Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Exp Biol ; 223(Pt 11)2020 05 29.
Article in English | MEDLINE | ID: mdl-32371443

ABSTRACT

An animal's temporal niche - the time of day at which it is active - is known to drive a variety of adaptations in the visual system. These include variations in the topography, spectral sensitivity and density of retinal photoreceptors, and changes in the eye's gross anatomy and spectral transmission characteristics. We have characterised visual spectral sensitivity in the murid rodent Rhabdomys pumilio (the four-striped grass mouse), which is in the same family as (nocturnal) mice and rats but exhibits a strong diurnal niche. As is common in diurnal species, the R. pumilio lens acts as a long-pass spectral filter, providing limited transmission of light <400 nm. Conversely, we found strong sequence homologies with the R. pumilio SWS and MWS opsins and those of related nocturnal species (mice and rats) whose SWS opsins are maximally sensitive in the near-UV. We continued to assess in vivo spectral sensitivity of cone vision using electroretinography and multi-channel recordings from the visual thalamus. These revealed that responses across the human visible range could be adequately described by those of a single pigment (assumed to be MWS opsin) maximally sensitive at ∼500 nm, but that sensitivity in the near-UV required inclusion of a second pigment whose peak sensitivity lay well into the UV range (λmax<400 nm, probably ∼360 nm). We therefore conclude that, despite the UV-filtering effects of the lens, R. pumilio retains an SWS pigment with a UV-A λmax In effect, this somewhat paradoxical combination of long-pass lens and UV-A λmax results in narrow-band sensitivity for SWS cone pathways in the UV-A range.


Subject(s)
Retinal Cone Photoreceptor Cells , Vision, Ocular , Animals , Mice , Opsins , Photoreceptor Cells, Vertebrate , Rats , Rod Opsins
2.
Stem Cells Transl Med ; 8(8): 775-784, 2019 08.
Article in English | MEDLINE | ID: mdl-31037833

ABSTRACT

Glaucoma is one of the leading causes of blindness, and there is an ongoing need for new therapies. Recent studies indicate that cell transplantation using Müller glia may be beneficial, but there is a need for novel sources of cells to provide therapeutic benefit. In this study, we have isolated Müller glia from retinal organoids formed by human induced pluripotent stem cells (hiPSCs) in vitro and have shown their ability to partially restore visual function in rats depleted of retinal ganglion cells by NMDA. Based on the present results, we suggest that Müller glia derived from retinal organoids formed by hiPSC may provide an attractive source of cells for human retinal therapies, to prevent and treat vision loss caused by retinal degenerative conditions. Stem Cells Translational Medicine 2019;8:775&784.


Subject(s)
Cell Transplantation/methods , Ependymoglial Cells/transplantation , Induced Pluripotent Stem Cells/cytology , Retinal Degeneration/therapy , Retinal Ganglion Cells/physiology , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Ependymoglial Cells/cytology , Humans , Induced Pluripotent Stem Cells/transplantation , Organoids/cytology , Phenotype , Rats , Regeneration , Retinal Ganglion Cells/pathology
3.
Mol Ther Nucleic Acids ; 12: 730-740, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30114557

ABSTRACT

Leber congenital amaurosis type 10 (LCA10) is a severe inherited retinal dystrophy associated with mutations in CEP290. The deep intronic c.2991+1655A>G mutation in CEP290 is the most common mutation in LCA10 individuals and represents an ideal target for oligonucleotide therapeutics. Here, a panel of antisense oligonucleotides was designed to correct the splicing defect associated with the mutation and screened for efficacy and safety. This identified QR-110 as the best-performing molecule. QR-110 restored wild-type CEP290 mRNA and protein expression levels in CEP290 c.2991+1655A>G homozygous and compound heterozygous LCA10 primary fibroblasts. Furthermore, in homozygous three-dimensional iPSC-derived retinal organoids, QR-110 showed a dose-dependent restoration of mRNA and protein function, as measured by percentage and length of photoreceptor cilia, without off-target effects. Localization studies in wild-type mice and rabbits showed that QR-110 readily reached all retinal layers, with an estimated half-life of 58 days. It was well tolerated following intravitreal injection in monkeys. In conclusion, the pharmacodynamic, pharmacokinetic, and safety properties make QR-110 a promising candidate for treating LCA10, and clinical development is currently ongoing.

4.
Nat Biotechnol ; 36(4): 328-337, 2018 04.
Article in English | MEDLINE | ID: mdl-29553577

ABSTRACT

Age-related macular degeneration (AMD) remains a major cause of blindness, with dysfunction and loss of retinal pigment epithelium (RPE) central to disease progression. We engineered an RPE patch comprising a fully differentiated, human embryonic stem cell (hESC)-derived RPE monolayer on a coated, synthetic basement membrane. We delivered the patch, using a purpose-designed microsurgical tool, into the subretinal space of one eye in each of two patients with severe exudative AMD. Primary endpoints were incidence and severity of adverse events and proportion of subjects with improved best-corrected visual acuity of 15 letters or more. We report successful delivery and survival of the RPE patch by biomicroscopy and optical coherence tomography, and a visual acuity gain of 29 and 21 letters in the two patients, respectively, over 12 months. Only local immunosuppression was used long-term. We also present the preclinical surgical, cell safety and tumorigenicity studies leading to trial approval. This work supports the feasibility and safety of hESC-RPE patch transplantation as a regenerative strategy for AMD.


Subject(s)
Human Embryonic Stem Cells/transplantation , Macular Degeneration/therapy , Retinal Pigment Epithelium/transplantation , Visual Acuity/physiology , Aged , Animals , Basement Membrane/diagnostic imaging , Basement Membrane/growth & development , Cell Differentiation/genetics , Female , Humans , Macular Degeneration/diagnostic imaging , Macular Degeneration/pathology , Male , Mice , Middle Aged , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/growth & development , Stem Cell Transplantation/adverse effects , Swine , Tomography, Optical Coherence
5.
Am J Hum Genet ; 102(4): 528-539, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29526280

ABSTRACT

Fuchs endothelial corneal dystrophy (FECD) is a common disease for which corneal transplantation is the only treatment option in advanced stages, and alternative treatment strategies are urgently required. Expansion (≥50 copies) of a non-coding trinucleotide repeat in TCF4 confers >76-fold risk for FECD in our large cohort of affected individuals. An FECD subject-derived corneal endothelial cell (CEC) model was developed to probe disease mechanism and investigate therapeutic approaches. The CEC model demonstrated that the repeat expansion leads to nuclear RNA foci, with the sequestration of splicing factor proteins (MBNL1 and MBNL2) to the foci and altered mRNA processing. Antisense oligonucleotide (ASO) treatment led to a significant reduction in the incidence of nuclear foci, MBNL1 recruitment to the foci, and downstream aberrant splicing events, suggesting functional rescue. This proof-of-concept study highlights the potential of a targeted ASO therapy to treat the accessible and tractable corneal tissue affected by this repeat expansion-mediated disease.


Subject(s)
Fuchs' Endothelial Dystrophy/genetics , Genetic Predisposition to Disease , Oligonucleotides, Antisense/pharmacology , Transcription Factor 4/genetics , Trinucleotide Repeat Expansion/genetics , Aged , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cohort Studies , Endothelial Cells/metabolism , Endothelium, Corneal/pathology , Female , Fuchs' Endothelial Dystrophy/pathology , Humans , Male , Mice, Inbred C57BL , Organ Specificity , RNA Precursors/genetics , RNA Processing, Post-Transcriptional , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , Risk Factors
6.
Transl Vis Sci Technol ; 5(4): 6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27486556

ABSTRACT

PURPOSE: We assessed the long-term efficacy and safety of human retinal progenitor cells (hRPC) using established rodent models. METHODS: Efficacy of hRPC was tested initially in Royal College of Surgeons (RCS) dystrophic rats immunosuppressed with cyclosporine/dexamethasone. Due to adverse effects of dexamethasone, this drug was omitted from a subsequent dose-ranging study, where different hRPC doses were tested for their ability to preserve visual function (measured by optokinetic head tracking) and retinal structure in RCS rats at 3 to 6 months after grafting. Safety of hRPC was assessed by subretinal transplantation into wild type (WT) rats and NIH-III nude mice, with analysis at 3 to 6 and 9 months after grafting, respectively. RESULTS: The optimal dose of hRPC for preserving visual function/retinal structure in dystrophic rats was 50,000 to 100,000 cells. Human retinal progenitor cells integrated/survived in dystrophic and WT rat retina up to 6 months after grafting and expressed nestin, vimentin, GFAP, and ßIII tubulin. Vision and retinal structure remained normal in WT rats injected with hRPC and there was no evidence of tumors. A comparison between dexamethasone-treated and untreated dystrophic rats at 3 months after grafting revealed an unexpected reduction in the baseline visual acuity of dexamethasone-treated animals. CONCLUSIONS: Human retinal progenitor cells appear safe and efficacious in the preclinical models used here. TRANSLATIONAL RELEVANCE: Human retinal progenitor cells could be deployed during early stages of retinal degeneration or in regions of intact retina, without adverse effects on visual function. The ability of dexamethasone to reduce baseline visual acuity in RCS dystrophic rats has important implications for the interpretation of preclinical and clinical cell transplant studies.

7.
Invest Ophthalmol Vis Sci ; 57(1): 115-25, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26780315

ABSTRACT

PURPOSE: Following on from reports of retrograde retinal signaling in mice, we sought to investigate the influence of age and retinal location on this phenomenon using mice that lack rods and the majority of cones. METHODS: We used functional anatomy for c-fos (Fos) and tyrosine hydroxylase (TH) to measure light-driven activation of dopamine neurons along a dorsal-ventral transect in C3H/He wild-type and rodless-coneless rd/rd cl (rdcl) mice aged 3, 5, and >14 months. A parallel series of retinae from 3-month-old mice was also stained for cone opsins and melanopsin. RESULTS: Analysis by confocal microscopy revealed light-driven Fos activation in TH cells residing in the middorsal retina of the youngest rdcl mice. This region was largely devoid of residual cones but contained a large number of intrinsically photosensitive retinal ganglion cells (ipRGCs) and the highest density of melanopsin neurites. With advancing age, there was a paradoxical increase in retrograde signaling from ∼3% Fos-positive (Fos+) TH cells at 3 months to ∼36% in rdcl mice >14 months. This increased activation occurred in more central and peripheral retinal regions. CONCLUSIONS: Our data provide new insights into the anatomy and plasticity of retrograde melanopsin signaling in mice with severe rod/cone dystrophy. The increased retrograde signaling we detect may result from either an increased potency of melanopsin signaling with advancing age and/or postsynaptic modification to dopaminergic neurons.


Subject(s)
Aging , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Animals , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C3H , Photic Stimulation , Photoreceptor Cells, Vertebrate/pathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Signal Transduction
8.
Front Neuroanat ; 8: 131, 2014.
Article in English | MEDLINE | ID: mdl-25477787

ABSTRACT

Here we have studied the population of intrinsically photosensitive retinal ganglion cells (ipRGCs) in adult pigmented and albino mice. Our data show that although pigmented (C57Bl/6) and albino (Swiss) mice have a similar total number of ipRGCs, their distribution is slightly different: while in pigmented mice ipRGCs are more abundant in the temporal retina, in albinos the ipRGCs are more abundant in superior retina. In both strains, ipRGCs are located in the retinal periphery, in the areas of lower Brn3a(+)RGC density. Both strains also contain displaced ipRGCs (d-ipRGCs) in the inner nuclear layer (INL) that account for 14% of total ipRGCs in pigmented mice and 5% in albinos. Tracing from both superior colliculli shows that 98% (pigmented) and 97% (albino) of the total ipRGCs, become retrogradely labeled, while double immunodetection of melanopsin and Brn3a confirms that few ipRGCs express this transcription factor in mice. Rather surprisingly, application of a retrograde tracer to the optic nerve (ON) labels all ipRGCs, except for a sub-population of the d-ipRGCs (14% in pigmented and 28% in albino, respectively) and melanopsin positive cells residing in the ciliary marginal zone (CMZ) of the retina. In the CMZ, between 20% (pigmented) and 24% (albino) of the melanopsin positive cells are unlabeled by the tracer and we suggest that this may be because they fail to send an axon into the ON. As such, this study provides the first evidence for a population of melanopsin interneurons in the mammalian retina.

9.
Exp Eye Res ; 119: 8-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316157

ABSTRACT

Maintenance of pupillary constriction in light-adapted rodents has traditionally been thought to involve a reflex between retina, brain and iris, with recent work identifying the melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) as the major conduits for retinal input to the brain. There is also a less well-understood phenomenon whereby the iris of some mammals, including mice, will constrict to light when either the eye, or the iris itself is physically isolated from the brain. The intrinsic pupillary light reflex (iPLR) is the term given to pupil constriction in the absence of retinal input to the brain. Here, using an intraocular axotomy approach, we show that the iPLR in conscious mice spans a dynamic range over 3 log units of irradiance. This iPLR response is absent in melanopsin knockout (MKO) mice and can be significantly inhibited by atropine. Immunohistochemistry for cfos and melanopsin, in combination with light exposure revealed a population of small ipRGCs in the retinal ciliary marginal zone (CMZ), which remain responsive to light in axotomised mice. We report that damage to the CMZ in a novel in vitro preparation removes a significant component of the iPLR response, while a detailed immunohistochemical analysis of the CMZ in wildtype mice revealed a melanopsin-rich plexus, which was consistently most intense in nasal retina. There were clear examples of melanopsin-positive, direct retino-ciliary projections, which appear to emanate from Brn3b negative, M1 type ipRGCs. These cells are clustered along the melanopsin-rich plexus nasally and may channel ipRGC signals from retina into the iris via ciliary body. Comparison between wildtype and MKO mice reveals that the ciliary body is also weakly stained for melanopsin. Our results show that the full extent of iPLR in mice requires cholinergic neurotransmission and intact signalling at the CMZ/ciliary body. This response may be mediated to some extent by ipRGCs, which send direct projections from the retina into ciliary body. In addition to the melanopsin-mediated iris sphincter constriction suggested by others, we propose a new mechanism, which may involve constriction of the ciliary body and ipRGC-mediated relaxation of the iris dilator muscle.


Subject(s)
Ciliary Body/cytology , Reflex, Pupillary/drug effects , Rod Opsins/pharmacology , Animals , Light , Light Signal Transduction/drug effects , Mice , Photic Stimulation/methods
10.
Curr Biol ; 22(12): 1134-41, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22633808

ABSTRACT

Photoreception in the mammalian retina is not restricted to rods and cones but extends to a small number of intrinsically photoreceptive retinal ganglion cells (ipRGCs), expressing the photopigment melanopsin. ipRGCs are known to support various accessory visual functions including circadian photoentrainment and pupillary reflexes. However, despite anatomical and physiological evidence that they contribute to the thalamocortical visual projection, no aspect of visual discrimination has been shown to rely upon ipRGCs. Based on their currently known roles, we hypothesized that ipRGCs may contribute to distinguishing brightness. This percept is related to an object's luminance-a photometric measure of light intensity relevant for cone photoreceptors. However, the perceived brightness of different sources is not always predicted by their respective luminance. Here, we used parallel behavioral and electrophysiological experiments to first show that melanopsin contributes to brightness discrimination in both retinally degenerate and fully sighted mice. We continued to use comparable paradigms in psychophysical experiments to provide evidence for a similar role in healthy human subjects. These data represent the first direct evidence that an aspect of visual discrimination in normally sighted subjects can be supported by inner retinal photoreceptors.


Subject(s)
Discrimination, Psychological/physiology , Light Signal Transduction/physiology , Light , Retinal Ganglion Cells/physiology , Rod Opsins/metabolism , Visual Perception/physiology , Adult , Animals , Humans , Light Signal Transduction/genetics , Mice , Nuclear Proteins/genetics , Photic Stimulation , Photometry , RNA-Binding Proteins , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/metabolism , Rod Opsins/physiology
11.
Vision Res ; 51(20): 2176-85, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21871912

ABSTRACT

The purpose of the current study was to determine the progress of cortical functional degeneration in the Royal College of Surgeons (RCS) rat. Cortical responses were measured with optical imaging of intrinsic signals using gratings of various spatial frequencies. Subsequently, electrophysiological recordings were also taken across cortical layers in response to a pulse of broad-spectrum light. We found significant degeneration in the cortical processing of visual information as early as 4 weeks of age. These results show that degeneration in the cortical response of the RCS rat starts before development has been properly completed.


Subject(s)
Retinal Degeneration/physiopathology , Visual Cortex/physiology , Visual Perception/physiology , Analysis of Variance , Animals , Electroretinography , Female , Light , Male , Models, Animal , Pattern Recognition, Visual/physiology , Photic Stimulation/methods , Rats , Sensory Thresholds/physiology
12.
Pharmacol Biochem Behav ; 100(1): 48-58, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820464

ABSTRACT

While prior exposure to drugs of abuse permanently changes many behaviors, the underlying psychological mechanisms are relatively obscure. Here, the effects of sensitization on the detection of an action-outcome relationship were assessed, using a particularly stringent contingency degradation procedure. Rats were trained to leverpress until the probability of reinforcement for a response on one lever, or alternative reinforcement for a response on a second lever was reduced to 0.05 per second. Sensitization was then carried out (1mg/kg d-amphetamine/day for 7 days). Then, one reinforcer was also made available for a lack of response on either lever (p=0.05/s), maintaining its contiguity with the original response but eliminating its contingent relationship. Sensitized animals were more active, particularly early in the contingency degradation phase, but reduced responding directed at the degraded action-outcome contingency at a similar rate as controls. However, controls also reduced responding directed at the nondegraded contingency until very late in training, while sensitized animals maintained nondegraded responding at baseline levels. It was suggested that the relatively specific response shown by sensitized animals may reflect either improved action-outcome utilization or discrimination of relevant task features.


Subject(s)
Amphetamine/administration & dosage , Conditioning, Operant/physiology , Psychomotor Performance/physiology , Reinforcement, Psychology , Animals , Conditioning, Operant/drug effects , Male , Psychomotor Performance/drug effects , Rats , Self Administration
13.
Mol Vis ; 17: 1701-15, 2011.
Article in English | MEDLINE | ID: mdl-21738400

ABSTRACT

PURPOSE: In several species the retinal pigment epithelium (RPE) has the potential to transdifferentiate into retinal cells to regenerate functional retinal tissue after injury. However, this capacity for regeneration is lost in mammals. The synthetic retinoic acid derivative, fenretinide [N(4-hydroxyphenyl) retinamide], induces a neuronal-like phenotype in the human adult retinal pigment epithelial cell line (ARPE-19). These changes are characterized by the appearance of neural-like processes and the expression of neuronal markers not normally associated with RPE cells. Here we assess whether fenretinide can induce a neuroretinal cell phenotype in ARPE-19 cells, by examining retinal cell marker expression. METHODS: ARPE-19 cells were treated daily with culture medium containing either 3 µM fenretinide or dimethyl sulfoxide as a control for 7 days. Cells were processed for immunocytochemistry, western blotting, and for analysis by PCR to examine the expression of a panel of RPE, neural, and retinal-associated cellular markers, including classical and non-canonical opsins. RESULTS: Treatment with fenretinide for 7 days induced the formation of neuronal-like processes in ARPE-19 cells. Fenretinide induced the expression of the cone long wavelength sensitive opsin (OPN1lw) but not rhodopsin (RHO), while decreasing the expression of RPE cell markers. Many of the neuronal and retinal specific markers examined were expressed in both control and fenretinide treated cells, including those involved in photoreceptor cell development and the multipotency of neural retinal progenitor cells. Interestingly, ARPE-19 cells also expressed both photoreceptor specific and non-specific canonical opsins. CONCLUSIONS: The expression of retinal-associated markers and loss of RPE cell markers in control ARPE-19 cells suggests that these cells might have dedifferentiated from an RPE cell phenotype under standard culture conditions. The expression of molecules, such as the transcription factors paired box 6 gene (PAX6), sex determining region Y-box 2 (SOX2), cone-rod homeobox (CRX), and neural retina leucine zipper (NRL), further implies that in culture these cells are predisposed toward a retinal progenitor-like state. The fenretinide-induced increase in photoreceptor cell markers, accompanied by a decrease in RPE cell markers, suggests that retinoids may play a role in the transdifferentiation of RPE cells. Importantly, our data show for the first time the expression of a vertebrate ciliary opsin (OPN1lw) and rhabdomeric-like opsin, opsin 4 (OPN4 also known as melanopsin) in a clonal cell line. Together these data suggest that ARPE-19 cells are primed for and possess the capacity to differentiate toward a retinal cell-like lineage.


Subject(s)
Biomarkers/metabolism , Cell Transdifferentiation/drug effects , Epithelial Cells/drug effects , Fenretinide/pharmacology , Retina/drug effects , Retinal Pigment Epithelium/drug effects , Adult , Blotting, Western , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression/drug effects , Humans , Immunohistochemistry , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Polymerase Chain Reaction , Retina/cytology , Retina/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinoids/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Invest Ophthalmol Vis Sci ; 52(10): 7148-59, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21743014

ABSTRACT

PURPOSE: Cultured retinal pigment epithelium (RPE) may become a therapeutic option for transplantation in retinal disease. However maintaining a native RPE phenotype in vitro has proven challenging. The human RPE cell-line ARPE-19 is used widely as an alternative to primary RPE. It is grown in DMEM/F12 medium as standard, but its phenotype is dependent on culture conditions, and many differentiation markers are usually absent. The purpose of this study was to examine how this sensitive phenotype of ARPE-19 can be modulated by growth media with or without the metabolite pyruvate to elucidate better RPE growth conditions. METHODS: ARPE-19 cells at passages p22 to p28 were cultured on filters for up to 3 months in DMEM/F12 or DMEM media with or without pyruvate and 1% fetal calf serum. Assessment of differentiation was performed using pigmentation, immunocytochemistry, protein/mRNA expression, transepithelial resistance, VEGF secretion, and ultrastructure. RESULTS: Pyruvate, in combination with DMEM, induced dark pigmentation and promoted differentiation markers such as CRALBP and MerTK. Importantly, RPE65 protein was detected by Western blotting and was enhanced by pyruvate, high glucose, and DMEM. ARPE-19 cells maintained in this medium could also phagocytose human photoreceptor outer segments (POS). VEGF secretion was greater in DMEM cultures and was affected by glucose but not by pyruvate. Pigmentation never occurred in DMEM/F12. CONCLUSIONS: This study demonstrated important differentiation markers, including pigmentation and Western blots of RPE65 protein, and showed human POS phagocytosis in ARPE-19 cultures using a simple differentiation protocol. The results favor the use of high-glucose DMEM with pyruvate for future RPE differentiation studies.


Subject(s)
Cell Differentiation/drug effects , Pyruvic Acid/pharmacology , Retinal Pigment Epithelium/cytology , Biomarkers/metabolism , Blotting, Western , Carrier Proteins/metabolism , Cells, Cultured , Culture Media/pharmacology , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Phagocytosis , Phenotype , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , c-Mer Tyrosine Kinase , cis-trans-Isomerases
15.
PLoS One ; 5(11): e15063, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-21124780

ABSTRACT

The mammalian visual system relies upon light detection by outer-retinal rod/cone photoreceptors and melanopsin-expressing retinal ganglion cells. Gnat1(-/-);Cnga3(-/-);Opn4(-/-) mice lack critical elements of each of these photoreceptive mechanisms via targeted disruption of genes encoding rod α transducin (Gnat1); the cone-specific α3 cyclic nucleotide gated channel subunit (Cnga3); and melanopsin (Opn4). Although assumed blind, we show here that these mice retain sufficiently widespread retinal photoreception to drive a reproducible flash electroretinogram (ERG). The threshold sensitivity of this ERG is similar to that of cone-based responses, however it is lost under light adapted conditions. Its spectral efficiency is consistent with that of rod opsin, but not cone opsins or melanopsin, indicating that it originates with light absorption by the rod pigment. The TKO light response survives intravitreal injection of U73122 (a phospholipase C antagonist), but is inhibited by a missense mutation of cone α transducin (Gnat2(cpfl3)), suggesting Gnat2-dependence. Visual responses in TKO mice extend beyond the retina to encompass the lateral margins of the lateral geniculate nucleus and components of the visual cortex. Our data thus suggest that a Gnat1-independent phototransduction mechanism downstream of rod opsin can support relatively widespread responses in the mammalian visual system. This anomalous rod opsin-based vision should be considered in experiments relying upon Gnat1 knockout to silence rod phototransduction.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Light Signal Transduction/physiology , Rod Opsins/metabolism , Transducin/metabolism , Visual Cortex/enzymology , Visual Perception/physiology , Adaptation, Ocular/drug effects , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Electroretinography , Estrenes/pharmacology , GTP-Binding Protein alpha Subunits/genetics , Geniculate Bodies/metabolism , Geniculate Bodies/physiology , Light Signal Transduction/genetics , Male , Mice , Mice, Knockout , Phosphodiesterase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Retina/metabolism , Retina/physiology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/physiology , Rod Opsins/genetics , Transducin/genetics , Visual Cortex/metabolism , Visual Cortex/physiology , Visual Perception/genetics
16.
PLoS One ; 5(11): e15009, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-21124784

ABSTRACT

Melanopsin photoreception plays a vital role in irradiance detection for non-image forming responses to light. However, little is known about the involvement of melanopsin in emotional processing of luminance. When confronted with a gradient in light, organisms exhibit spatial movements relative to this stimulus. In rodents, behavioural light aversion (BLA) is a well-documented but poorly understood phenomenon during which animals attribute salience to light and remove themselves from it. Here, using genetically modified mice and an open field behavioural paradigm, we investigate the role of melanopsin in BLA. While wildtype (WT), melanopsin knockout (Opn4(-/-)) and rd/rd cl (melanopsin only (MO)) mice all exhibit BLA, our novel methodology reveals that isolated melanopsin photoreception produces a slow, potentiating response to light. In order to control for the involvement of pupillary constriction in BLA we eliminated this variable with topical atropine application. This manipulation enhanced BLA in WT and MO mice, but most remarkably, revealed light aversion in triple knockout (TKO) mice, lacking three elements deemed essential for conventional photoreception (Opn4(-/-) Gnat1(-/-) Cnga3(-/-)). Using a number of complementary strategies, we determined this response to be generated at the level of the retina. Our findings have significant implications for the understanding of how melanopsin signalling may modulate aversive responses to light in mice and humans. In addition, we also reveal a clear potential for light perception in TKO mice.


Subject(s)
Avoidance Learning/radiation effects , Light , Motor Activity/radiation effects , Rod Opsins/physiology , Animals , Atropine/pharmacology , Avoidance Learning/drug effects , Channelrhodopsins , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/physiology , Electroretinography , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/physiology , Humans , Immunohistochemistry , Kinetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Parasympatholytics/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Regression Analysis , Retina/metabolism , Rod Opsins/genetics , Transducin/genetics , Transducin/physiology , Visual Cortex/metabolism , Visual Cortex/radiation effects
17.
Retina ; 30(7): 983-1001, 2010.
Article in English | MEDLINE | ID: mdl-20616678

ABSTRACT

BACKGROUND: Retinal diseases such as age-related macular degeneration and retinitis pigmentosa remain major causes of severe vision loss in humans. Clinical trials for treatment of retinal degenerations are underway and advancements in our understanding of retinal biology in health/disease have implications for novel therapies. METHODS: A review of retinal biology is used to inform a discussion of current strategies to maintain/repair neural circuitry in age-related macular degeneration, retinitis pigmentosa, and Type 2 Leber congenital amaurosis. RESULTS: In age-related macular degeneration/retinitis pigmentosa, a progressive loss of rods/cones results in corruption of bipolar cell circuitry, although retinal output neurons/photoreceptive melanopsin cells survive. Visual function can be stabilized/enhanced after treatment in age-related macular degeneration, but in advanced degenerations, reorganization of retinal circuitry may preclude attempts to restore cone function. In Type 2 Leber congenital amaurosis, useful vision can be restored by gene therapy where central cones survive. Remarkable progress has been made in restoring vision to rodents using light-responsive ion channels inserted into bipolar cells/retinal ganglion cells. CONCLUSION: Advances in genetic, cellular, and prosthetic therapies show varying degrees of promise for treating retinal degenerations. While functional benefits can be obtained after early therapeutic interventions, efforts should be made to minimize circuitry changes as soon as possible after rod/cone loss. Advances in retinal anatomy/physiology and genetic technologies should allow refinement of future reparative strategies.


Subject(s)
Photoreceptor Cells, Vertebrate/physiology , Retinal Degeneration/physiopathology , Retinal Degeneration/therapy , Visual Pathways/physiopathology , Humans
18.
Cell Transplant ; 19(10): 1291-306, 2010.
Article in English | MEDLINE | ID: mdl-20447347

ABSTRACT

Human fetal retinal cells have been widely advocated for the development of cellular replacement therapies in patients with retinal dystrophies and age-related macular degeneration. A major limitation, however, is the lack of an abundant and renewable source of cells to meet therapeutic demand, although theoretically this may be addressed through the use of immortalized retinal progenitor cell lines. Here, we have used the temperature-sensitive tsA58 simian virus SV40 T antigen to conditionally immortalize human retinal progenitor cells isolated from retinal tissue at 10-12 weeks of gestation. We show that immortalized human fetal retinal cells retain their progenitor cell properties over many passages, and are comparable with nonimmortalized human fetal retinal cultures from the same gestational period with regard to expression of certain retinal genes. To evaluate the capacity of these cells to integrate into the diseased retina and to screen for potential tumorigenicity, cells were grafted into neonatal hooded Lister rats and RCS dystrophic rats. Both cell lines exhibited scarce integration into the host retina and failed to express markers of mature differentiated retinal cells. Moreover, although immortalized cells showed a greater propensity to survive, the cell lines demonstrated poor long-term survival. All grafts were infiltrated with host macrophage/microglial cells throughout their duration of survival. This study demonstrates that immortalized human fetal retinal progenitor cells retain their progenitor characteristics and may therefore have therapeutic potential in strategies that demand a renewable and consistent supply of donor cells for the treatment of degenerative retinal diseases.


Subject(s)
Retina/cytology , Retinal Diseases/therapy , Stem Cells/cytology , Animals , Antigens, Polyomavirus Transforming/metabolism , Biomarkers/metabolism , Cell Line, Transformed , Cell Survival , Fetus/cytology , Graft Survival , Humans , Rats , Retina/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , Temperature
19.
PLoS One ; 4(12): e8152, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19997644

ABSTRACT

Transformation of somatic cells with a set of embryonic transcription factors produces cells with the pluripotent properties of embryonic stem cells (ESCs). These induced pluripotent stem (iPS) cells have the potential to differentiate into any cell type, making them a potential source from which to produce cells as a therapeutic platform for the treatment of a wide range of diseases. In many forms of human retinal disease, including age-related macular degeneration (AMD), the underlying pathogenesis resides within the support cells of the retina, the retinal pigment epithelium (RPE). As a monolayer of cells critical to photoreceptor function and survival, the RPE is an ideally accessible target for cellular therapy. Here we report the differentiation of human iPS cells into RPE. We found that differentiated iPS-RPE cells were morphologically similar to, and expressed numerous markers of developing and mature RPE cells. iPS-RPE are capable of phagocytosing photoreceptor material, in vitro and in vivo following transplantation into the Royal College of Surgeons (RCS) dystrophic rat. Our results demonstrate that iPS cells can be differentiated into functional iPS-RPE and that transplantation of these cells can facilitate the short-term maintenance of photoreceptors through phagocytosis of photoreceptor outer segments. Long-term visual function is maintained in this model of retinal disease even though the xenografted cells are eventually lost, suggesting a secondary protective host cellular response. These findings have identified an alternative source of replacement tissue for use in human retinal cellular therapies, and provide a new in vitro cellular model system in which to study RPE diseases affecting human patients.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Retinal Diseases/therapy , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/transplantation , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Polarity , Cell Shape , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/transplantation , Humans , Immunohistochemistry , Macrophages/cytology , Phagocytosis , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/ultrastructure , Proto-Oncogene Proteins c-fos/metabolism , Rats , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/ultrastructure , Vision, Ocular/physiology
20.
Mol Vis ; 15: 283-95, 2009.
Article in English | MEDLINE | ID: mdl-19204785

ABSTRACT

PURPOSE: To examine the ability of retinal pigment epithelial (RPE) cells derived from human embryonic stem cells (HESC) to phagocytose photoreceptor outer segments, and to determine whether exposure to human retina induces any morphological changes in these cells. METHODS: HESC-RPE cells were derived from a super-confluent preparation of the Shef1 HESC line. Pigmented colonies were isolated and expanded into pigmented monolayers on Matrigel matrix-coated dishes or filters. Cells were exposed to fluorescently labeled outer segments isolated from the porcine eye and assessed for phagocytic activity at regular intervals. Expression of molecules associated with RPE phagocytosis was analyzed by RT-PCR, immunocytochemistry, and western blot. The role of Mer Tyrosine Kinase (MERTK) in the phagocytosis of outer segments was investigated using antibodies directed against MERTK to block function. In a novel approach, cells were also exposed to fresh human neural retina tissue then examined by electron microscopy for evidence of phagocytosis and changes in cell morphology. RESULTS: HESC-derived RPE cells are capable of phagocytosing isolated porcine outer segments and express molecules associated with RPE-specific phagocytosis, including MERTK. Pre-incubation with antibodies against MERTK blocked phagocytosis of photoreceptor outer segments, but not polystyrene beads. HESC-RPE cells also phagocytosed outer segments in a novel human retinal explant system. Furthermore co-culture adjacent to human retina tissue in this preparation resulted in the appearance of features in HESC-derived RPE cells normally observed only as the RPE matures. CONCLUSIONS: The ingestion of photoreceptor outer segments from an isolated population and an artificial ex vivo human retina system demonstrates HESC-derived RPE cells are functional. HESC-derived RPE possess the relevant molecules required for phagocytosis, including MERTK, which is essential for the phagocytosis of outer segments but not latex beads. Furthermore, some changes observed in cell morphology after co-culture with human retina may have implications for understanding the full development and differentiation of RPE cells.


Subject(s)
Embryonic Stem Cells/cytology , Phagocytosis/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Retinal Photoreceptor Cell Outer Segment/physiology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Analysis of Variance , Animals , Cells, Cultured , Embryonic Stem Cells/ultrastructure , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Immunohistochemistry , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Retina/physiology , Retina/ultrastructure , Retinal Pigment Epithelium/ultrastructure , Swine , c-Mer Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...