in protecting retinal photoreceptor cells from the damaging effects of constant light and slowing the rate of inherited photoreceptor degenerations. It is currently unclear whether, if continuously available, all NTFs can be protective for many or most retinal degenerations (RDs). We used transgenic mice that continuously overexpress the neurotrophin NT-3 from lens fibers under the control of the alphaA-crystallin promoter to test for neuroprotection in light-damage experiments and in four naturally occurring or transgenically induced RDs in mice. Lens-specific expression of NT-3 mRNA was demonstrated both by in situ hybridization in embryos and by reverse-transcriptase polymerase chain reaction (RT-PCR) in adult mice. Furthermore, NT-3 protein was found in abundance in the lens, ocular fluids, and retina by enzyme-linked immunosorbent assay (ELISA) and immunocytochemistry. Overexpression of NT-3 had no adverse effects on the structure or function of the retina for up to at least 14 months of age. Mice expressing the NT-3 transgene were protected from the damaging effects of constant light to a much greater degree than those receiving bolus injections of NT-3. When the NT-3 transgene was transferred into rd/rd, Rds/+, Q344ter mutant rhodopsin or Mertk knockout mice, overexpression of NT-3 had no protective effect on the RDs in these mice. Thus, specificity of the neuroprotective effect of NT-3 is clearly demonstrated, and different molecular mechanisms are inferred to mediate the protective effect in light-induced and inherited RDs.


Subject(s)
Cytoprotection/genetics , Epithelial Cells/metabolism , Lens, Crystalline/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Retinal Degeneration/therapy , Animals , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lens, Crystalline/physiopathology , Lens, Crystalline/radiation effects , Light/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/radiation effects , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Retinal Degeneration/etiology , Retinal Degeneration/physiopathology , Time Factors , Transgenes/genetics , c-Mer Tyrosine Kinase
20.
Invest Ophthalmol Vis Sci ; 49(7): 2863-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18344445

ABSTRACT

PURPOSE: In previous studies of light-induced (LRD) and age-related (ageRD) retinal degeneration (RD) between the BALB/cByJ (BALB) and B6(Cg)-Tyr(c-2J)/J (B6a) albino mouse strains, RD-modifying quantitative trait loci (QTLs) were identified. After breeding BALB- and B6a-rd3/rd3 congenic strains and finding significant differences in RD, an F1 intercross to determine rd3 QTLs that influence this inherited RD was performed. METHODS: N10, F2 BALB- and B6a-rd3/rd3 strains were measured for retinal outer nuclear layer (ONL) thickness from 5 to 12 weeks of age. Since 10 weeks showed significant differences in the ONL, F2 progeny from an F1 intercross were measured for ONL thickness. F2 DNAs were genotyped for SNPs by the Center for Inherited Disease Research. Correlation of genotype with phenotype was made with Map Manager QTX. RESULTS: One hundred forty-eight SNPs approximately 10 cM apart were typed in the F2 progeny and analyzed. Significant QTLs were identified on chromosomes (Chrs) 17, 8, 14, and 6 (B6a alleles protective) and two on Chr 5 (BALB alleles protective). Suggestive QTLs were found as well. For the strongest QTLs, follow-up SNPs were analyzed to narrow the critical intervals. Additional studies demonstrated that rd3 disease is exacerbated by light but not protected by the absence of rhodopsin regeneration. CONCLUSIONS: QTLs were identified that modulate rd3-RD. These overlapped some QTLs from previous ageRD and LRD studies. The presence of some of the same QTLs in several studies suggests partial commonality in RD pathways. Identifying natural gene/alleles that modify RDs opens avenues of study that may lead to therapies for RD diseases.


Subject(s)
Mice, Mutant Strains/genetics , Mice, Mutant Strains/metabolism , Quantitative Trait Loci , Retinal Degeneration/genetics , Animals , Chromosomes, Mammalian , Eye/metabolism , Genotype , Light , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymorphism, Single Nucleotide , Retina/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Pigments/metabolism , Rhodopsin/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL