Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Vis ; 21(5): 15, 2021 05 03.
Article in English | MEDLINE | ID: mdl-34003243

ABSTRACT

Across saccades, perceptual detectability of brief visual stimuli is strongly diminished. We recently observed that this perceptual suppression phenomenon is jumpstarted in the retina, suggesting that the phenomenon might be significantly more visual in nature than normally acknowledged. Here, we explicitly compared saccadic suppression strength when saccades were made across a uniform image of constant luminance versus when saccades were made across image patches of different luminance, width, and trans-saccadic luminance polarity. We measured perceptual contrast thresholds of human subjects for brief peri-saccadic flashes of positive (luminance increments) or negative (luminance decrements) polarity. Thresholds were >6-7 times higher when saccades translated a luminance stripe or edge across the retina than when saccades were made over a completely uniform image patch. Critically, both background luminance and flash luminance polarity strongly modulated peri-saccadic contrast thresholds. In addition, all of these very same visual dependencies also occurred in the absence of any saccades, but with qualitatively similar rapid translations of image patches across the retina. This similarity of visual dependencies with and without saccades supports the notion that perceptual saccadic suppression may be fundamentally a visual phenomenon, which strongly motivates neurophysiological and theoretical investigations on the role of saccadic eye movement commands in modulating its properties.


Subject(s)
Saccades , Visual Perception , Humans , Light , Retina , Vision, Ocular
2.
J Vis ; 19(2): 1, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30707751

ABSTRACT

Spatial resolution is a key property of eyes when it comes to understanding how animals' visual signals are perceived. This property can be robustly estimated by measuring the contrast sensitivity as a function of different spatial frequencies, defined as the number of achromatic vertical bright and dark stripe pairs within one degree of visual angle. This contrast sensitivity function (CSF) has been estimated for different animal groups, but data on fish are limited to two free-swimming, freshwater species (i.e., goldfish and bluegill sunfish). In this study, we describe the CSF of a small marine cryptobenthic fish (Tripterygion delaisi) using an optokinetic reflex approach. Tripterygion delaisi features a contrast sensitivity that is as excellent as other fish species, up to 125 (reciprocal of Michelson contrast) at the optimal spatial frequency of 0.375 c/°. The maximum spatial resolution is instead relatively coarse, around 2.125 c/°. By comparing our results with acuity values derived from anatomical estimates of ganglion cells' density, we conclude that the optokinetic reflex seems to be adapted to process low spatial frequency information from stimuli in the peripheral visual field and show that small marine fish can feature excellent contrast sensitivity at optimal spatial frequency.


Subject(s)
Contrast Sensitivity/physiology , Perciformes/physiology , Visual Fields/physiology , Animals , Nystagmus, Optokinetic/physiology
3.
J Neurophysiol ; 117(5): 1894-1910, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28202573

ABSTRACT

Microsaccades occur during gaze fixation to correct for miniscule foveal motor errors. The mechanisms governing such fine oculomotor control are still not fully understood. In this study, we explored microsaccade control by analyzing the impacts of transient visual stimuli on these movements' kinematics. We found that such kinematics can be altered in systematic ways depending on the timing and spatial geometry of visual transients relative to the movement goals. In two male rhesus macaques, we presented peripheral or foveal visual transients during an otherwise stable period of fixation. Such transients resulted in well-known reductions in microsaccade frequency, and our goal was to investigate whether microsaccade kinematics would additionally be altered. We found that both microsaccade timing and amplitude were modulated by the visual transients, and in predictable manners by these transients' timing and geometry. Interestingly, modulations in the peak velocity of the same movements were not proportional to the observed amplitude modulations, suggesting a violation of the well-known "main sequence" relationship between microsaccade amplitude and peak velocity. We hypothesize that visual stimulation during movement preparation affects not only the saccadic "Go" system driving eye movements but also a "Pause" system inhibiting them. If the Pause system happens to be already turned off despite the new visual input, movement kinematics can be altered by the readout of additional visually evoked spikes in the Go system coding for the flash location. Our results demonstrate precise control over individual microscopic saccades and provide testable hypotheses for mechanisms of saccade control in general.NEW & NOTEWORTHY Microsaccadic eye movements play an important role in several aspects of visual perception and cognition. However, the mechanisms for microsaccade control are still not fully understood. We found that microsaccade kinematics can be altered in a systematic manner by visual transients, revealing a previously unappreciated and exquisite level of control by the oculomotor system of even the smallest saccades. Our results suggest precise temporal interaction between visual, motor, and inhibitory signals in microsaccade control.


Subject(s)
Evoked Potentials, Visual , Saccades , Animals , Biomechanical Phenomena , Fixation, Ocular , Macaca mulatta , Male , Models, Neurological , Visual Perception
4.
Proc Natl Acad Sci U S A ; 108(46): E1174-83, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22006315

ABSTRACT

Cilia-based locomotion is the major form of locomotion for microscopic planktonic organisms in the ocean. Given their negative buoyancy, these organisms must control ciliary activity to maintain an appropriate depth. The neuronal bases of depth regulation in ciliary swimmers are unknown. To gain insights into depth regulation we studied ciliary locomotor control in the planktonic larva of the marine annelid, Platynereis. We found several neuropeptides expressed in distinct sensory neurons that innervate locomotor cilia. Neuropeptides altered ciliary beat frequency and the rate of calcium-evoked ciliary arrests. These changes influenced larval orientation, vertical swimming, and sinking, resulting in upward or downward shifts in the steady-state vertical distribution of larvae. Our findings indicate that Platynereis larvae have depth-regulating peptidergic neurons that directly translate sensory inputs into locomotor output on effector cilia. We propose that the simple circuitry found in these ciliated larvae represents an ancestral state in nervous system evolution.


Subject(s)
Locomotion , Neuropeptides/metabolism , Polychaeta/embryology , Polychaeta/physiology , Animals , Behavior, Animal , Cilia/metabolism , Electrophysiology/methods , FMRFamide/pharmacology , Image Processing, Computer-Assisted/methods , Larva/metabolism , Larva/physiology , Molecular Sequence Data , Muscles/physiology , Neurons/metabolism , Swimming
5.
Nat Neurosci ; 11(6): 667-75, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18432197

ABSTRACT

Genetically encoded optical neuromodulators create an opportunity for circuit-specific intervention in neurological diseases. One of the diseases most amenable to this approach is retinal degeneration, where the loss of photoreceptors leads to complete blindness. To restore photosensitivity, we genetically targeted a light-activated cation channel, channelrhodopsin-2, to second-order neurons, ON bipolar cells, of degenerated retinas in vivo in the Pde6b(rd1) (also known as rd1) mouse model. In the absence of 'classical' photoreceptors, we found that ON bipolar cells that were engineered to be photosensitive induced light-evoked spiking activity in ganglion cells. The rescue of light sensitivity was selective to the ON circuits that would naturally respond to increases in brightness. Despite degeneration of the outer retina, our intervention restored transient responses and center-surround organization of ganglion cells. The resulting signals were relayed to the visual cortex and were sufficient for the animals to successfully perform optomotor behavioral tasks.


Subject(s)
Light , Retinal Bipolar Cells/physiology , Retinal Degeneration , Rhodopsin/physiology , Vision, Ocular/physiology , Animals , Behavior, Animal , Disease Models, Animal , Electroporation/methods , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Evoked Potentials, Visual/radiation effects , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Motor Activity/radiation effects , Patch-Clamp Techniques , Photic Stimulation/methods , Piperazines/pharmacology , Quinoxalines/pharmacology , Retinal Bipolar Cells/radiation effects , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinal Degeneration/therapy , Retinal Ganglion Cells/physiology , Time Factors , Vision, Ocular/radiation effects , Visual Pathways/drug effects , Visual Pathways/physiology , Visual Pathways/radiation effects
6.
Commun Biol ; 5(1): 692, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35821404

ABSTRACT

Visual perception remains stable across saccadic eye movements, despite the concurrent strongly disruptive visual flow. This stability is partially associated with a reduction in visual sensitivity, known as saccadic suppression, which already starts in the retina with reduced ganglion cell sensitivity. However, the retinal circuit mechanisms giving rise to such suppression remain unknown. Here, we describe these mechanisms using electrophysiology in mouse, pig, and macaque retina, 2-photon calcium imaging, computational modeling, and human psychophysics. We find that sequential stimuli, like those that naturally occur during saccades, trigger three independent suppressive mechanisms in the retina. The main mechanism is triggered by contrast-reversing sequential stimuli and originates within the receptive field center of ganglion cells. It does not involve inhibition or other known suppressive mechanisms like saturation or adaptation. Instead, it relies on temporal filtering of the inherently slow response of cone photoreceptors coupled with downstream nonlinearities. Two further mechanisms of suppression are present predominantly in ON ganglion cells and originate in the receptive field surround, highlighting another disparity between ON and OFF ganglion cells. The mechanisms uncovered here likely play a role in shaping the retinal output following eye movements and other natural viewing conditions where sequential stimulation is ubiquitous.


Subject(s)
Retina , Saccades , Animals , Humans , Mice , Photic Stimulation/methods , Retina/physiology , Swine , Vision, Ocular , Visual Perception/physiology
7.
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
8.
Nat Commun ; 11(1): 1977, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332756

ABSTRACT

Visual sensitivity, probed through perceptual detectability of very brief visual stimuli, is strongly impaired around the time of rapid eye movements. This robust perceptual phenomenon, called saccadic suppression, is frequently attributed to active suppressive signals that are directly derived from eye movement commands. Here we show instead that visual-only mechanisms, activated by saccade-induced image shifts, can account for all perceptual properties of saccadic suppression that we have investigated. Such mechanisms start at, but are not necessarily exclusive to, the very first stage of visual processing in the brain, the retina. Critically, neural suppression originating in the retina outlasts perceptual suppression around the time of saccades, suggesting that extra-retinal movement-related signals, rather than causing suppression, may instead act to shorten it. Our results demonstrate a far-reaching contribution of visual processing mechanisms to perceptual saccadic suppression, starting in the retina, without the need to invoke explicit motor-based suppression commands.


Subject(s)
Retina/physiology , Saccades/physiology , Visual Perception , Adult , Animals , Female , Fixation, Ocular , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Photic Stimulation , Reaction Time , Swine , Vision, Ocular , Visual Fields , Young Adult
9.
Neuron ; 46(1): 117-27, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15820698

ABSTRACT

The excitatory and inhibitory inputs to directionally selective (DS) ganglion cells are themselves directionally selective. Directionality is achieved because excitation is reduced during null-direction movement along a GABAergic pathway. Inhibition is reduced during preferred-direction movement along a pathway that includes cholinergic synapses. Both excitation and inhibition are made directional by laterally offset inhibitory signals similar to the spatial offset of the direct inhibitory input to the DS cell dendrites. Thus, spatially offset lateral inhibition generates directionality at three different levels in the DS circuitry. We also found that for stimuli falling within the dendritic field, cholinergic input is delivered to the OFF but not the ON dendrites. Cholinergic pathways from outside the dendritic field reach both ON and OFF dendrites, but both of these pathways are normally inactivated by GABAergic synapses.


Subject(s)
Neural Inhibition/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Visual Perception/physiology , Acetylcholine/metabolism , Amacrine Cells/physiology , Animals , Evoked Potentials, Visual/physiology , Glutamic Acid/metabolism , Organ Culture Techniques , Rabbits , gamma-Aminobutyric Acid/metabolism
10.
Nat Commun ; 8(1): 1813, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180667

ABSTRACT

Rod and cone photoreceptors support vision across large light intensity ranges. Rods, active under dim illumination, are thought to saturate at higher (photopic) irradiances. The extent of rod saturation is not well defined; some studies report rod activity well into the photopic range. Using electrophysiological recordings from retina and dorsal lateral geniculate nucleus of cone-deficient and visually intact mice, we describe stimulus and physiological factors that influence photopic rod-driven responses. We find that rod contrast sensitivity is initially strongly reduced at high irradiances, but progressively recovers to allow responses to moderate contrast stimuli. Surprisingly, rods recover faster at higher light levels. A model of rod phototransduction suggests that phototransduction gain adjustments and bleaching adaptation underlie rod recovery. Consistently, exogenous chromophore reduces rod responses at bright background. Thus, bleaching adaptation renders mouse rods responsive to modest contrast at any irradiance. Paradoxically, raising irradiance across the photopic range increases the robustness of rod responses.


Subject(s)
Adaptation, Physiological , Light Signal Transduction/physiology , Light/adverse effects , Photobleaching/radiation effects , Retinal Rod Photoreceptor Cells/physiology , Animals , Color Vision/physiology , Geniculate Bodies/physiology , Mice , Mice, Transgenic , Models, Animal , Photic Stimulation , Retinal Cone Photoreceptor Cells/physiology
11.
Invest Ophthalmol Vis Sci ; 57(2): 658-63, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26903226

ABSTRACT

PURPOSE: Ischemic stroke in retinal arteries leads to death of neural tissue and ultimately to blindness. The retina is known to die within 4 hours after onset of ischemia. It is debated whether hypothermia might increase the time window for medical treatment and thereby the chance of recovering sight. In order to characterize the time course of cell death during ischemia and potential beneficial effects of hypothermia in more detail, we investigated the survival of ganglion cells in ischemic pig and human retina as a function of time and temperature. METHODS: Eyes were obtained from minipigs and from human donors post mortem. Enucleated minipig eyes were stored for defined durations at three different temperatures (37 °C, 21 °C, and 4 °C). In order to assess the viability of the tissue, we measured ganglion cell activity (spiking) with multielectrode arrays. RESULTS: Minipig retinal ganglion cell function was severely compromised after 2 hours of ischemia at body temperature. After 4 hours, ganglion cells did not fire action potentials anymore. However, at 21 °C, ganglion cell activity was maintained under ischemic conditions for up to 12 hours, and for at least 50 hours at 4 °C. In postmortem human retina, we recorded ganglion cell activity in retinas received up to 27 hours after death. CONCLUSIONS: Our results demonstrate that hypothermia greatly increases survival of retinal ganglion cells exposed to ischemia. These results might be relevant for the future treatment of retinal ischemia.


Subject(s)
Hypothermia, Induced/methods , Ischemia/therapy , Retinal Diseases/therapy , Retinal Ganglion Cells/pathology , Animals , Cadaver , Cell Count , Cell Death , Cell Survival , Disease Models, Animal , Humans , Ischemia/pathology , Retinal Diseases/pathology , Swine , Swine, Miniature
12.
Invest Ophthalmol Vis Sci ; 56(8): 4835-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26218912

ABSTRACT

PURPOSE: Mutations in the OPA1 gene cause autosomal dominant optic atrophy (ADOA), a visual disorder associated with degeneration of retinal ganglion cells (RGCs). Here, we characterized the disease progression in a homologous mouse model B6;C3-Opa1 329-355del and asked whether the pronounced cell death affects certain RGC types more than others. METHODS: The influence of the Opa1 mutation was assessed by morphologic (retina and optic nerve histology) and functional (multielectrode array) methods. RESULTS: The RGC loss of approximately 50% within 18 months was significantly more pronounced in RGCs with small-caliber axons. Small-caliber axon RGCs comprise a variety of functional RGC types. Accordingly, electrophysiological analyses of RGCs did not show a dropout of distinct functional RGC subgroups. However, the response properties of RGCs were affected significantly by the mutation. Surprisingly, these functional changes were different under different luminance conditions (scotopic, mesopic, and photopic). Finally, melanopsin cells are known to be less susceptible to retinal insults. We found that these cells are also spared in the Opa1 mouse model, and demonstrated for the first time that this resistance persisted even when the melanopsin gene had been knocked-out. CONCLUSIONS: Small-caliber axons show a higher vulnerability to the Opa1 mutation in our mouse model for ADOA. Luminance-dependent functional changes suggest an influence of the Opa1 mutation on the retinal circuitry upstream of RGCs. Photoresponsive RGCs are protected against cell death due to the Opa1 mutation, but not by melanopsin expression itself.


Subject(s)
DNA/genetics , GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Retinal Ganglion Cells/pathology , Animals , DNA Mutational Analysis , Disease Models, Animal , Female , GTP Phosphohydrolases/metabolism , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Optic Atrophy, Autosomal Dominant/metabolism , Optic Atrophy, Autosomal Dominant/pathology , Retinal Ganglion Cells/metabolism
13.
J Comp Neurol ; 523(18): 2788-806, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26011242

ABSTRACT

The Jimpy mutant mouse has a point mutation in the proteolipid protein gene (plp1). The resulting misfolding of the protein leads to oligodendrocyte death, myelin destruction, and failure to produce adequately myelinated axons in the central nervous system (CNS). It is not known how the absence of normal myelination during development influences neural function. We characterized the Jimpy mouse retina to find out whether lack of myelination in the optic nerve during development has an effect on normal functioning and morphology of the retina. Optokinetic reflex measurements showed that Jimpy mice had, in general, a functional visual system. Both PLP1 antibody staining and reverse transcriptase-polymerase chain reaction for plp1 mRNA showed that plp1 is not expressed in the wild-type retina. However, in the optic nerve, plp1 is normally expressed, and consequently, in Jimpy mutant mice, myelination of axons in the optic nerve was mostly absent. Nevertheless, neither axon count nor axon ultrastructure in the optic nerve was affected. Physiological recordings of ganglion cell activity using microelectrode arrays revealed a decrease of stimulus-evoked activity at mesopic light levels. Morphological analysis of the retina did not show any significant differences in the gross morphology, such as thickness of retinal layers or cell number in the inner and outer nuclear layer. The cell bodies in the inner nuclear layer, however, were larger in the peripheral retina of Jimpy mutant mice. Antibody labeling against cell type-specific markers showed that the number of rod bipolar and horizontal cells was increased in Jimpy mice. In conclusion, whereas the Jimpy mutation has dramatic effects on the myelination of retinal ganglion cell axons, it has moderate effects on retinal morphology and function.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Mutation/genetics , Myelin Proteolipid Protein/genetics , Retina/pathology , Action Potentials , Animals , Animals, Newborn , Calbindins/metabolism , Choline O-Acetyltransferase/metabolism , Ectodysplasins/genetics , Ectodysplasins/metabolism , Glutamate-Ammonia Ligase/metabolism , Male , Mice , Mice, Jimpy , Microscopy, Electron, Transmission , Myelin Basic Protein/metabolism , Neurons/metabolism , Neurons/ultrastructure , Nystagmus, Optokinetic/genetics , Protein Kinase C , Retina/ultrastructure
14.
Nat Neurosci ; 18(1): 66-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485757

ABSTRACT

The collective activity pattern of retinal ganglion cells, the retinal code, underlies higher visual processing. How does the ambient illuminance of the visual scene influence this retinal output? We recorded from isolated mouse and pig retina and from mouse dorsal lateral geniculate nucleus in vivo at up to seven ambient light levels covering the scotopic to photopic regimes. Across each luminance transition, most ganglion cells exhibited qualitative response changes, whereas they maintained stable responses within each luminance. We commonly observed the appearance and disappearance of ON responses in OFF cells and vice versa. Such qualitative response changes occurred for a variety of stimuli, including full-field and localized contrast steps and naturalistic movies. Our results suggest that the retinal code is not fixed but varies with every change of ambient luminance. This finding raises questions about signal processing within the retina and has implications for visual processing in higher brain areas.


Subject(s)
Lighting , Retina/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/physiology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Environment , GABA Antagonists/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/physiology , Retina/drug effects , Retinal Ganglion Cells/physiology , Swine , gamma-Aminobutyric Acid/physiology
15.
Methods Mol Biol ; 1148: 149-60, 2014.
Article in English | MEDLINE | ID: mdl-24718800

ABSTRACT

Blindness is one of the most devastating conditions affecting the quality of life. Hereditary degenerative diseases, such as retinitis pigmentosa, are characterized by the progressive loss of photoreceptors, leading to complete blindness. No treatment is known, the current state-of-the-art of restoring vision are implanted electrode arrays. As a recently discovered alternative, optical neuromodulators, such as channelrhodopsin, allow new strategies for treating these diseases by imparting light-sensitivity onto the remaining retinal neurons after photoreceptor cell death. Retinal degeneration is a heterogeneous set of diseases with diverse secondary effects on the retinal circuitry. Successful treatment strategies have to take into account this diversity, as only the existing retinal hardware can serve as substrate for optogenetic intervention. The goal is to salvage the retinal ruins and to revert the leftover tissue into a functional visual sensor that operates as optimally as possible. Here, we discuss three different successful approaches that have been applied to degenerated mouse retina.


Subject(s)
Blindness/therapy , Retina/physiopathology , Animals , Dependovirus/genetics , Genetic Therapy , Humans , Optogenetics , Retina/pathology , Retinal Ganglion Cells/metabolism , Rhodopsin/biosynthesis , Rhodopsin/genetics , Transduction, Genetic
16.
PLoS One ; 9(8): e106148, 2014.
Article in English | MEDLINE | ID: mdl-25165854

ABSTRACT

Multi-electrode arrays are a state-of-the-art tool in electrophysiology, also in retina research. The output cells of the retina, the retinal ganglion cells, form a monolayer in many species and are well accessible due to their proximity to the inner retinal surface. This structure has allowed the use of multi-electrode arrays for high-throughput, parallel recordings of retinal responses to presented visual stimuli, and has led to significant new insights into retinal organization and function. However, using conventional arrays where electrodes are embedded into a glass or ceramic plate can be associated with three main problems: (1) low signal-to-noise ratio due to poor contact between electrodes and tissue, especially in the case of strongly curved retinas from small animals, e.g. rodents; (2) insufficient oxygen and nutrient supply to cells located on the bottom of the recording chamber; and (3) displacement of the tissue during recordings. Perforated multi-electrode arrays (pMEAs) have been found to alleviate all three issues in brain slice recordings. Over the last years, we have been using such perforated arrays to study light evoked activity in the retinas of various species including mouse, pig, and human. In this article, we provide detailed step-by-step instructions for the use of perforated MEAs to record visual responses from the retina, including spike recordings from retinal ganglion cells and in vitro electroretinograms (ERG). In addition, we provide in-depth technical and methodological troubleshooting information, and show example recordings of good quality as well as examples for the various problems which might be encountered. While our description is based on the specific equipment we use in our own lab, it may also prove useful when establishing retinal MEA recordings with other equipment.


Subject(s)
Retina/physiology , Animals , Electric Stimulation/instrumentation , Electric Stimulation/methods , Electrodes , Electroretinography/methods , Evoked Potentials, Visual , Humans , Mice , Swine
17.
Invest Ophthalmol Vis Sci ; 55(8): 5431-44, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25103259

ABSTRACT

PURPOSE: Age-related macular degeneration (AMD) is a major leading cause of visual impairment and blindness with no cure currently established. Cell replacement of RPE is discussed as a potential therapy for AMD. Previous studies were performed in animal models with severe limitations in recapitulating the disease progression. In detail, we describe the effect of systemic injection of sodium iodate in the mouse retina. We further evaluate the usefulness of this animal model to analyze cell-specific effects following transplantation of human embryonic stem cell (hESC)-derived RPE cells. METHODS: Morphologic, functional, and behavioral changes following sodium iodate injection were monitored by histology, gene expression analysis, electroretinography, and optokinetic head tracking. Human embryonic stem cell-derived RPE cells were transplanted 1 week after sodium iodate injection and experimental retinae were analyzed 3 weeks later. RESULTS: Injection of sodium iodate caused complete RPE cell loss, photoreceptor degeneration, and altered gene and protein expression in outer and inner nuclear layers. Retinal function was severely affected by day 3 and abolished from day 14. Following transplantation, donor hESC-derived RPE cells formed extensive monolayers that displayed wild-type RPE cell morphology, organization, and function, including phagocytosis of host photoreceptor outer segments. CONCLUSIONS: Systemic injection of sodium iodate has considerable effects on RPE, photoreceptors, and inner nuclear layer neurons, and provides a model to assay reconstitution and maturation of RPE cell transplants. The availability of an RPE-free Bruch's membrane in this model likely allows the unprecedented formation of extensive polarized cell monolayers from donor hESC-derived RPE cell suspensions.


Subject(s)
Cell Transplantation/methods , Disease Models, Animal , Retinal Diseases/therapy , Retinal Pigment Epithelium/transplantation , Animals , Iodates/pharmacology , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Retinal Diseases/chemically induced , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Pigment Epithelium/drug effects
18.
Curr Biol ; 23(20): R925-7, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24156812

ABSTRACT

Mice show an innate protective behavior to looming shadows approaching from above: they either run for cover or freeze in place. This newly discovered 'looming response' adds to the repertoire of stereotyped behaviors that can be utilized to study visual pathways.


Subject(s)
Escape Reaction , Mice/physiology , Neural Pathways/physiology , Retina/physiology , Visual Perception , Animals , Female , Male
19.
PLoS One ; 8(11): e81278, 2013.
Article in English | MEDLINE | ID: mdl-24312285

ABSTRACT

Some hereditary diseases, such as retinitis pigmentosa, lead to blindness due to the death of photoreceptors, though the rest of the visual system might be only slightly affected. Optogenetics is a promising tool for restoring vision after retinal degeneration. In optogenetics, light-sensitive ion channels ("channelrhodopsins") are expressed in neurons so that the neurons can be activated by light. Currently existing variants of channelrhodopsin--engineered for use in neurophysiological research--do not necessarily support the goal of vision restoration optimally, due to two factors: First, the nature of the light stimulus is fundamentally different in "optogenetic vision" compared to "optogenetic neuroscience". Second, the retinal target neurons have specific properties that need to be accounted for, e.g. most retinal neurons are non-spiking. In this study, by using a computational model, we investigate properties of channelrhodopsin that might improve successful vision restoration. We pay particular attention to the operational brightness range and suggest strategies that would allow optogenetic vision over a wider intensity range than currently possible, spanning the brightest 5 orders of naturally occurring luminance. We also discuss the biophysical limitations of channelrhodopsin, and of the expressing cells, that prevent further expansion of this operational range, and we suggest design strategies for optogenetic tools which might help overcoming these limitations. Furthermore, the computational model used for this study is provided as an interactive tool for the research community.


Subject(s)
Models, Biological , Optogenetics/methods , Rhodopsin/genetics , Rhodopsin/metabolism , Vision, Ocular/genetics , Environment , Light , Photic Stimulation , Vision, Ocular/radiation effects
20.
Behav Neurosci ; 127(5): 788-96, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23957722

ABSTRACT

Testing optokinetic head or eye movements is an established method to determine visual performance of laboratory animals, including chickens, guinea pigs, mice, or fish. It is based on the optokinetic reflex which causes the animals to track a drifting stripe pattern with eye and head movements. We have developed an improved version of the optomotor test with better control over the stimulus parameters, as well as a high degree of automation. The stripe pattern is presented on computer monitors surrounding the animal. By tracking the head position of freely moving animals in real time, the visual angle under which the stripes of the pattern appeared was kept constant even for changing head positions. Furthermore, an algorithm was developed for automated evaluation of the tracking performance of the animal. Comparing the automatically determined behavioral score with manual assessment of the animals' tracking behavior confirmed the reliability of our methodology. As an example, we reproduced the known contrast sensitivity function of wild type mice. Furthermore, the progressive decline in visual performance of a mouse model of retinal degeneration, rd10, was demonstrated.


Subject(s)
Eye Movements/physiology , Pattern Recognition, Automated/methods , Reflex/physiology , Visual Acuity/physiology , Algorithms , Animals , Data Collection , Female , Male , Mice , Mice, Inbred C57BL , Photic Stimulation , Video Recording
SELECTION OF CITATIONS
SEARCH DETAIL