Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Elife ; 122024 May 10.
Article in English | MEDLINE | ID: mdl-38727583

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinal disease in which there is a loss of cone-mediated daylight vision. As there are >100 disease genes, our goal is to preserve cone vision in a disease gene-agnostic manner. Previously we showed that overexpressing TXNIP, an α-arrestin protein, prolonged cone vision in RP mouse models, using an AAV to express it only in cones. Here, we expressed different alleles of Txnip in the retinal pigmented epithelium (RPE), a support layer for cones. Our goal was to learn more of TXNIP's structure-function relationships for cone survival, as well as determine the optimal cell type expression pattern for cone survival. The C-terminal half of TXNIP was found to be sufficient to remove GLUT1 from the cell surface, and improved RP cone survival, when expressed in the RPE, but not in cones. Knock-down of HSP90AB1, a TXNIP-interactor which regulates metabolism, improved the survival of cones alone and was additive for cone survival when combined with TXNIP. From these and other results, it is likely that TXNIP interacts with several proteins in the RPE to indirectly support cone survival, with some of these interactions different from those that lead to cone survival when expressed only in cones.


Subject(s)
Carrier Proteins , Disease Models, Animal , Retinal Cone Photoreceptor Cells , Retinitis Pigmentosa , Animals , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Mice , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation, Missense , Cell Survival , Alleles , Gene Deletion , Thioredoxins/genetics , Thioredoxins/metabolism , Retinal Pigment Epithelium/metabolism
2.
bioRxiv ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38370727

ABSTRACT

Retinitis pigmentosa (RP) is a prevalent inherited retinal degenerative disease worldwide, affecting 1 in 4,000 people. The disease is characterized by an initial loss of night vision followed by a loss of daylight and color vision. Many of the RP disease genes are expressed in the rod photoreceptors, the cell type that initiates dim light vision. Following loss of rods, the cone photoreceptors, which initiate daylight vision, also are affected and can die leading to total loss of vision. The reasons for loss of cone vision are not entirely clear, but appear to be due to loss of the rods. Previously we showed that overexpressing Txnip, an α-arrestin protein, in mouse models of RP using AAV gene therapy prolonged the survival of RP cones (Xue et al., 2021). At least part of the mechanism for cone survival was a switch in the fuel source, from glucose to lactate. In addition, the mitochondria of cones were both morphologically and functionally improved by delivery of Txnip. We have gone on to test several alleles of Txnip for the ability to prolong cone survival in rd1, a mouse model of RP. In addition, proteins that bind to Txnip and/or have homology to Txnip were tested. Five different deletion alleles of Txnip were expressed in cones or the retinal pigmented epithelium (RPE). Here we show that the C-terminal half of Txnip (149-397aa) is sufficient to remove GLUT1 from the RPE cell surface, and improved rd1 cone survival when expressed specifically in the RPE. Overexpressing Arrdc4, an α-arrestin that shares 60% similar protein sequence to Txnip, reduced rd1 cone survival. Reduction of the expression of HSP90AB1, a protein that interacts with Txnip and regulates metabolism, improved the survival of rd1 cones alone and was additive for cone survival when combined with Txnip. However, full length Txnip with a single amino acid change, C247S, as we tested in our original study, remains the most highly efficacious form of the gene for cone rescue. The above observations suggest that only a subset of the hypothesized and known activities of Txnip play a role in promoting RP cone survival, and that the activities of Txnip in the RPE differ from those in cone photoreceptors.

3.
bioRxiv ; 2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37961274

ABSTRACT

Vision is initiated by the reception of light by photoreceptors and subsequent processing via parallel retinal circuits. Proper circuit organization depends on the multi-functional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here we investigated the retinal function of Fat3 mutant mice and found decreases in physiological and perceptual responses to high frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. Indeed, similar deficits were observed in mice lacking the bipolar cell glutamate receptors GRIK1 (OFF-bipolar cells) and GRM6 (ON-bipolar cells). Mechanistically, FAT3 binds to the synaptic protein PTPσ and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. How FAT3 impacts ON-cone bipolar cell function at high temporal frequency remains to be uncovered. These findings expand the repertoire of FAT3's functions and reveal the importance of both ON- and OFF-bipolar cells for high frequency light response.

4.
Adv Exp Med Biol ; 1415: 143-146, 2023.
Article in English | MEDLINE | ID: mdl-37440027

ABSTRACT

Retinitis pigmentosa (RP) is a hereditary retinal degenerative disease that can lead to blindness. In RP, rod photoreceptors die first, followed by cone photoreceptors death due to unknown mechanisms. However, one clue for cone death concerns their metabolism. Early changes suggest that they do not have enough glucose, which normally fuels their metabolism. We sought to design adeno-associated virus (AAV)-based gene therapy to address their metabolic challenges and found that overexpressing Txnip is an effective gene therapy that extends cone survival and vision in three strains of RP mice. The Txnip-mediated rescue was found to be dependent upon lactate dehydrogenase b (Ldhb), which is required for lactate catabolism. Txnip also was found to improve mitochondrial health. Herein, we propose a model in which Txnip shifts cones from their normal reliance on glucose to enhanced utilization of lactate to benefit cones in a condition where the glucose supply is limiting.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retinal Degeneration/therapy , Genetic Therapy , Glucose/metabolism , Disease Models, Animal , Carrier Proteins/genetics , Carrier Proteins/metabolism , Thioredoxins/genetics
5.
Article in English | MEDLINE | ID: mdl-37460158

ABSTRACT

Retinitis pigmentosa is a blinding disease wherein rod photoreceptors are affected first, due to the expression of a disease gene, leading to the loss of dim light vision. In many cases, cones do not express the disease gene, yet they are also affected and eventually die, typically after most of the rods in their neighborhood have died. The cause of secondary cone death is unclear. Photoreceptors are one of the most energy-demanding cell types in the body and consume a high amount of glucose. At an early stage of degeneration, the cones appear to have a shortage of glucose to fuel their metabolism. This review focuses on gene therapy approaches that address this potential metabolic shortcoming.

6.
Proc Natl Acad Sci U S A ; 120(23): e2217885120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252956

ABSTRACT

Retinitis pigmentosa (RP) is an ocular disease characterized by the loss of night vision, followed by the loss of daylight vision. Daylight vision is initiated in the retina by cone photoreceptors, which are gradually lost in RP, often as bystanders in a disease process that initiates in their neighboring rod photoreceptors. Using physiological assays, we investigated the timing of cone electroretinogram (ERG) decline in RP mouse models. A correlation between the time of loss of the cone ERG and the loss of rods was found. To investigate a potential role of the visual chromophore supply in this loss, mouse mutants with alterations in the regeneration of the retinal chromophore, 11-cis retinal, were examined. Reducing chromophore supply via mutations in Rlbp1 or Rpe65 resulted in greater cone function and survival in a RP mouse model. Conversely, overexpression of Rpe65 and Lrat, genes that can drive the regeneration of the chromophore, led to greater cone degeneration. These data suggest that abnormally high chromophore supply to cones upon the loss of rods is toxic to cones, and that a potential therapy in at least some forms of RP is to slow the turnover and/or reduce the level of visual chromophore in the retina.


Subject(s)
Color Vision , Retinitis Pigmentosa , Mice , Animals , Retina , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/genetics , Disease Models, Animal
7.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34197341

ABSTRACT

Inherited retinal diseases, such as retinitis pigmentosa (RP), can be caused by thousands of different mutations, a small number of which have been successfully treated with gene replacement. However, this approach has yet to scale and may not be feasible in many cases, highlighting the need for interventions that could benefit more patients. Here, we found that microglial phagocytosis is upregulated during cone degeneration in RP, suggesting that expression of "don't-eat-me" signals such as CD47 might confer protection to cones. To test this, we delivered an adeno-associated viral (AAV) vector expressing CD47 on cones, which promoted cone survival in 3 mouse models of RP and preserved visual function. Cone rescue with CD47 required a known interacting protein, signal regulatory protein α (SIRPα), but not an alternative interacting protein, thrombospondin-1 (TSP1). Despite the correlation between increased microglial phagocytosis and cone death, microglia were dispensable for the prosurvival activity of CD47, suggesting that CD47 interacts with SIRPα on nonmicroglial cells to alleviate degeneration. These findings establish augmentation of CD47/SIRPα signaling as a potential treatment strategy for RP and possibly other forms of neurodegeneration.


Subject(s)
CD47 Antigen/genetics , Receptors, Immunologic/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Animals , CD47 Antigen/metabolism , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Knockout , Microglia/pathology , Phagocytosis , Receptors, Immunologic/metabolism , Retinitis Pigmentosa/pathology , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
8.
Elife ; 102021 04 13.
Article in English | MEDLINE | ID: mdl-33847261

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinal disease affecting >20 million people worldwide. Loss of daylight vision typically occurs due to the dysfunction/loss of cone photoreceptors, the cell type that initiates our color and high-acuity vision. Currently, there is no effective treatment for RP, other than gene therapy for a limited number of specific disease genes. To develop a disease gene-agnostic therapy, we screened 20 genes for their ability to prolong cone photoreceptor survival in vivo. Here, we report an adeno-associated virus vector expressing Txnip, which prolongs the survival of cone photoreceptors and improves visual acuity in RP mouse models. A Txnip allele, C247S, which blocks the association of Txnip with thioredoxin, provides an even greater benefit. Additionally, the rescue effect of Txnip depends on lactate dehydrogenase b (Ldhb) and correlates with the presence of healthier mitochondria, suggesting that Txnip saves RP cones by enhancing their lactate catabolism.


Retinitis pigmentosa is an inherited eye disease affecting around one in every 4,000 people. It results from genetic defects in light sensitive cells of the retina, called photoreceptor cells, which line the back of the eye. Though vision loss can occur from birth, retinitis pigmentosa usually involves a gradual loss of vision, sometimes leading to blindness. Rod photoreceptors, which are responsible for vision in low light, are impacted first. The disease then affects cone photoreceptors, the cells that detect light during the day, providing both color and sharp vision. Around 100 mutated genes associated with retinitis pigmentosa have been identified, but only a handful of families with one of these mutant genes have been treated with a gene therapy specific for their mutated gene. There are currently no therapies available to treat the vast number of people with this disease. The mutations that cause retinitis pigmentosa directly affect the rod cells that detect dim light, leading to loss of night vision. There is also an indirect effect that causes cone photoreceptors to stop working and die. One theory to explain this two-step disease process relates to the fact that cone photoreceptors are very active cells, requiring a high level of energy, nutrients and oxygen. If surrounding rod cells die, cone photoreceptors may be deprived of some essential supplies, leading to cone cell death and daylight vision loss. To examine this theory, Xue et al. tested a new gene therapy designed to alleviate the potential shortfall in nutrients. The experiments used three different strains of mice that had the same genetic mutations as humans with retinitis pigmentosa. The gene therapy used a virus, called adeno-associated virus (AAV), to deliver 20 different genes to cone cells. Each of the 20 genes tested plays a different role in cells' processing of nutrients to provide energy. After administering the treatment, Xue et al. monitored the mice to see whether or not their vision was affected, and how cone cells responded. Only one of the 20 genes, Txnip, delivered using gene therapy, had a beneficial effect, prolonging cone cell survival in all three mouse strains. The mice that received Txnip also retained their ability to discern moving stripes on vision tests. Further investigations demonstrated that activating Txnip forced the cones to start using a molecule called lactate as an energy source, which could be more available to them than glucose, their usual fuel. These cells also had healthier mitochondria ­ the compartments inside cells that produce and manage energy supplies. This dual effect on fuel use and mitochondrial health is thought to be the basis for the extended cone survival and function. These experiments by Xue et al. have identified a good gene therapy candidate for treating retinitis pigmentosa independently of which genes are causing the disease. Further research will be required to test the safety of the gene therapy, and whether its beneficial effects translate to humans with retinitis pigmentosa, and potentially other diseases with unhealthy photoreceptors.


Subject(s)
Carrier Proteins/genetics , Color Vision/genetics , Dependovirus/physiology , Retinitis Pigmentosa/genetics , Thioredoxins/genetics , Animals , Disease Models, Animal , Mice , Microorganisms, Genetically-Modified/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/physiopathology
9.
Sci Transl Med ; 13(580)2021 02 10.
Article in English | MEDLINE | ID: mdl-33568518

ABSTRACT

Nucleic acids are used in many therapeutic modalities, including gene therapy, but their ability to trigger host immune responses in vivo can lead to decreased safety and efficacy. In the case of adeno-associated viral (AAV) vectors, studies have shown that the genome of the vector activates Toll-like receptor 9 (TLR9), a pattern recognition receptor that senses foreign DNA. Here, we engineered AAV vectors to be intrinsically less immunogenic by incorporating short DNA oligonucleotides that antagonize TLR9 activation directly into the vector genome. The engineered vectors elicited markedly reduced innate immune and T cell responses and enhanced gene expression in clinically relevant mouse and pig models across different tissues, including liver, muscle, and retina. Subretinal administration of higher-dose AAV in pigs resulted in photoreceptor pathology with microglia and T cell infiltration. These adverse findings were avoided in the contralateral eyes of the same animals that were injected with the engineered vectors. However, intravitreal injection of higher-dose AAV in macaques, a more immunogenic route of administration, showed that the engineered vector delayed but did not prevent clinical uveitis, suggesting that other immune factors in addition to TLR9 may contribute to intraocular inflammation in this model. Our results demonstrate that linking specific immunomodulatory noncoding sequences to much longer therapeutic nucleic acids can "cloak" the vector from inducing unwanted immune responses in multiple, but not all, models. This "coupled immunomodulation" strategy may widen the therapeutic window for AAV therapies as well as other DNA-based gene transfer methods.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy , Immunity, Innate , Mice , Swine
10.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33491671

ABSTRACT

Nrf2, a transcription factor that regulates the response to oxidative stress, has been shown to rescue cone photoreceptors and slow vision loss in mouse models of retinal degeneration (rd). The retinal pigment epithelium (RPE) is damaged in these models, but whether it also could be rescued by Nrf2 has not been previously examined. We used an adeno-associated virus (AAV) with an RPE-specific (Best1) promoter to overexpress Nrf2 in the RPE of rd mice. Control rd mice showed disruption of the regular array of the RPE, as well as loss of RPE cells. Cones were lost in circumscribed regions within the cone photoreceptor layer. Overexpression of Nrf2 specifically in the RPE was sufficient to rescue the RPE, as well as the disruptions in the cone photoreceptor layer. Electron microscopy showed compromised apical microvilli in control rd mice but showed preserved microvilli in Best1-Nrf2-treated mice. The rd mice treated with Best1-Nrf2 had slightly better visual acuity. Transcriptome profiling showed that Nrf2 upregulates multiple oxidative defense pathways, reversing declines seen in the glutathione pathway in control rd mice. In summary, Nrf2 overexpression in the RPE preserves RPE morphology and survival in rd mice, and it is a potential therapeutic for diseases involving RPE degeneration, including age-related macular degeneration (AMD).


Subject(s)
NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/physiology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/physiopathology , Retinitis Pigmentosa/therapy , Animals , Disease Models, Animal , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/therapy , Mice , Mice, Mutant Strains , Mice, Transgenic , Microscopy, Electron, Scanning , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Up-Regulation , Visual Acuity/genetics , Visual Acuity/physiology
11.
FASEB J ; 34(8): 10242-10249, 2020 08.
Article in English | MEDLINE | ID: mdl-32539195

ABSTRACT

The nuclei of cone photoreceptors are located on the apical side of the outer nuclear layer (ONL) in vertebrate retinas. However, the functional role of this evolutionarily conserved localization of cone nuclei is unknown. We previously showed that Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) are essential for the apical migration of cone nuclei during development. Here, we developed an efficient genetic strategy to disrupt cone LINC complexes in mice. Experiments with animals from both sexes revealed that disrupting cone LINC complexes resulted in mislocalization of cone nuclei to the basal side of ONL in mouse retina. This, in turn, disrupted cone pedicle morphology, and appeared to reduce the efficiency of synaptic transmission from cones to bipolar cells. Although we did not observe other developmental or phototransduction defects in cones with mislocalized nuclei, their dark adaptation was impaired, consistent with a deficiency in chromophore recycling. These findings demonstrate that the apical localization of cone nuclei in the ONL is required for the timely dark adaptation and efficient synaptic transmission in cone photoreceptors.


Subject(s)
Cell Nucleus/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Animals , Cytoskeleton/physiology , Dark Adaptation/physiology , Female , Male , Mice
12.
J Clin Invest ; 130(8): 4360-4369, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32352930

ABSTRACT

Retinitis pigmentosa (RP) is a genetically heterogenous group of eye diseases in which initial degeneration of rods triggers secondary degeneration of cones, leading to significant loss of daylight, color, and high-acuity vision. Gene complementation with adeno-associated viral (AAV) vectors is one strategy to treat RP. Its implementation faces substantial challenges, however; for example, the tremendous number of loci with causal mutations. Gene therapy targeting secondary cone degeneration is an alternative approach that could provide a much-needed generic treatment for many patients with RP. Here, we show that microglia are required for the upregulation of potentially neurotoxic inflammatory factors during cone degeneration in RP, creating conditions that might contribute to cone dysfunction and death. To ameliorate the effects of such factors, we used AAV vectors to express isoforms of the antiinflammatory cytokine transforming growth factor beta (TGF-ß). AAV-mediated delivery of TGF-ß1 rescued degenerating cones in 3 mouse models of RP carrying different pathogenic mutations. Treatment with TGF-ß1 protected vision, as measured by 2 behavioral assays, and could be pharmacologically disrupted by either depleting microglia or blocking the TGF-ß receptors. Our results suggest that TGF-ß1 may be broadly beneficial for patients with cone degeneration, and potentially other forms of neurodegeneration, through a pathway dependent upon microglia.


Subject(s)
Microglia/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa , Transforming Growth Factor beta1/biosynthesis , Animals , Dependovirus , Disease Models, Animal , Genetic Vectors , Humans , Mice , Mice, Transgenic , Microglia/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/prevention & control , Transduction, Genetic , Transforming Growth Factor beta1/genetics
13.
Proc Natl Acad Sci U S A ; 116(20): 10140-10149, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31036641

ABSTRACT

Retinitis pigmentosa (RP) is a disease that initially presents as night blindness due to genetic deficits in the rod photoreceptors of the retina. Rods then die, causing dysfunction and death of cone photoreceptors, the cell type that mediates high acuity and color vision, ultimately leading to blindness. We investigated immune responses in mouse models of RP and found evidence of microglia activation throughout the period of cone degeneration. Using adeno-associated vectors (AAVs), delivery of genes encoding microglial regulatory signals led to the identification of AAV serotype 8 (AAV8) soluble CX3CL1 (sCX3CL1) as a promising therapy for degenerating cones. Subretinal injection of AAV8-sCX3CL1 significantly prolonged cone survival in three strains of RP mice. Rescue of cones was accompanied by improvements in visual function. AAV8-sCX3CL1 did not affect rod survival, microglia localization, or inflammatory cytokine levels in the retina. Furthermore, although RNA sequencing of microglia demonstrated marked transcriptional changes with AAV8-sCX3CL1, pharmacological depletion of up to ∼99% of microglia failed to abrogate the effect of AAV8-sCX3CL1 on cone survival. These findings indicate that AAV8-sCX3CL1 can rescue cones in multiple mouse models of RP via a pathway that does not require normal numbers of microglia. Gene therapy with sCX3CL1 is a promising mutation-independent approach to preserve vision in RP and potentially other forms of retinal degeneration.


Subject(s)
Chemokine CX3CL1/genetics , Genetic Therapy/methods , Microglia/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/therapy , Animals , Dependovirus , Disease Models, Animal , Mice , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/immunology , Vision, Ocular
14.
Proc Natl Acad Sci U S A ; 116(12): 5785-5794, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30833387

ABSTRACT

Adeno-associated viral vectors (AAVs) have become popular for gene therapy, given their many advantages, including their reduced inflammatory profile compared with that of other viruses. However, even in areas of immune privilege such as the eye, AAV vectors are capable of eliciting host-cell responses. To investigate the effects of such responses on several ocular cell types, we tested multiple AAV genome structures and capsid types using subretinal injections in mice. Assays of morphology, inflammation, and physiology were performed. Pathological effects on photoreceptors and the retinal pigment epithelium (RPE) were observed. Müller glia and microglia were activated, and the proinflammatory cytokines TNF-α and IL-1ß were up-regulated. There was a strong correlation between cis-regulatory sequences and toxicity. AAVs with any one of three broadly active promoters, or an RPE-specific promoter, were toxic, while AAVs with four different photoreceptor-specific promoters were not toxic at the highest doses tested. There was little correlation between toxicity and transgene, capsid type, preparation method, or cellular contaminants within a preparation. The toxic effect was dose-dependent, with the RPE being more sensitive than photoreceptors. Our results suggest that ocular AAV toxicity is associated with certain AAV cis-regulatory sequences and/or their activity and that retinal damage occurs due to responses by the RPE and/or microglia. By applying multiple, sensitive assays of toxicity, AAV vectors can be designed so that they can be used safely at high dose, potentially providing greater therapeutic efficacy.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Transduction, Genetic/methods , Animals , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Mice , Mice, Inbred C57BL , Photoreceptor Cells/metabolism , Promoter Regions, Genetic/genetics , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Transgenes , Vision, Ocular/genetics , Vision, Ocular/physiology
15.
Sci Rep ; 7(1): 2390, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539612

ABSTRACT

Pigment regeneration is critical for the function of cone photoreceptors in bright and rapidly-changing light conditions. This process is facilitated by the recently-characterized retina visual cycle, in which Müller cells recycle spent all-trans-retinol visual chromophore back to 11-cis-retinol. This 11-cis-retinol is oxidized selectively in cones to the 11-cis-retinal used for pigment regeneration. However, the enzyme responsible for the oxidation of 11-cis-retinol remains unknown. Here, we sought to determine whether retinol dehydrogenase 10 (RDH10), upregulated in rod/cone hybrid retinas and expressed abundantly in Müller cells, is the enzyme that drives this reaction. We created mice lacking RDH10 either in cone photoreceptors, Müller cells, or the entire retina. In vivo electroretinography and transretinal recordings revealed normal cone photoresponses in all RDH10-deficient mouse lines. Notably, their cone-driven dark adaptation both in vivo and in isolated retina was unaffected, indicating that RDH10 is not required for the function of the retina visual cycle. We also generated transgenic mice expressing RDH10 ectopically in rod cells. However, rod dark adaptation was unaffected by the expression of RDH10 and transgenic rods were unable to use cis-retinol for pigment regeneration. We conclude that RDH10 is not the dominant retina 11-cis-RDH, leaving its primary function in the retina unknown.


Subject(s)
Alcohol Oxidoreductases/genetics , Ependymoglial Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Dark Adaptation/physiology , Electroretinography , Ependymoglial Cells/cytology , Gene Expression , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Oxidation-Reduction , Retinal Cone Photoreceptor Cells/cytology , Retinal Pigment Epithelium/cytology , Retinal Rod Photoreceptor Cells/cytology , Retinaldehyde/metabolism , Transgenes , Vision, Ocular/physiology , Vitamin A/metabolism
16.
Mol Biol Cell ; 27(12): 1928-37, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27075175

ABSTRACT

Lamin B1 and lamin B2 are essential building blocks of the nuclear lamina, a filamentous meshwork lining the nucleoplasmic side of the inner nuclear membrane. Deficiencies in lamin B1 and lamin B2 impair neurodevelopment, but distinct functions for the two proteins in the development and homeostasis of the CNS have been elusive. Here we show that embryonic depletion of lamin B1 in retinal progenitors and postmitotic neurons affects nuclear integrity, leads to the collapse of the laminB2 meshwork, impairs neuronal survival, and markedly reduces the cellularity of adult retinas. In stark contrast, a deficiency of lamin B2 in the embryonic retina has no obvious effect on lamin B1 localization or nuclear integrity in embryonic retinas, suggesting that lamin B1, but not lamin B2, is strictly required for nucleokinesis during embryonic neurogenesis. However, the absence of lamin B2 prevents proper lamination of adult retinal neurons, impairs synaptogenesis, and reduces cone photoreceptor survival. We also show that lamin B1 and lamin B2 are extremely long-lived proteins in rod and cone photoreceptors. OF interest, a complete absence of both proteins during postnatal life has little or no effect on the survival and function of cone photoreceptors.


Subject(s)
Lamin Type B/metabolism , Animals , Lamin Type B/genetics , Mice , Mice, Knockout , Neurogenesis/physiology , Neurons/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Retina/embryology , Retina/metabolism , Retina/physiology
17.
Sci Rep ; 5: 17616, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626567

ABSTRACT

Continuous visual perception and the dark adaptation of vertebrate photoreceptors after bright light exposure require recycling of their visual chromophore through a series of reactions in the retinal pigmented epithelium (RPE visual cycle). Light-driven chromophore consumption by photoreceptors is greater in daytime vs. nighttime, suggesting that correspondingly higher activity of the visual cycle may be required. However, as rod photoreceptors are saturated in bright light, the continuous turnover of their chromophore by the visual cycle throughout the day would not contribute to vision. Whether the recycling of chromophore that drives rod dark adaptation is regulated by the circadian clock and light exposure is unknown. Here, we demonstrate that mouse rod dark adaptation is slower during the day or after light pre-exposure. This surprising daytime suppression of the RPE visual cycle was accompanied by light-driven reduction in expression of Rpe65, a key enzyme of the RPE visual cycle. Notably, only rods in melatonin-proficient mice were affected by this daily visual cycle modulation. Our results demonstrate that the circadian clock and light exposure regulate the recycling of chromophore in the RPE visual cycle. This daily melatonin-driven modulation of rod dark adaptation could potentially protect the retina from light-induced damage during the day.


Subject(s)
Circadian Clocks/physiology , Dark Adaptation/physiology , Light , Retinal Rod Photoreceptor Cells/metabolism , Vision, Ocular/physiology , Animals , Mice , Mice, Mutant Strains , Retinal Rod Photoreceptor Cells/cytology
18.
J Clin Invest ; 125(2): 727-38, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25607845

ABSTRACT

Mutations in the cellular retinaldehyde-binding protein (CRALBP, encoded by RLBP1) can lead to severe cone photoreceptor-mediated vision loss in patients. It is not known how CRALBP supports cone function or how altered CRALBP leads to cone dysfunction. Here, we determined that deletion of Rlbp1 in mice impairs the retinal visual cycle. Mice lacking CRALBP exhibited M-opsin mislocalization, M-cone loss, and impaired cone-driven visual behavior and light responses. Additionally, M-cone dark adaptation was largely suppressed in CRALBP-deficient animals. While rearing CRALBP-deficient mice in the dark prevented the deterioration of cone function, it did not rescue cone dark adaptation. Adeno-associated virus-mediated restoration of CRALBP expression specifically in Müller cells, but not retinal pigment epithelial (RPE) cells, rescued the retinal visual cycle and M-cone sensitivity in knockout mice. Our results identify Müller cell CRALBP as a key component of the retinal visual cycle and demonstrate that this pathway is important for maintaining normal cone-driven vision and accelerating cone dark adaptation.


Subject(s)
Carrier Proteins/metabolism , Dark Adaptation/physiology , Ependymoglial Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Vision, Ocular/physiology , Animals , Blindness/genetics , Blindness/metabolism , Blindness/pathology , Carrier Proteins/genetics , Dependovirus , Ependymoglial Cells/pathology , Mice , Mice, Knockout , Opsins/genetics , Opsins/metabolism , Protein Transport/physiology , Retinal Cone Photoreceptor Cells/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...