Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 434
Filter
1.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38719453

ABSTRACT

Retinal prosthetics are one of the leading therapeutic strategies to restore lost vision in patients with retinitis pigmentosa and age-related macular degeneration. Much work has described patterns of spiking in retinal ganglion cells (RGCs) in response to electrical stimulation, but less work has examined the underlying retinal circuitry that is activated by electrical stimulation to drive these responses. Surprisingly, little is known about the role of inhibition in generating electrical responses or how inhibition might be altered during degeneration. Using whole-cell voltage-clamp recordings during subretinal electrical stimulation in the rd10 and wild-type (wt) retina, we found electrically evoked synaptic inputs differed between ON and OFF RGC populations, with ON cells receiving mostly excitation and OFF cells receiving mostly inhibition and very little excitation. We found that the inhibition of OFF bipolar cells limits excitation in OFF RGCs, and a majority of both pre- and postsynaptic inhibition in the OFF pathway arises from glycinergic amacrine cells, and the stimulation of the ON pathway contributes to inhibitory inputs to the RGC. We also show that this presynaptic inhibition in the OFF pathway is greater in the rd10 retina, compared with that in the wt retina.


Subject(s)
Electric Stimulation , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/physiology , Retinal Degeneration/physiopathology , Mice, Inbred C57BL , Retinal Bipolar Cells/physiology , Patch-Clamp Techniques , Visual Pathways/physiology , Visual Pathways/physiopathology , Neural Inhibition/physiology , Female , Male , Retina/physiology , Amacrine Cells/physiology
2.
Nat Commun ; 15(1): 2965, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580652

ABSTRACT

VGluT3-expressing mouse retinal amacrine cells (VG3s) respond to small-object motion and connect to multiple types of bipolar cells (inputs) and retinal ganglion cells (RGCs, outputs). Because these input and output connections are intermixed on the same dendrites, making sense of VG3 circuitry requires comparing the distribution of synapses across their arbors to the subcellular flow of signals. Here, we combine subcellular calcium imaging and electron microscopic connectomic reconstruction to analyze how VG3s integrate and transmit visual information. VG3s receive inputs from all nearby bipolar cell types but exhibit a strong preference for the fast type 3a bipolar cells. By comparing input distributions to VG3 dendrite responses, we show that VG3 dendrites have a short functional length constant that likely depends on inhibitory shunting. This model predicts that RGCs that extend dendrites into the middle layers of the inner plexiform encounter VG3 dendrites whose responses vary according to the local bipolar cell response type.


Subject(s)
Amacrine Cells , Retina , Mice , Animals , Amacrine Cells/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Synapses/metabolism , Microscopy, Electron , Dendrites/physiology
3.
Neural Comput ; 36(6): 1041-1083, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38669693

ABSTRACT

We consider a model of basic inner retinal connectivity where bipolar and amacrine cells interconnect and both cell types project onto ganglion cells, modulating their response output to the brain visual areas. We derive an analytical formula for the spatiotemporal response of retinal ganglion cells to stimuli, taking into account the effects of amacrine cells inhibition. This analysis reveals two important functional parameters of the network: (1) the intensity of the interactions between bipolar and amacrine cells and (2) the characteristic timescale of these responses. Both parameters have a profound combined impact on the spatiotemporal features of retinal ganglion cells' responses to light. The validity of the model is confirmed by faithfully reproducing pharmacogenetic experimental results obtained by stimulating excitatory DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) expressed on ganglion cells and amacrine cells' subclasses, thereby modifying the inner retinal network activity to visual stimuli in a complex, entangled manner. Our mathematical model allows us to explore and decipher these complex effects in a manner that would not be feasible experimentally and provides novel insights in retinal dynamics.


Subject(s)
Retina , Retinal Ganglion Cells , Retinal Ganglion Cells/physiology , Retina/physiology , Animals , Models, Neurological , Amacrine Cells/physiology , Computer Simulation , Humans , Visual Pathways/physiology , Photic Stimulation/methods , Nerve Net/physiology , Visual Fields/physiology , Retinal Bipolar Cells/physiology
4.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38514178

ABSTRACT

An organizational feature of neural circuits is the specificity of synaptic connections. A striking example is the direction-selective (DS) circuit of the retina. There are multiple subtypes of DS retinal ganglion cells (DSGCs) that prefer motion along one of four preferred directions. This computation is mediated by selective wiring of a single inhibitory interneuron, the starburst amacrine cell (SAC), with each DSGC subtype preferentially receiving input from a subset of SAC processes. We hypothesize that the molecular basis of this wiring is mediated in part by unique expression profiles of DSGC subtypes. To test this, we first performed paired recordings from isolated mouse retinas of both sexes to determine that postnatal day 10 (P10) represents the age at which asymmetric synapses form. Second, we performed RNA sequencing and differential expression analysis on isolated P10 ON-OFF DSGCs tuned for either nasal or ventral motion and identified candidates which may promote direction-specific wiring. We then used a conditional knock-out strategy to test the role of one candidate, the secreted synaptic organizer cerebellin-4 (Cbln4), in the development of DS tuning. Using two-photon calcium imaging, we observed a small deficit in directional tuning among ventral-preferring DSGCs lacking Cbln4, though whole-cell voltage-clamp recordings did not identify a significant change in inhibitory inputs. This suggests that Cbln4 does not function primarily via a cell-autonomous mechanism to instruct wiring of DS circuits. Nevertheless, our transcriptomic analysis identified unique candidate factors for gaining insights into the molecular mechanisms that instruct wiring specificity in the DS circuit.


Subject(s)
Mice, Inbred C57BL , Retina , Retinal Ganglion Cells , Synapses , Animals , Mice , Retina/metabolism , Retina/physiology , Male , Synapses/physiology , Synapses/metabolism , Female , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Amacrine Cells/metabolism , Motion Perception/physiology , Nerve Net/physiology , Nerve Net/metabolism , Visual Pathways/physiology , Visual Pathways/metabolism
5.
J Fish Biol ; 104(5): 1299-1307, 2024 May.
Article in English | MEDLINE | ID: mdl-38308449

ABSTRACT

We studied the topography of retinal ganglion cells (GCs) and estimated spatial resolving power (SRP) in the pajama cardinalfish Sphaeramia nematoptera (Bleeker, 1856), a relatively small brightly colored fish inhabiting coral reefs and lagoons in the Western Pacific. S. nematoptera is an active night predator feeding on near-bottom animal plankton and benthos. DAPI staining was used to label nuclei of GCs and non-GCs in the inner plexiform and ganglion cell layers. Non-GCs were distinguished from GCs in Nissl-stained retinal wholemounts based on cell size, shape, and staining intensity. The proportion of displaced amacrine cells (DACs) varied from 15.46 ± 1.12 (visual streak [VS]) to 17.99 ± 1.06% (dorsal periphery) (mean ± S.E.M., N = 5); the respective proportions of glial cells were 6.61 ± 0.84 and 5.89 ± 0.76%. Thus, 76%-78% of cells in the ganglion cell layer and inner plexiform layer were GCs. The minimum spatial coverage of GCs (3600-4600 cells/mm2) was detected in the dorsal and ventral periphery. It gradually increased toward the central retina to form a moderate VS. The maximum GC density (11,400-12,400 cells/mm2) was registered in the central portion of the VS. No pronounced concentric retinal specializations were found. The total number of GCs ranged within 595.2-635.9 × 103. The anatomical spatial resolving power was minimum in the ventral periphery (4.91-5.53 cpd) and maximum in the central portion of the VS (8.47-9.07 cpd). The respective minimum separable angles were 0.18-0.20° and 0.11-0.12°. The relatively high spatial resolving power and presence of the VS in the pajama cardinalfish are in line with its highly visual behavior.


Subject(s)
Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Amacrine Cells/cytology
6.
Nat Commun ; 15(1): 1819, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38418467

ABSTRACT

Dendritic mechanisms driving input-output transformation in starburst amacrine cells (SACs) are not fully understood. Here, we combine two-photon subcellular voltage and calcium imaging and electrophysiological recording to determine the computational architecture of mouse SAC dendrites. We found that the perisomatic region integrates motion signals over the entire dendritic field, providing a low-pass-filtered global depolarization to dendrites. Dendrites integrate local synaptic inputs with this global signal in a direction-selective manner. Coincidental local synaptic inputs and the global motion signal in the outward motion direction generate local suprathreshold calcium transients. Moreover, metabotropic glutamate receptor 2 (mGluR2) signaling in SACs modulates the initiation of calcium transients in dendrites but not at the soma. In contrast, voltage-gated potassium channel 3 (Kv3) dampens fast voltage transients at the soma. Together, complementary mGluR2 and Kv3 signaling in different subcellular regions leads to dendritic compartmentalization and direction selectivity, highlighting the importance of these mechanisms in dendritic computation.


Subject(s)
Amacrine Cells , Receptors, Metabotropic Glutamate , Animals , Mice , Amacrine Cells/physiology , Calcium , Signal Transduction , Dendrites/physiology
7.
PLoS Biol ; 22(2): e3002538, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38422167

ABSTRACT

In mammals, starburst amacrine cells are centrally involved in motion vision and a new study in PLOS Biology, by Yan and colleagues finds that zebrafish have them, too. They coexist with a second pair of starburst-like neurons, but neither appears to be strongly motion selective.


Subject(s)
Amacrine Cells , Zebrafish , Animals , Amacrine Cells/physiology , Retina/physiology , Mammals , Cholinergic Agents
8.
Nat Commun ; 15(1): 599, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238324

ABSTRACT

In early sensory systems, cell-type diversity generally increases from the periphery into the brain, resulting in a greater heterogeneity of responses to the same stimuli. Surround suppression is a canonical visual computation that begins within the retina and is found at varying levels across retinal ganglion cell types. Our results show that heterogeneity in the level of surround suppression occurs subcellularly at bipolar cell synapses. Using single-cell electrophysiology and serial block-face scanning electron microscopy, we show that two retinal ganglion cell types exhibit very different levels of surround suppression even though they receive input from the same bipolar cell types. This divergence of the bipolar cell signal occurs through synapse-specific regulation by amacrine cells at the scale of tens of microns. These findings indicate that each synapse of a single bipolar cell can carry a unique visual signal, expanding the number of possible functional channels at the earliest stages of visual processing.


Subject(s)
Retina , Retinal Ganglion Cells , Animals , Mice , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Synapses/physiology
9.
J Neurosci ; 44(10)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-37957014

ABSTRACT

Classic ON-OFF direction-selective ganglion cells (DSGCs) that encode the four cardinal directions were recently shown to also be orientation-selective. To clarify the mechanisms underlying orientation selectivity, we employed a variety of electrophysiological, optogenetic, and gene knock-out strategies to test the relative contributions of glutamate, GABA, and acetylcholine (ACh) input that are known to drive DSGCs, in male and female mouse retinas. Extracellular spike recordings revealed that DSGCs respond preferentially to either vertical or horizontal bars, those that are perpendicular to their preferred-null motion axes. By contrast, the glutamate input to all four DSGC types measured using whole-cell patch-clamp techniques was found to be tuned along the vertical axis. Tuned glutamatergic excitation was heavily reliant on type 5A bipolar cells, which appear to be electrically coupled via connexin 36 containing gap junctions to the vertically oriented processes of wide-field amacrine cells. Vertically tuned inputs are transformed by the GABAergic/cholinergic "starburst" amacrine cells (SACs), which are critical components of the direction-selective circuit, into distinct patterns of inhibition and excitation. Feed-forward SAC inhibition appears to "veto" preferred orientation glutamate excitation in dorsal/ventral (but not nasal/temporal) coding DSGCs "flipping" their orientation tuning by 90° and accounts for the apparent mismatch between glutamate input tuning and the DSGC's spiking response. Together, these results reveal how two distinct synaptic motifs interact to generate complex feature selectivity, shedding light on the intricate circuitry that underlies visual processing in the retina.


Subject(s)
Retina , Retinal Ganglion Cells , Mice , Animals , Male , Female , Retinal Ganglion Cells/physiology , Retina/physiology , Amacrine Cells/physiology , Visual Perception , Glutamic Acid , Photic Stimulation/methods , Neural Inhibition/physiology
10.
Cell Rep ; 42(11): 113440, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37976158

ABSTRACT

Retinal ribbon synapses undergo functional changes after eye opening that remain uncharacterized. Using light-flash stimulation and paired patch-clamp recordings, we examined the maturation of the ribbon synapse between rod bipolar cells (RBCs) and AII-amacrine cells (AII-ACs) after eye opening (postnatal day 14) in the mouse retina at near physiological temperatures. We find that light-evoked excitatory postsynaptic currents (EPSCs) in AII-ACs exhibit a slow sustained component that increases in magnitude with advancing age, whereas a fast transient component remains unchanged. Similarly, paired recordings reveal a dual-component EPSC with a slower sustained component that increases during development, even though the miniature EPSC (mEPSC) amplitude and kinetics do not change significantly. We thus propose that the readily releasable pool of vesicles from RBCs increases after eye opening, and we estimate that a short light flash can evoke the release of ∼4,000 vesicles onto a single mature AII-AC.


Subject(s)
Amacrine Cells , Synapses , Mice , Animals , Amacrine Cells/physiology , Synapses/physiology , Retina/physiology , Retinal Bipolar Cells/physiology , Synaptic Transmission/physiology
11.
Cell Rep ; 42(11): 113390, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37930888

ABSTRACT

In the retina, rod and cone pathways mediate visual signals over a billion-fold range in luminance. AII ("A-two") amacrine cells (ACs) receive signals from both pathways via different bipolar cells, enabling AIIs to operate at night and during the day. Previous work has examined luminance-dependent changes in AII gap junction connectivity, but less is known about how surrounding circuitry shapes AII receptive fields across light levels. Here, we report that moderate contrast stimuli elicit surround inhibition in AIIs under all but the dimmest visual conditions, due to actions of horizontal cells and at least two ACs that inhibit presynaptic bipolar cells. Under photopic (daylight) conditions, surround inhibition transforms AII response kinetics, which are inherited by downstream ganglion cells. Ablating neuronal nitric oxide synthase type-1 (nNOS-1) ACs removes AII surround inhibition under mesopic (dusk/dawn), but not photopic, conditions. Our findings demonstrate how multiple layers of neural circuitry interact to encode signals across a wide physiological range.


Subject(s)
Amacrine Cells , Retina , Amacrine Cells/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells , Gap Junctions/physiology
12.
eNeuro ; 10(8)2023 08.
Article in English | MEDLINE | ID: mdl-37527925

ABSTRACT

Synaptic plasticity is a fundamental feature of the CNS that controls the magnitude of signal transmission between communicating cells. Many electrical synapses exhibit substantial plasticity that modulates the degree of coupling within groups of neurons, alters the fidelity of signal transmission, or even reconfigures functional circuits. In several known examples, such plasticity depends on calcium and is associated with neuronal activity. Calcium-driven signaling is known to promote potentiation of electrical synapses in fish Mauthner cells, mammalian retinal AII amacrine cells, and inferior olive neurons, and to promote depression in thalamic reticular neurons. To measure local calcium dynamics in situ, we developed a transgenic mouse expressing a GCaMP calcium biosensor fused to Connexin 36 (Cx36) at electrical synapses. We examined the sources of calcium for activity-dependent plasticity in retina slices using confocal or Super-Resolution Radial Fluctuations imaging. More than half of Cx36-GCaMP gap junctions responded to puffs of glutamate with transient increases in fluorescence. The responses were strongly dependent on NMDA receptors, in keeping with known activity-dependent signaling in some amacrine cells. We also found that some responses depended on the activity of voltage-gated calcium channels, representing a previously unrecognized source of calcium to control retinal electrical synaptic plasticity. The high prevalence of calcium signals at electrical synapses in response to glutamate application indicates that a large fraction of electrical synapses has the potential to be regulated by neuronal activity. This provides a means to tune circuit connectivity dynamically based on local activity.


Subject(s)
Calcium , Gap Junctions , Mice , Animals , Gap Junctions/physiology , Retina , Connexins/genetics , Amacrine Cells/physiology , Mammals , Gap Junction delta-2 Protein
13.
Sci Rep ; 13(1): 2834, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36808155

ABSTRACT

Stage II cholinergic retinal waves are one of the first instances of neural activity in the visual system as they are present at a developmental timepoint in which light-evoked activity remains largely undetectable. These waves of spontaneous neural activity sweeping across the developing retina are generated by starburst amacrine cells, depolarize retinal ganglion cells, and drive the refinement of retinofugal projections to numerous visual centers in the brain. Building from several well-established models, we assemble a spatial computational model of starburst amacrine cell-mediated wave generation and wave propagation that includes three significant advancements. First, we model the intrinsic spontaneous bursting of the starburst amacrine cells, including the slow afterhyperpolarization, which shapes the stochastic process of wave generation. Second, we establish a mechanism of wave propagation using reciprocal acetylcholine release, synchronizing the bursting activity of neighboring starburst amacrine cells. Third, we model the additional starburst amacrine cell release of GABA, changing the spatial propagation of retinal waves and in certain instances, the directional bias of the retinal wave front. In total, these advancements comprise a now more comprehensive model of wave generation, propagation, and direction bias.


Subject(s)
Amacrine Cells , Retina , Amacrine Cells/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Cholinergic Agents , Acetylcholine
14.
Cell Rep ; 42(2): 112055, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36757846

ABSTRACT

The vertebrate inner retina is driven by photoreceptors whose outputs are already pre-processed; in zebrafish, outer retinal circuits split "color" from "grayscale" information across four cone-photoreceptor types. It remains unclear how the inner retina processes incoming spectral information while also combining cone signals to shape grayscale functions. We address this question by imaging the light-driven responses of amacrine cells (ACs) and bipolar cells (BCs) in larval zebrafish in the presence and pharmacological absence of inner retinal inhibition. We find that ACs enhance opponency in some bipolar cells while at the same time suppressing pre-existing opponency in others, so that, depending on the retinal region, the net change in the number of color-opponent units is essentially zero. To achieve this "dynamic balance," ACs counteract intrinsic color opponency of BCs via the On channel. Consistent with these observations, Off-stratifying ACs are exclusively achromatic, while all color-opponent ACs stratify in the On sublamina.


Subject(s)
Amacrine Cells , Zebrafish , Animals , Amacrine Cells/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology
15.
Vision Res ; 205: 108187, 2023 04.
Article in English | MEDLINE | ID: mdl-36758452

ABSTRACT

By analyzing light-evoked spike responses, cation currents (ΔIC) and chloride currents (ΔICl) of over 100 morphologically-identified retinal ganglion cells (GCs) in dark-adapted mouse retina, we found there are at least 14 functionally- and morphologically-distinct types of RGCs. These cells can be divided into 5 groups based on their patterns of spike response to whole field light steps (SRWFLS), a GC identification scheme commonly used in studies with extracellular recording techniques. We also found that all GCs in the mouse retina express strychnine-sensitive glycine receptors, and receive light-elicited chloride current (ΔICl) accompanied by a conductance increase from narrow-field, glycinergic amacrine cells. As the dark membrane potential of RGC are near the chloride-equilibrium potential, mouse GCs' spike responses are mediated primarily by bipolar cells inputs, and modulated by "shunting inhibition" from narrow-field amacrine cells. Analysis of strychnine actions on light-evoked cation current ΔIC (bipolar cell inputs) in GCs suggests that narrow-field amacrine cells modulate GCs by sending ON-OFF crossover feedback signals to presynaptic bipolar cell axon terminals via sign-inverting glycinergic synapses, and the feedback signals are synergistic to the bipolar cell light responses. Therefore narrow-field amacrine cells enhance light-evoked bipolar cell inputs to GCs by presynaptic "synergistic addition", besides the abovementioned postsynaptic "shunting inhibition" in GCs.


Subject(s)
Amacrine Cells , Retinal Ganglion Cells , Animals , Mice , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Retina/physiology , Strychnine , Chlorides , Cations
16.
J Anat ; 243(2): 204-222, 2023 08.
Article in English | MEDLINE | ID: mdl-35292986

ABSTRACT

The precise specification of cellular fate is thought to ensure the production of the correct number of neurons within a population. Programmed cell death may be an additional mechanism controlling cell number, believed to refine the proper ratio of pre- to post-synaptic neurons for a given species. Here, we consider the size of three different neuronal populations in the rod pathway of the mouse retina: rod photoreceptors, rod bipolar cells, and AII amacrine cells. Across a collection of 28 different strains of mice, large variation in the numbers of all three cell types is present. The variation in their numbers is not correlated, so that the ratio of rods to rod bipolar cells, as well as rod bipolar cells to AII amacrine cells, varies as well. Establishing connectivity between such variable pre- and post-synaptic populations relies upon plasticity that modulates process outgrowth and morphological differentiation, which we explore experimentally for both rod bipolar and AII amacrine cells in a mouse retina with elevated numbers of each cell type. While both rod bipolar dendritic and axonal arbors, along with AII lobular arbors, modulate their areal size in relation to local homotypic cell densities, the dendritic appendages of the AII amacrine cells do not. Rather, these processes exhibit a different form of plasticity, regulating the branching density of their overlapping arbors. Each form of plasticity should ensure uniformity in retinal coverage in the presence of the independent specification of afferent and target cell number.


Subject(s)
Dendrites , Retina , Mice , Animals , Dendrites/physiology , Amacrine Cells/physiology , Axons
17.
Cell Rep ; 41(2): 111484, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223749

ABSTRACT

Midget and parasol ganglion cells (GCs) represent the major output channels from the primate eye to the brain. On-type midget and parasol GCs exhibit a higher background spike rate and thus can respond more linearly to contrast changes than their Off-type counterparts. Here, we show that a calcium-permeable AMPA receptor (CP-AMPAR) antagonist blocks background spiking and sustained light-evoked firing in On-type GCs while preserving transient light responses. These effects are selective for On-GCs and are occluded by a gap-junction blocker suggesting involvement of AII amacrine cells (AII-ACs). Direct recordings from AII-ACs, cobalt uptake experiments, and analyses of transcriptomic data confirm that CP-AMPARs are expressed by primate AII-ACs. Overall, our data demonstrate that under some background light levels, CP-AMPARs at the rod bipolar to AII-AC synapse drive sustained signaling in On-type GCs and thus contribute to the more linear contrast signaling of the primate On- versus Off-pathway.


Subject(s)
Amacrine Cells , Receptors, AMPA , Amacrine Cells/physiology , Animals , Calcium , Cobalt , Primates , Receptors, Calcium-Sensing , Retina/physiology
18.
Vis Neurosci ; 39: E004, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35534787

ABSTRACT

The vertebrate retina contains a large number of different types of neurons that can be distinguished by their morphological properties. Assuming that no location should be without a contribution from the circuitry and function linked to a specific type of neuron, it is expected that the dendritic trees of neurons belonging to a type will cover the retina in a regular manner. Thus, for most types of neurons, the contribution to visual processing is thought to be independent of the exact location of individual neurons across the retina. Here, we have investigated the distribution of AII amacrine cells in rat retina. The AII is a multifunctional amacrine cell found in mammals and involved in synaptic microcircuits that contribute to visual processing under both scotopic and photopic conditions. Previous investigations have suggested that AIIs are regularly distributed, with a nearest-neighbor distance regularity index of ~4. It has been argued, however, that this presumed regularity results from treating somas as points, without taking into account their actual spatial extent which constrains the location of other cells of the same type. When we simulated random distributions of cell bodies with size and density similar to real AIIs, we confirmed that the simulated distributions could not be distinguished from the distributions observed experimentally for AIIs in different regions and eccentricities of the retina. The developmental mechanisms that generate the observed distributions of AIIs remain to be investigated.


Subject(s)
Amacrine Cells , Retina , Amacrine Cells/physiology , Animals , Cell Body , Mammals , Rats , Retina/physiology , Software
19.
Nat Commun ; 13(1): 2862, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35606344

ABSTRACT

From mouse to primate, there is a striking discontinuity in our current understanding of the neural coding of motion direction. In non-primate mammals, directionally selective cell types and circuits are a signature feature of the retina, situated at the earliest stage of the visual process. In primates, by contrast, direction selectivity is a hallmark of motion processing areas in visual cortex, but has not been found in the retina, despite significant effort. Here we combined functional recordings of light-evoked responses and connectomic reconstruction to identify diverse direction-selective cell types in the macaque monkey retina with distinctive physiological properties and synaptic motifs. This circuitry includes an ON-OFF ganglion cell type, a spiking, ON-OFF polyaxonal amacrine cell and the starburst amacrine cell, all of which show direction selectivity. Moreover, we discovered that macaque starburst cells possess a strong, non-GABAergic, antagonistic surround mediated by input from excitatory bipolar cells that is critical for the generation of radial motion sensitivity in these cells. Our findings open a door to investigation of a precortical circuitry that computes motion direction in the primate visual system.


Subject(s)
Connectome , Macaca , Retina , Amacrine Cells/physiology , Animals , Evoked Potentials, Visual/physiology , Macaca/physiology , Mammals , Mice , Primates/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Synapses/physiology
20.
Curr Biol ; 32(11): 2529-2538.e4, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35588744

ABSTRACT

The detection of motion direction is a fundamental visual function and a classic model for neural computation. In the non-primate retina, direction selectivity arises in starburst amacrine cell (SAC) dendrites, which provide selective inhibition to direction-selective retinal ganglion cells (dsRGCs). Although SACs are present in primates, their connectivity and the existence of dsRGCs remain open questions. Here, we present a connectomic reconstruction of the primate ON SAC circuit from a serial electron microscopy volume of the macaque central retina. We show that the structural basis for the SACs' ability to confer directional selectivity on postsynaptic neurons is conserved. SACs selectively target a candidate homolog to the mammalian ON-sustained dsRGCs that project to the accessory optic system (AOS) and contribute to gaze-stabilizing reflexes. These results indicate that the capacity to compute motion direction is present in the retina, which is earlier in the primate visual system than classically thought.


Subject(s)
Amacrine Cells , Connectome , Amacrine Cells/physiology , Animals , Dendrites/physiology , Mammals , Primates , Retina/physiology , Retinal Ganglion Cells/physiology , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...