Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 319
Filter
1.
Sci Rep ; 14(1): 11642, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773346

ABSTRACT

Vision restoration is one of the most promising applications of optogenetics. However, it is limited due to the poor-sensitivity, slow-kinetics and narrow band absorption spectra of opsins. Here, a detailed theoretical study of retinal ganglion neurons (RGNs) expressed with ChRmine, ReaChR, CoChR, CatCh and their mutants, with near monochromatic LEDs, and broadband sunlight, halogen lamp, RGB LED light, and pure white light sources has been presented. All the opsins exhibit improved light sensitivity and larger photocurrent on illuminating with broadband light sources compared to narrow band LEDs. ChRmine allows firing at ambient sunlight (1.5 nW/mm2) and pure white light (1.2 nW/mm2), which is lowest among the opsins considered. The broadband activation spectrum of ChRmine and its mutants is also useful to restore color sensitivity. Although ChRmine exhibits slower turn-off kinetics with broadband light, high-fidelity spikes can be evoked upto 50 Hz. This limit extends upto 80 Hz with the improved hsChRmine mutant although it requires double the irradiance compared to ChRmine. The present study shows that ChRmine and its mutants allow activation of RGNs with ambient light which is useful for goggle-free white light optogenetic retinal prostheses with improved quality of restored vision.


Subject(s)
Light , Optogenetics , Retinal Ganglion Cells , Optogenetics/methods , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Humans , Mutation , Animals , Opsins/genetics , Opsins/metabolism , Vision, Ocular/physiology
2.
Biochem Biophys Res Commun ; 592: 93-98, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35033872

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) are able to synthesize the photosensitive protein melanopsin, which is involved in the regulation of circadian rhythms, the papillary light reflex and other nonimaging visual functions. To investigate whether ipRGCs are involved in mediating the light modulation of sleep-wakefulness in rodents, melanopsin knockout mice (MKO), melanopsin-only mice (MO) and coneless, rodless, melanopsin knockout mice (TKO) were used in this study to record electroencephalogram and electromyography variations in the normal 12:12 h light:dark cycle, and 1 h and 3 h light pulses were administered at 1 h after the light was turned off. In the normal 12:12 h light-dark cycle, the WT, MKO and MO mice had a regular day-night rhythm and no significant difference in wakefulness, rapid eye movement (REM) or nonrapid eye movement (NREM) sleep. However, TKO mice could not be entrained according to the light-dark cycle and exhibited a free-running rhythm. Extending the light pulse durations significantly changed the sleep and wakefulness activities of the WT and MO mice but did not have an effect on the MKO mice. These results indicate that melanopsin significantly affects REM and NREM sleep and that ipRGCs play an important role in light-induced sleep in mice.


Subject(s)
Light , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Sleep/physiology , Sleep/radiation effects , Wakefulness/physiology , Wakefulness/radiation effects , Animals , Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Male , Mice, Inbred C57BL , Mice, Knockout , Rod Opsins/deficiency , Rod Opsins/metabolism , Sleep Stages/physiology , Sleep Stages/radiation effects
3.
Nat Commun ; 12(1): 5115, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433830

ABSTRACT

Light regulates daily sleep rhythms by a neural circuit that connects intrinsically photosensitive retinal ganglion cells (ipRGCs) to the circadian pacemaker, the suprachiasmatic nucleus. Light, however, also acutely affects sleep in a circadian-independent manner. The neural circuits involving the acute effect of light on sleep remain unknown. Here we uncovered a neural circuit that drives this acute light response, independent of the suprachiasmatic nucleus, but still through ipRGCs. We show that ipRGCs substantially innervate the preoptic area (POA) to mediate the acute light effect on sleep in mice. Consistently, activation of either the POA projecting ipRGCs or the light-responsive POA neurons increased non-rapid eye movement (NREM) sleep without influencing REM sleep. In addition, inhibition of the light-responsive POA neurons blocked the acute light effects on NREM sleep. The predominant light-responsive POA neurons that receive ipRGC input belong to the corticotropin-releasing hormone subpopulation. Remarkably, the light-responsive POA neurons are inhibitory and project to well-known wakefulness-promoting brain regions, such as the tuberomammillary nucleus and the lateral hypothalamus. Therefore, activation of the ipRGC-POA circuit inhibits arousal brain regions to drive light-induced NREM sleep. Our findings reveal a functional retina-brain circuit that is both necessary and sufficient for the acute effect of light on sleep.


Subject(s)
Neuronal Plasticity/radiation effects , Retinal Ganglion Cells/radiation effects , Sleep/radiation effects , Suprachiasmatic Nucleus/physiology , Animals , Light , Male , Mice , Photoreceptor Cells/radiation effects , Preoptic Area/physiology , Preoptic Area/radiation effects , Suprachiasmatic Nucleus/radiation effects , Wakefulness/radiation effects
4.
Int J Mol Sci ; 22(16)2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34445085

ABSTRACT

Retinal ganglion cells (RGCs) undergo dendritic pruning in a variety of neurodegenerative diseases, including glaucoma and autosomal dominant optic atrophy (ADOA). Axotomising RGCs by severing the optic nerve generates an acute model of RGC dendropathy, which can be utilized to assess the therapeutic potential of treatments for RGC degeneration. Photobiomodulation (PBM) with red light provided neuroprotection to RGCs when administered ex vivo to wild-type retinal explants. In the current study, we used aged (13-15-month-old) wild-type and heterozygous B6;C3-Opa1Q285STOP (Opa1+/-) mice, a model of ADOA exhibiting RGC dendropathy. These mice were pre-treated with 4 J/cm2 of 670 nm light for five consecutive days before the eyes were enucleated and the retinas flat-mounted into explant cultures for 0-, 8- or 16-h ex vivo. RGCs were imaged by confocal microscopy, and their dendritic architecture was quantified by Sholl analysis. In vivo 670 nm light pretreatment inhibited the RGC dendropathy observed in untreated wild-type retinas over 16 h ex vivo and inhibited dendropathy in ON-center RGCs in wild-type but not Opa1+/- retinas. Immunohistochemistry revealed that aged Opa1+/- RGCs exhibited increased nitrosative damage alongside significantly lower activation of NF-κB and upregulation of DJ-1. PBM restored NF-κB activation in Opa1+/- RGCs and enhanced DJ-1 expression in both genotypes, indicating a potential molecular mechanism priming the retina to resist future oxidative insult. These data support the potential of PBM as a treatment for diseases involving RGC degeneration.


Subject(s)
Optic Atrophy, Autosomal Dominant/therapy , Phototherapy , Protein Deglycase DJ-1/analysis , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/radiation effects , Animals , Disease Models, Animal , Light , Mice , Neuroprotection/radiation effects , Optic Atrophy, Autosomal Dominant/pathology , Retinal Degeneration , Retinal Ganglion Cells/cytology , Up-Regulation/radiation effects
5.
PLoS One ; 16(2): e0246952, 2021.
Article in English | MEDLINE | ID: mdl-33592045

ABSTRACT

The retinal output is the sole source of visual information for the brain. Studies in non-primate mammals estimate that this information is carried by several dozens of retinal ganglion cell types, each informing the brain about different aspects of a visual scene. Even though morphological studies of primate retina suggest a similar diversity of ganglion cell types, research has focused on the function of only a few cell types. In human retina, recordings from individual cells are anecdotal or focus on a small subset of identified types. Here, we present the first systematic ex-vivo recording of light responses from 342 ganglion cells in human retinas obtained from donors. We find a great variety in the human retinal output in terms of preferences for positive or negative contrast, spatio-temporal frequency encoding, contrast sensitivity, and speed tuning. Some human ganglion cells showed similar response behavior as known cell types in other primate retinas, while we also recorded light responses that have not been described previously. This first extensive description of the human retinal output should facilitate interpretation of primate data and comparison to other mammalian species, and it lays the basis for the use of ex-vivo human retina for in-vitro analysis of novel treatment approaches.


Subject(s)
Retinal Ganglion Cells/physiology , Animals , Contrast Sensitivity/radiation effects , Humans , Light , Photic Stimulation , Retinal Ganglion Cells/radiation effects
6.
J Biol Rhythms ; 36(2): 146-159, 2021 04.
Article in English | MEDLINE | ID: mdl-33357136

ABSTRACT

"Non-image-forming" (NIF) effects of light are mediated primarily by a subset of intrinsically photosensitive retinal ganglion cells (ipRGCs) expressing the photopigment, melanopsin (OPN4). These NIF functions include circadian entrainment, pupillary reflexes, and photic effects on sleep, mood, and cognition. We recently reported that mice of multiple genotypes exhibit reduced voluntary ethanol intake under both constant darkness (DD) and constant light (LL) relative to standard light-dark (LD) conditions. In the present study, we sought to determine whether these effects are mediated by melanopsin-expressing ipRGCs and their potential relationship to photic effects on the circadian system. To this end, we examined the effects of environmental lighting regimen on both ethanol intake and circadian activity rhythms in a genetically engineered mouse model (Opn4aDTA/aDTA) in which melanopsin expression is completely blocked while ipRGCs are progressively ablated due to activation of attenuated diphtheria toxin A (aDTA) transgene under the control of the Opn4 promoter. As expected from previous studies, Opn4aDTA/aDTA mice displayed dramatic attenuation of circadian photosensitivity, but surprisingly, showed identical suppression of ethanol intake under both DD and LL as that seen in controls. These results demonstrate that the effects of lighting regimen on voluntary ethanol intake are independent of melanopsin-expressing ipRGCs and ipRGC-mediated photic effects on the circadian system. Rather, these effects are likely mediated by classical retinal photoreceptors and central pathways.


Subject(s)
Circadian Rhythm/radiation effects , Ethanol/administration & dosage , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Rod Opsins/metabolism , Animals , Female , Lighting , Male , Mice , Rod Opsins/deficiency , Rod Opsins/genetics
7.
Invest Ophthalmol Vis Sci ; 61(12): 15, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33064129

ABSTRACT

Purpose: Functional adaptation to ambient light is a key characteristic of retinal ganglion cells (RGCs), but little is known about how adaptation is affected by factors that are harmful to RGC health. We explored adaptation-induced changes to RGC physiology when exposed to increased intraocular pressure (IOP), a major risk factor for glaucoma. Methods: Wild-type mice of both sexes were subjected to 2 weeks of IOP elevation using the bead model. Retinas were assessed using a multielectrode array to record RGC responses to checkerboard white noise stimulation under both scotopic and photopic light levels. This information was used to calculate a spike-triggered average (STA) for each RGC with which to compare between lighting levels. Results: Low but not high IOP elevation resulted in several distinct RGC functional changes: (1) diminished adaptation-dependent receptive field (RF) center-surround interactions; (2) increased likelihood of a scotopic STA; and (3) increased spontaneous firing rate. Center RF size change with lighting level varied among RGCs, and both the center and surround STA peak times were consistently increased under scotopic illumination, although none of these properties were impacted by IOP level. Conclusions: These findings provide novel evidence that RGCs exhibit reduced light-dependent adaptation and increased excitability when IOP is elevated to low but not high levels. These results may reveal functional changes that occur early in glaucoma, which can potentially be used to identify patients with glaucoma at earlier stages when intervention is most beneficial.


Subject(s)
Adaptation, Ocular/physiology , Intraocular Pressure/physiology , Light , Ocular Hypertension/physiopathology , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/radiation effects , Animals , Color Vision/physiology , Contrast Sensitivity/physiology , Disease Models, Animal , Electrophysiology , Female , Male , Mice , Mice, Inbred C57BL , Night Vision/physiology , Photic Stimulation
8.
Invest Ophthalmol Vis Sci ; 61(12): 10, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33049058

ABSTRACT

Purpose: Intrinsically photosensitive retinal ganglion cells (ipRGCs) that express the visual pigment melanopsin regulate non-image-forming visual tasks, such as circadian photoentrainment and pupil constriction, as well as contrast detection for image formation. Sustained ipRGC function throughout the day is, therefore, of great importance. Melanopsin is a bistable rhabdomeric-type (R-type) visual pigment, which is thought to use light to regenerate its chromophore from all-trans-retinal back to 11-cis-retinal and does not depend on constant chromophore supply to the extent required by visual pigment in rod and cone photoreceptors. Like the majority of photopigments and G-protein-coupled receptors (GPCRs), melanopsin deactivation requires C-terminal phosphorylation and subsequent ß-arrestin binding. We hypothesize that melanopsin utilizes canonical GPCR resensitization mechanisms, including dephosphorylation and endocytosis, during the light, and together, they provide a mechanism for prolonged light responses. Methods: Here, we examined expression of protein phosphatases from a variety of subfamilies by RT-PCR and immunohistochemical analyses of the mouse retina. The expression of protein phosphatase 2A (PP2A) in ipRGCs was assessed. We also examine the role of phosphatase and endocytic activity in sustaining melanopsin signaling using transiently-transfected HEK293 cells. Results: Our analyses suggest that melanopsin-mediated light responses can be rapidly and extensively enhanced by PP2A activity. Light-activated melanopsin undergoes endocytosis in a clathrin-dependent manner. This endocytic activity enhances light responses upon repeated stimulation, implicating a role for endocytic activity in resensitization. Conclusions: Thus, we propose that melanopsin phototransduction is maintained by utilizing canonical GPCR resensitization mechanisms rather than reliance on chromophore replenishment from supporting cells.


Subject(s)
Clathrin/physiology , Endocytosis/physiology , Gene Expression Regulation/physiology , Protein Phosphatase 2/physiology , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Animals , Calcium Signaling/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Photic Stimulation , Plasmids , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/radiation effects , Transfection , Vision, Ocular/physiology
9.
Nat Commun ; 11(1): 4605, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929073

ABSTRACT

From starlight to sunlight, adaptation alters retinal output, changing both the signal and noise among populations of retinal ganglion cells (RGCs). Here we determine how these light level-dependent changes impact decoding of retinal output, testing the importance of accounting for RGC noise correlations to optimally read out retinal activity. We find that at moonlight conditions, correlated noise is greater and assuming independent noise severely diminishes decoding performance. In fact, assuming independence among a local population of RGCs produces worse decoding than using a single RGC, demonstrating a failure of population codes when correlated noise is substantial and ignored. We generalize these results with a simple model to determine what conditions dictate this failure of population processing. This work elucidates the circumstances in which accounting for noise correlations is necessary to take advantage of population-level codes and shows that sensory adaptation can strongly impact decoding requirements on downstream brain areas.


Subject(s)
Retina/physiology , Adaptation, Ocular/radiation effects , Animals , Bayes Theorem , Light , Linear Models , Night Vision/physiology , Photic Stimulation , Rats, Long-Evans , Retina/radiation effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects
10.
Mol Neurobiol ; 57(11): 4530-4548, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32748371

ABSTRACT

Aquaporin 9 (AQP9) is an aquaglyceroporin that can transport lactate. Accumulating evidence suggests that astrocyte-to-neuron lactate shuttle (ANLS) plays a critical role in energy metabolism in neurons, including retinal ganglion cells (RGCs). To test the hypothesis that AQP9, in concert with monocarboxylate transporters (MCTs), participates in ANLS to maintain function and survival of RGCs, Aqp9-null mice and wild-type (WT) littermates were subjected to optic nerve crush (ONC) with or without intravitreal injection of an MCT2 inhibitor. RGC density was similar between the Aqp9-null mice and WT mice without ONC, while ONC resulted in significantly more RGC density reduction in the Aqp9-null mice than in the WT mice at day 7. Positive scotopic threshold response (pSTR) amplitude values were similar between the two groups without ONC, but were significantly more reduced in the Aqp9-null mice than in the WT mice 7days after ONC. MCT2 inhibitor injection accelerated RGC death and pSTR amplitude reduction only in the WT mice with ONC. Immunolabeling revealed that both RGCs and astrocytes expressed AQP9, that ONC predominantly reduced astrocytic AQP9 expression, and that MCTs 1, 2, and 4 were co-localized with AQP9 at the ganglion cell layer. These retinal MCTs were also co-immunoprecipitated with AQP9 in the WT mice. ONC decreased the co-immunoprecipitation of MCTs 1 and 4, but did not impact co-immunoprecipitation of MCT2. Retinal glucose transporter 1 expression was increased in Aqp9-null mice. Aqp9 gene deletion reduced and increased the intraretinal L-lactate and D-glucose concentrations, respectively. Results suggest that AQP9 acts as the ANLS to maintain function and survival of RGCs.


Subject(s)
Aquaporins/genetics , Astrocytes/metabolism , Gene Deletion , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Neurons/metabolism , Optic Nerve/pathology , Retinal Ganglion Cells/pathology , Animals , Aquaporins/metabolism , Aquaporins/radiation effects , Astrocytes/radiation effects , Biological Transport/radiation effects , Cell Death , Cell Survival/radiation effects , Dark Adaptation/radiation effects , Electroretinography , Energy Metabolism/radiation effects , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/metabolism , Light , Mice, Inbred C57BL , Mice, Knockout , Monocarboxylic Acid Transporters/antagonists & inhibitors , Nerve Crush , Neurons/radiation effects , Night Vision/radiation effects , Optic Nerve/physiopathology , Optic Nerve/radiation effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Sensory Thresholds/radiation effects
11.
Invest Ophthalmol Vis Sci ; 61(10): 40, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32832970

ABSTRACT

Purpose: To determine the effects of narrowband light exposure on choroidal thickness and the pupil response in humans. Methods: Twenty subjects, ages 21 to 43 years, underwent 1 hour of exposure to broadband, short wavelength "blue," or long wavelength "red" light, or darkness. Choroidal thickness, imaged with spectral domain optical coherence tomography, axial length, determined from biometry, and rod/cone- and intrinsically photosensitive retinal ganglion cell-driven pupil responses were measured before and after exposure. Pupil stimuli were six 1 second alternating red (651 nm) and blue (456 nm) stimuli, 60 seconds apart. Pupil metrics included maximum constriction and the 6 second post-illumination pupil response (PIPR). Results: Compared with before exposure, the choroid significantly thinned after broadband light, red light, and dark exposure (all P < 0.05), but not after blue light exposure (P = 0.39). The maximum constriction to 1 second red stimuli significantly decreased after all light exposures (all P < 0.001), but increased after dark exposure (P = 0.02), compared with before exposure. Maximum constriction and 6-second PIPR to 1 second blue stimuli significantly decreased after all light exposures compared with before exposure (all P < 0.005), with no change after dark exposure (P > 0.05). There were no differences in axial length change or 6-second PIPR to red stimuli between exposures. Conclusions: Narrowband blue and red light exposure induced differential changes in choroidal thickness. Maximum constriction, a function of rod/cone activity, and the intrinsically photosensitive retinal ganglion cell-mediated PIPR were attenuated after all light exposures. Findings demonstrate differing effects of short-term narrowband light and dark exposure on the choroid, rod/cone activity, and intrinsically photosensitive retinal ganglion cells.


Subject(s)
Choroid/radiation effects , Reflex, Pupillary/radiation effects , Adult , Choroid/anatomy & histology , Choroid/diagnostic imaging , Female , Humans , Light , Male , Photic Stimulation , Pupil/radiation effects , Retinal Ganglion Cells/radiation effects , Tomography, Optical Coherence , Young Adult
12.
Nat Neurosci ; 23(7): 869-880, 2020 07.
Article in English | MEDLINE | ID: mdl-32483349

ABSTRACT

Besides generating vision, light modulates various physiological functions, including mood. While light therapy applied in the daytime is known to have anti-depressive properties, excessive light exposure at night has been reportedly associated with depressive symptoms. The neural mechanisms underlying this day-night difference in the effects of light are unknown. Using a light-at-night (LAN) paradigm in mice, we showed that LAN induced depressive-like behaviors without disturbing the circadian rhythm. This effect was mediated by a neural pathway from retinal melanopsin-expressing ganglion cells to the dorsal perihabenular nucleus (dpHb) to the nucleus accumbens (NAc). Importantly, the dpHb was gated by the circadian rhythm, being more excitable at night than during the day. This indicates that the ipRGC→dpHb→NAc pathway preferentially conducts light signals at night, thereby mediating LAN-induced depressive-like behaviors. These findings may be relevant when considering the mental health effects of the prevalent nighttime illumination in the industrial world.


Subject(s)
Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Depression/physiopathology , Light/adverse effects , Visual Pathways/physiology , Animals , Depression/etiology , Habenula/physiology , Habenula/radiation effects , Mice , Nucleus Accumbens/physiology , Nucleus Accumbens/radiation effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Visual Pathways/radiation effects
13.
Science ; 368(6495): 1108-1113, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32499439

ABSTRACT

Enabling near-infrared light sensitivity in a blind human retina may supplement or restore visual function in patients with regional retinal degeneration. We induced near-infrared light sensitivity using gold nanorods bound to temperature-sensitive engineered transient receptor potential (TRP) channels. We expressed mammalian or snake TRP channels in light-insensitive retinal cones in a mouse model of retinal degeneration. Near-infrared stimulation increased activity in cones, ganglion cell layer neurons, and cortical neurons, and enabled mice to perform a learned light-driven behavior. We tuned responses to different wavelengths, by using nanorods of different lengths, and to different radiant powers, by using engineered channels with different temperature thresholds. We targeted TRP channels to human retinas, which allowed the postmortem activation of different cell types by near-infrared light.


Subject(s)
Blindness/therapy , Gold , Infrared Rays , Nanotubes , Retinal Degeneration/therapy , Sensory Thresholds/radiation effects , TRPC Cation Channels/physiology , Vision, Ocular/radiation effects , Animals , Blindness/physiopathology , Disease Models, Animal , Evoked Potentials, Visual/physiology , Evoked Potentials, Visual/radiation effects , Genetic Engineering , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Photic Stimulation , Rats , Retinal Cone Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Sensory Thresholds/physiology , Snakes , TRPC Cation Channels/genetics , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Vision, Ocular/physiology , Visual Cortex/physiopathology , Visual Cortex/radiation effects
14.
Science ; 368(6490): 527-531, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32355031

ABSTRACT

Retinal ganglion cells (RGCs) drive diverse, light-evoked behaviors that range from conscious visual perception to subconscious, non-image-forming behaviors. It is thought that RGCs primarily drive these functions through the release of the excitatory neurotransmitter glutamate. We identified a subset of melanopsin-expressing intrinsically photosensitive RGCs (ipRGCs) in mice that release the inhibitory neurotransmitter γ-aminobutyric acid (GABA) at non-image-forming brain targets. GABA release from ipRGCs dampened the sensitivity of both the pupillary light reflex and circadian photoentrainment, thereby shifting the dynamic range of these behaviors to higher light levels. Our results identify an inhibitory RGC population in the retina and provide a circuit-level mechanism that contributes to the relative insensitivity of non-image-forming behaviors at low light levels.


Subject(s)
Neural Pathways/physiology , Retinal Ganglion Cells/physiology , Visual Perception/physiology , Animals , Female , GABAergic Neurons/physiology , Glutamate Decarboxylase/metabolism , Light , Male , Mice , Mice, Mutant Strains , Neural Pathways/drug effects , Reflex, Pupillary/physiology , Reflex, Pupillary/radiation effects , Retinal Ganglion Cells/radiation effects , Rod Opsins/metabolism , Unconscious, Psychology , Visual Perception/radiation effects , gamma-Aminobutyric Acid/metabolism
15.
Nature ; 581(7807): 194-198, 2020 05.
Article in English | MEDLINE | ID: mdl-32404998

ABSTRACT

Daily changes in light and food availability are major time cues that influence circadian timing1. However, little is known about the circuits that integrate these time cues to drive a coherent circadian output1-3. Here we investigate whether retinal inputs modulate entrainment to nonphotic cues such as time-restricted feeding. Photic information is relayed to the suprachiasmatic nucleus (SCN)-the central circadian pacemaker-and the intergeniculate leaflet (IGL) through intrinsically photosensitive retinal ganglion cells (ipRGCs)4. We show that adult mice that lack ipRGCs from the early postnatal stages have impaired entrainment to time-restricted feeding, whereas ablation of ipRGCs at later stages had no effect. Innervation of ipRGCs at early postnatal stages influences IGL neurons that express neuropeptide Y (NPY) (hereafter, IGLNPY neurons), guiding the assembly of a functional IGLNPY-SCN circuit. Moreover, silencing IGLNPY neurons in adult mice mimicked the deficits that were induced by ablation of ipRGCs in the early postnatal stages, and acute inhibition of IGLNPY terminals in the SCN decreased food-anticipatory activity. Thus, innervation of ipRGCs in the early postnatal period tunes the IGLNPY-SCN circuit to allow entrainment to time-restricted feeding.


Subject(s)
Circadian Rhythm/physiology , Feeding Behavior/physiology , Light , Neural Pathways , Retina/physiology , Animals , Axons/physiology , Axons/radiation effects , Circadian Rhythm/radiation effects , Cues , Eating/physiology , Eating/radiation effects , Feeding Behavior/radiation effects , Female , Geniculate Bodies/cytology , Geniculate Bodies/physiology , Geniculate Bodies/radiation effects , Male , Mice , Neural Pathways/radiation effects , Neuropeptide Y/metabolism , Retina/cytology , Retina/radiation effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Signal Transduction/radiation effects , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/radiation effects , Time Factors
16.
Middle East Afr J Ophthalmol ; 27(3): 177-181, 2020.
Article in English | MEDLINE | ID: mdl-33488015

ABSTRACT

PURPOSE: In response to growing concern about the effect of blue light on ocular tissue, companies have created mobile device screen protectors to block blue light. This project evaluates one of these screen protectors' ability to reduce blue light intensity. METHODS: The intensity of light at 450 nm from an iPhone 8, iPhone X, and iPad was measured in a dark room. The averages of three measurements were taken with and without the screen protector at different distances, settings of brightness, and Apple's night shift (NS) mode. Results were analyzed using paired t-tests. RESULTS: At 33 cm, 100% brightness, and 0% NS, the screen protector decreased intensity by 43.9%, 32.3%, and 34.9% for the iPhone 8, iPhone X, and iPad, respectively. At 33 cm and 100% brightness, increasing NS mode from 0% to 100% decreased intensity by 81.2%, 84.2%, and 86.5%. At 33 cm without NS, decreasing the brightness from 100% to 0% decreased intensity by 99.5%, 99.8%, and 97.8%. CONCLUSIONS: The screen protector decreased the intensity at 450 nm for every setting other than those at 0% brightness. Decreasing brightness and applying NS mode were more effective in reducing blue light. More research is needed to determine the benefits of decreasing blue light exposure from electronic devices.


Subject(s)
Computers, Handheld , Light/adverse effects , Macular Degeneration/prevention & control , Radiation Injuries, Experimental/prevention & control , Radiation Protection/instrumentation , Smartphone , Animals , Circadian Rhythm/radiation effects , Humans , Macular Degeneration/etiology , Radiation Dosage , Radiation Injuries, Experimental/etiology , Retinal Ganglion Cells/radiation effects , Retinal Pigment Epithelium/radiation effects
17.
Mol Neurobiol ; 57(1): 435-449, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31376070

ABSTRACT

Enriched environment (EE) protects the retina from adult rats against ischemia/reperfusion (I/R) injury; however, how the components of EE contribute to the recovery after retinal ischemic damage remains unclear. We analyzed the contribution of social, cognitive, and visual stimulation on functional and histological alterations induced by I/R. Male Wistar rats were submitted to unilateral ischemia by increasing intraocular pressure to 120 mmHg for 40 min. After ischemia, animals were housed in the following conditions: standard environment (SE), enriched environment (EE), novelty environment (NE), standard social environment (SoE), standard visual environment (SVE), or visual environment (VE). In another set of experiments, rats were submitted to bilateral ischemia and housed in SE or EE. At 2 weeks post-ischemia, rats were subjected to electroretinography and histological analysis. EE (but not SoE or NE) afforded functional and histological protection against unilateral ischemia. EE did not induce protection in animals submitted to bilateral ischemia. VE protected retinal function and histology and increased retinal BDNF levels, while a TrkB receptor antagonist prevented the protective effect of VE against I/R damage. In animals submitted to unilateral ischemia, EE and VE induced an increase in c-fos immunoreactivity in the ipsi and contralateral superior colliculus, whereas in animals submitted to bilateral ischemia, no changes in c-fos-immunoreactivity were observed in either superior colliculus from EE-housed animals. These results support that visual stimulation could be a potent stimulus for driving retinal protection in adult rats through a BDNF/TrkB-dependent mechanism, likely involving the superior colliculus.


Subject(s)
Ischemia/pathology , Photic Stimulation , Retina/pathology , Retina/radiation effects , Animals , Azepines/pharmacology , Benzamides/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Electroretinography , Locomotion/drug effects , Male , Rats, Wistar , Retina/drug effects , Retina/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/radiation effects
18.
J Drug Target ; 28(3): 330-338, 2020 03.
Article in English | MEDLINE | ID: mdl-31479288

ABSTRACT

Direct exposure to Ultraviolet (UV) radiation causes progressive damages in retinal cells, which is one of the hypothetical mechanisms underlying age-related retinopathy or macular degeneration. The protective effects of Apocynin against UV damages were firstly tested in retinal pigment epithelium cells (RPEs) and retinal ganglion cells (RGCs). Subsequently the beneficial effect of Apocynin on mouse retinas against light damage were examined. Next, microarray profiling was used to identify the genes regulated by Apocynin in both RPEs and RGCs. A candidate gene was isolated for functional characterisation by knock-down study. Apocynin was shown to inhibit cell death, reduce oxidative stress and deoxyribonucleic acid damages in both RPEs and RGCs challenged with UV. Intravitreal application of Apocynin also improved retinal dysfunction caused by light damage. Sirtuin 1 (SIRT1) was identified as induced by Apocynin by microarray study. The induction was confirmed by realtime-PCR and western blotting. Knocking down SIRT1 antagonised the protective effect of Apocynin against UV damages in both RPEs and RGCs. Apocynin is a novel agent that shows both in vitro and in vivo efficacies against UV radiation induced retina damages. SIRT1 pathway is implicated in UV radiation protection of Apocynin in retinal cells.


Subject(s)
Acetophenones/pharmacology , Retinal Ganglion Cells/drug effects , Retinal Pigment Epithelium/drug effects , Sirtuin 1/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Retinal Ganglion Cells/radiation effects , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/radiation effects , Sirtuin 1/genetics , Ultraviolet Rays/adverse effects
19.
Sci Rep ; 9(1): 18396, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804570

ABSTRACT

Rapid dilation of retinal vessels in response to flickering light (functional hyperemia) is a well-known autoregulatory response driven by increased neural activity in the inner retina. Little is known about flicker-induced changes of activity of retinal neurons themselves. We non-invasively investigated flicker-induced changes of retinal ganglion cell (RGC) function in common inbred mouse strains using the pattern electroretinogram (PERG), a sensitive measure of RGC function. Flicker was superimposed on the pattern stimulus at frequencies that did not generate measurable flicker-ERG and alter the PERG response. Transition from flicker at 101 Hz (control) to flicker at 11 Hz (test) at constant mean luminance induced a slow reduction of PERG amplitude to a minimum (39% loss in C57BL/6J mice and 52% loss in DBA/2J mice) 4-5 minutes after 11 Hz flicker onset, followed by a slow recovery to baseline over 20 minutes. Results demonstrate that the magnitude and temporal dynamics of RGC response induced by flicker at 11 Hz can be non-invasively assessed with PERG in the mouse. This allows investigating the functional phenotype of different mouse strains as well as pathological changes in glaucoma and optic nerve disease. The non-contact flicker-PERG method opens the possibility of combined assessment of neural and vascular response dynamics.


Subject(s)
Adaptation, Physiological , Hyperemia/physiopathology , Retinal Ganglion Cells/radiation effects , Retinal Vessels/radiation effects , Animals , Electroretinography/methods , Hyperemia/etiology , Light , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Photic Stimulation/methods , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Retinal Vessels/physiopathology
20.
Sci Rep ; 9(1): 15110, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641196

ABSTRACT

In the visual system, retinal ganglion cells (RGCs) of various subtypes encode preprocessed photoreceptor signals into a spike output which is then transmitted towards the brain through parallel feature pathways. Spike timing determines how each feature signal contributes to the output of downstream neurons in visual brain centers, thereby influencing efficiency in visual perception. In this study, we demonstrate a marked population-wide variability in RGC response latency that is independent of trial-to-trial variability and recording approach. RGC response latencies to simple visual stimuli vary considerably in a heterogenous cell population but remain reliable when RGCs of a single subtype are compared. This subtype specificity, however, vanishes when the retinal circuitry is bypassed via direct RGC electrical stimulation. This suggests that latency is primarily determined by the signaling speed through retinal pathways that provide subtype specific inputs to RGCs. In addition, response latency is significantly altered when GABA inhibition or gap junction signaling is disturbed, which further supports the key role of retinal microcircuits in latency tuning. Finally, modulation of stimulus parameters affects individual RGC response delays considerably. Based on these findings, we hypothesize that retinal microcircuits fine-tune RGC response latency, which in turn determines the context-dependent weighing of each signal and its contribution to visual perception.


Subject(s)
Reaction Time/physiology , Retina/physiology , Signal Transduction , Animals , Calcium Signaling/radiation effects , Gap Junctions/radiation effects , Light , Mice, Inbred C57BL , Neural Inhibition/radiation effects , Photic Stimulation , Reaction Time/radiation effects , Retina/radiation effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Signal Transduction/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...