Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Dev Biol ; 476: 218-239, 2021 08.
Article in English | MEDLINE | ID: mdl-33848537

ABSTRACT

Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.


Subject(s)
Photoreceptor Cells/cytology , Synapses/metabolism , Vision, Ocular/physiology , Animals , Humans , Interneurons/physiology , Photoreceptor Cells/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology
2.
Dev Biol ; 476: 88-100, 2021 08.
Article in English | MEDLINE | ID: mdl-33774011

ABSTRACT

During retinal development, multipotent and restricted progenitor cells generate all of the neuronal cells of the retina. Among these are horizontal cells, which are interneurons that modulate the light-induced signal from photoreceptors. This study utilizes the identification of novel cis-regulatory elements as a method to examine the gene regulatory networks that direct the development of horizontal cells. Here we describe a screen for cis-regulatory elements, or enhancers, for the horizontal cell-associated genes PTF1A, ONECUT1 (OC1), TFAP2A (AP2A), and LHX1. The OC1ECR22 and Tfap2aACR5 elements were shown to be potential enhancers for OC1 and TFAP2A, respectively, and to be specifically active in developing horizontal cells. The OC1ECR22 element is activated by PTF1A and RBPJ, which translates to regulation of OC1 expression and suggests that PTF1A is a direct activator of OC1 expression in developing horizontal cells. The region within the Tfap2aACR5 element that is responsible for its activation was determined to be a 100 bp sequence named Motif 4. Both OC1ECR22 and Tfap2aACR5 are negatively regulated by the nuclear receptors THRB and RXRG, as is the expression of OC1 and AP2A, suggesting that nuclear receptors may have a role in the negative regulation of horizontal cell development.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Retina/embryology , Retinal Horizontal Cells/metabolism , Animals , Cell Differentiation/physiology , Chick Embryo , Gene Expression/genetics , Gene Regulatory Networks/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Neurons/metabolism , Onecut Transcription Factors , Retina/metabolism , Retinal Horizontal Cells/physiology , Stem Cells/metabolism , Transcription Factor AP-2 , Transcription Factors/metabolism
3.
J Comp Neurol ; 529(8): 1756-1767, 2021 06.
Article in English | MEDLINE | ID: mdl-33070331

ABSTRACT

Horizontal cells (HCs) are neurons of the outer retina, which provide inhibitory feedback onto photoreceptors and contribute to image processing. HCs in teleosts are classified into four subtypes (H1-H4), each having different roles: H1-H3 feed back onto different sets of cones, H4 feed back onto rods, and only H1 store and release the inhibitory neurotransmitter, γ-aminobutyric acid (GABA). Dissociated HCs exhibit spontaneous Ca2+ -based action potentials (APs), yet it is unclear if APs occur in situ, or if all subtypes exhibit APs. We measured intracellular Ca2+ and report APs in slice preparations of the goldfish retina. In HCs furthest from photoreceptors (i.e., H3/H4), APs were less frequent, with greater duration and area under the curve (a measure of Ca2+ flux). Next, we classified acutely dissociated HCs into subtypes by integrating the ratio of dendritic field size vs. soma size (rd/s ). H1 and H2 subtypes had low rd/s values (<8); H3/H4 had high rd/s (>12). To verify this model, H1s were identified by immunoreactivity for GABA and 95% of these cells had an rd/s < 4. In Ca2+ imaging experiments, as rd/s increased, AP duration and area under the curve increased, while frequency decreased. Our results demonstrate the presence of Ca2+ -based APs in the goldfish retina in situ and show that HC subtypes H1 through H4 exhibit progressively longer and less frequent spontaneous APs. These results suggest that APs may play an important role in inhibitory feedback, and may have implications for understanding the relative contributions of HC subtypes in the outer retina.


Subject(s)
Action Potentials/physiology , Retinal Horizontal Cells/physiology , Visual Perception/physiology , Animals , Goldfish
4.
Neuron ; 107(2): 320-337.e6, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32473094

ABSTRACT

In the eye, the function of same-type photoreceptors must be regionally adjusted to process a highly asymmetrical natural visual world. Here, we show that UV cones in the larval zebrafish area temporalis are specifically tuned for UV-bright prey capture in their upper frontal visual field, which may use the signal from a single cone at a time. For this, UV-photon detection probability is regionally boosted more than 10-fold. Next, in vivo two-photon imaging, transcriptomics, and computational modeling reveal that these cones use an elevated baseline of synaptic calcium to facilitate the encoding of bright objects, which in turn results from expressional tuning of phototransduction genes. Moreover, the light-driven synaptic calcium signal is regionally slowed by interactions with horizontal cells and later accentuated at the level of glutamate release driving retinal networks. These regional differences tally with variations between peripheral and foveal cones in primates and hint at a common mechanistic origin.


Subject(s)
Photoreceptor Cells, Vertebrate/physiology , Predatory Behavior/physiology , Retinal Cone Photoreceptor Cells/physiology , Zebrafish/physiology , Animals , Calcium Signaling , Computer Simulation , Glutamic Acid/metabolism , Larva , Light , Light Signal Transduction , Retinal Horizontal Cells/physiology , Synapses/physiology , Transcriptome , Ultraviolet Rays , Visual Fields
5.
J Neurophysiol ; 122(6): 2284-2293, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31596629

ABSTRACT

Horizontal cells (HCs) are interneurons of the outer retina that undergo graded changes in membrane potential during the light response and provide feedback to photoreceptors. We characterized spontaneous Ca2+-based action potentials (APs) in isolated goldfish (Carassius auratus) HCs with electrophysiological and intracellular imaging techniques. Transient changes in intracellular Ca2+ concentration ([Ca2+]i) were observed with fura-2 and were abolished by removal of extracellular Ca2+ or by inhibition of Ca2+ channels by 50 µM Cd2+ or 100 µM nifedipine. Inhibition of Ca2+ release from stores with 20 µM ryanodine or 50 µM dantrolene abolished Ca2+ transients and increased baseline [Ca2+]i. This increased baseline was prevented by blocking L-type Ca2+ channels with nifedipine, suggesting that Ca2+-induced Ca2+ release from stores may be needed to inactivate membrane Ca2+ channels. Caffeine (3 mM) increased the frequency of Ca2+ transients, and the store-operated channel antagonist 2-aminoethyldiphenylborinate (100 µM) counteracted this effect. APs were detected with voltage-sensitive dye imaging (FluoVolt) and current-clamp electrophysiology. In current-clamp recordings, regenerative APs were abolished by removal of extracellular Ca2+ or in the presence of 5 mM Co2+ or 100 µM nifedipine, and APs were amplified with 15 mM Ba2+. Collectively, our data suggest that during APs Ca2+ enters through L-type Ca2+ channels and that Ca2+ stores (gated by ryanodine receptors) contribute to the rise in [Ca2+]i. This work may lead to further understanding of the possible role APs have in vision, such as transitioning from light to darkness or modulating feedback from HCs to photoreceptors.NEW & NOTEWORTHY Horizontal cells (HCs) are interneurons of the outer retina that provide inhibitory feedback onto photoreceptors. HCs respond to light via graded changes in membrane potential. We characterized spontaneous action potentials in HCs from goldfish and linked action potential generation to a rise in intracellular Ca2+ via plasma membrane channels and ryanodine receptors. Action potentials may play a role in vision, such as transitioning from light to darkness, or in modulating feedback from HCs to photoreceptors.


Subject(s)
Action Potentials/physiology , Calcium/metabolism , Retinal Horizontal Cells/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Goldfish
6.
PLoS One ; 14(8): e0218818, 2019.
Article in English | MEDLINE | ID: mdl-31461464

ABSTRACT

Although it is well established that the vertebrate retina contains endogenous circadian clocks that regulate retinal physiology and function during day and night, the processes that the clocks affect and the means by which the clocks control these processes remain unresolved. We previously demonstrated that a circadian clock in the goldfish retina regulates rod-cone electrical coupling so that coupling is weak during the day and robust at night. The increase in rod-cone coupling at night introduces rod signals into cones so that the light responses of both cones and cone horizontal cells, which are post-synaptic to cones, become dominated by rod input. By comparing the light responses of cones, cone horizontal cells and rod horizontal cells, which are post-synaptic to rods, under dark-adapted conditions during day and night, we determined whether the daily changes in the strength of rod-cone coupling could account entirely for rhythmic changes in the light response properties of cones and cone horizontal cells. We report that although some aspects of the day/night changes in cone and cone horizontal cell light responses, such as response threshold and spectral tuning, are consistent with modulation of rod-cone coupling, other properties cannot be solely explained by this phenomenon. Specifically, we found that at night compared to the day the time course of spectrally-isolated cone photoresponses was slower, cone-to-cone horizontal cell synaptic transfer was highly non-linear and of lower gain, and the delay in cone-to-cone horizontal cell synaptic transmission was longer. However, under bright light-adapted conditions in both day and night, cone-to-cone horizontal cell synaptic transfer was linear and of high gain, and no additional delay was observed at the cone-to-cone horizontal cell synapse. These findings suggest that in addition to controlling rod-cone coupling, retinal clocks shape the light responses of cone horizontal cells by modulating cone-to-cone horizontal cell synaptic transmission.


Subject(s)
Circadian Clocks/physiology , Goldfish , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synaptic Transmission/physiology , Animals , Circadian Clocks/radiation effects , Light , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Horizontal Cells/radiation effects , Synaptic Transmission/radiation effects
7.
PLoS Biol ; 17(4): e3000200, 2019 04.
Article in English | MEDLINE | ID: mdl-30933967

ABSTRACT

The stream of visual information sent from photoreceptors to second-order bipolar cells is intercepted by laterally interacting horizontal cells that generate feedback to optimize and improve the efficiency of signal transmission. The mechanisms underlying the regulation of graded photoreceptor synaptic output in this nonspiking network have remained elusive. Here, we analyze with patch clamp recording the novel mechanisms by which horizontal cells control pH in the synaptic cleft to modulate photoreceptor neurotransmitter release. First, we show that mammalian horizontal cells respond to their own GABA release and that the results of this autaptic action affect cone voltage-gated Ca2+ channel (CaV channel) gating through changes in pH. As a proof-of-principle, we demonstrate that chemogenetic manipulation of horizontal cells with exogenous anion channel expression mimics GABA-mediated cone CaV channel inhibition. Activation of these GABA receptor anion channels can depolarize horizontal cells and increase cleft acidity via Na+/H+ exchanger (NHE) proton extrusion, which results in inhibition of cone CaV channels. This action is effectively counteracted when horizontal cells are sufficiently hyperpolarized by increased GABA receptor (GABAR)-mediated HCO3- efflux, alkalinizing the cleft and disinhibiting cone CaV channels. This demonstrates how hybrid actions of GABA operate in parallel to effect voltage-dependent pH changes, a novel mechanism for regulating synaptic output.


Subject(s)
Photoreceptor Cells, Vertebrate/physiology , Retinal Horizontal Cells/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology , Animals , Calcium Channels/metabolism , Feedback , Feedback, Physiological/physiology , Female , Guinea Pigs , Hydrogen-Ion Concentration , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Retina/cytology , Retina/metabolism , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Signal Transduction/physiology , Synapses/metabolism , Synaptic Transmission/physiology
8.
J Neurosci ; 39(4): 651-662, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30504272

ABSTRACT

Lateral inhibition in the vertebrate retina depends on a negative feedback synapse between horizontal cells (HCs) and rod and cone photoreceptors. A change in pH is thought to be the signal for negative feedback, but its spatial profile in the synaptic cleft is unknown. Here we use three different membrane proteins, each fused to the same genetically-encoded pH-sensitive Green Fluorescent Protein (GFP) (pHluorin), to probe synaptic pH in retina from transgenic zebrafish (Danio rerio) of either sex. We used the cone transducin promoter to express SynaptopHluorin (pHluorin on vesicle-associated membrane protein (VAMP2)) or CalipHluorin (pHluorin on an L-type Ca2+ channel) and the HC-specific connexin-55.5 promoter to express AMPApHluorin (pHluorin on an AMPA receptor). Stimulus light led to increased fluorescence of all three probes, consistent with alkalinization of the synaptic cleft. The receptive field size, sensitivity to surround illumination, and response to activation of an alien receptor expressed exclusively in HCs, are consistent with lateral inhibition as the trigger for alkalinization. However, SynaptopHluorin and AMPApHluorin, which are displaced farther from cone synaptic ribbons than CalipHluorin, reported a smaller pH change. Hence, unlike feedforward glutamatergic transmission, which spills over to allow cross talk between terminals in the cone network, the pH change underlying HC feedback is compartmentalized to individual synaptic invaginations within a cone terminal, consistent with private line communication.SIGNIFICANCE STATEMENT Lateral inhibition (LI) is a fundamental feature of information processing in sensory systems, enhancing contrast sensitivity and enabling edge discrimination. Horizontal cells (HCs) are the first cellular substrate of LI in the vertebrate retina, but the synaptic mechanisms underlying LI are not completely understood, despite decades of study. This paper makes a significant contribution to our understanding of LI, by showing that each HC-cone synapse is a "private-line" that operates independently from other HC-cone connections. Using transgenic zebrafish expressing pHluorin, a pH-sensitive GFP variant spliced onto three different protein platforms expressed either in cones or HCs we show that the feedback pH signal is constrained to individual cone terminals, and more stringently, to individual synaptic contact sites within each terminal.


Subject(s)
Feedback, Physiological/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Connexins/metabolism , Female , Glutamates/physiology , Hydrogen-Ion Concentration , Male , Protons , Receptors, AMPA/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Horizontal Cells/ultrastructure , Synapses/ultrastructure , Synaptic Transmission/physiology , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/physiology , Zebrafish
10.
Neuron ; 99(1): 117-134.e11, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29937281

ABSTRACT

Many brain regions contain local interneurons of distinct types. How does an interneuron type contribute to the input-output transformations of a given brain region? We addressed this question in the mouse retina by chemogenetically perturbing horizontal cells, an interneuron type providing feedback at the first visual synapse, while monitoring the light-driven spiking activity in thousands of ganglion cells, the retinal output neurons. We uncovered six reversible perturbation-induced effects in the response dynamics and response range of ganglion cells. The effects were enhancing or suppressive, occurred in different response epochs, and depended on the ganglion cell type. A computational model of the retinal circuitry reproduced all perturbation-induced effects and led us to assign specific functions to horizontal cells with respect to different ganglion cell types. Our combined experimental and theoretical work reveals how a single interneuron type can differentially shape the dynamical properties of distinct output channels of a brain region.


Subject(s)
Feedback , Interneurons/physiology , Retinal Ganglion Cells/physiology , Retinal Horizontal Cells/physiology , Vision, Ocular/physiology , Animals , Calcium/metabolism , Mice , Models, Neurological , Photoreceptor Cells, Vertebrate , Retinal Bipolar Cells , Synapses
11.
Methods Mol Biol ; 1753: 179-189, 2018.
Article in English | MEDLINE | ID: mdl-29564789

ABSTRACT

Genetic manipulation of horizontal cells using a Connexin57-iCre mouse (Cx57-iCre) line combined with calcium imaging is proving to be a valuable method to study horizontal cell feedback inhibition onto photoreceptor terminals. While it is accepted that horizontal cells provide lateral inhibitory feedback to photoreceptors, the cellular mechanisms that underlie this feedback inhibition remain only partially elucidated. Feedback inhibition of photoreceptors acts via modulation of their voltage-gated calcium channels at their synaptic terminal. Calcium imaging of photoreceptors in retinal slices, therefore, reflects the impact of inhibitory feedback from horizontal cells. The development of a Cx57-iCre mouse line permits genetic manipulation of horizontal cells. In wild-type mouse retina, depolarization of horizontal cells by kainate provokes a decrease in photoreceptor Ca2+i, whereas hyperpolarization by NBQX elicits an increase in photoreceptor Ca2+i. These responses indicate increased feedback inhibition occurred when horizontal cells are depolarized, and decreased feedback inhibition, when hyperpolarized. This system was used to test the role of GABA release from horizontal cells in feedback inhibition by the selective elimination of VGAT/VIAAT, the inhibitory amino acid transmitter transporter that loads GABA into the synaptic vesicles of horizontal cells. Combined with calcium imaging of photoreceptors in retinal slices, the knockout of specific proteins, e.g., VGAT, provides a robust technique to test the role of GABA in feedback inhibition by horizontal cells.


Subject(s)
Feedback, Physiological/physiology , Molecular Imaging/methods , Optical Imaging/methods , Photoreceptor Cells, Vertebrate/physiology , Retinal Horizontal Cells/physiology , Animals , Calcium/chemistry , Calcium/metabolism , Calcium Channels/metabolism , Connexins/genetics , Feedback, Physiological/drug effects , Immunohistochemistry/instrumentation , Immunohistochemistry/methods , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Molecular Imaging/instrumentation , Optical Imaging/instrumentation , Photoreceptor Cells, Vertebrate/drug effects , Quinoxalines/pharmacology , Retinal Horizontal Cells/drug effects
12.
J Neurophysiol ; 119(4): 1437-1449, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29357459

ABSTRACT

A persistent change in illumination causes light-adaptive changes in retinal neurons. Light adaptation improves visual encoding by preventing saturation and by adjusting spatiotemporal integration to increase the signal-to-noise ratio (SNR) and utilize signaling bandwidth efficiently. In dim light, the visual input contains a greater relative amount of quantal noise, and vertebrate receptive fields are extended in space and time to increase SNR. Whereas in bright light, SNR of the visual input is high, the rate of synaptic vesicle release from the photoreceptors is low so that quantal noise in synaptic output may limit SNR postsynaptically. Whether and how reduced synaptic SNR impacts spatiotemporal integration in postsynaptic neurons remains unclear. To address this, we measured spatiotemporal integration in retinal horizontal cells and ganglion cells in the guinea pig retina across a broad illumination range, from low to high photopic levels. In both cell types, the extent of spatial and temporal integration changed according to an inverted U-shaped function consistent with adaptation to low SNR at both low and high light levels. We show how a simple mechanistic model with interacting, opponent filters can generate the observed changes in ganglion cell spatiotemporal receptive fields across light-adaptive states and postulate that retinal neurons postsynaptic to the cones in bright light adopt low-pass spatiotemporal response characteristics to improve visual encoding under conditions of low synaptic SNR.


Subject(s)
Adaptation, Ocular/physiology , Electrophysiological Phenomena/physiology , Retinal Ganglion Cells/physiology , Retinal Horizontal Cells/physiology , Animals , Female , Guinea Pigs , Male , Photic Stimulation , Signal-To-Noise Ratio
13.
Proc Natl Acad Sci U S A ; 114(48): 12803-12808, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29138314

ABSTRACT

Ganglion cells (GCs) are fundamental to retinal neural circuitry, processing photoreceptor signals for transmission to the brain via their axons. However, much remains unknown about their role in vision and their vulnerability to disease leading to blindness. A major bottleneck has been our inability to observe GCs and their degeneration in the living human eye. Despite two decades of development of optical technologies to image cells in the living human retina, GCs remain elusive due to their high optical translucency. Failure of conventional imaging-using predominately singly scattered light-to reveal GCs has led to a focus on multiply-scattered, fluorescence, two-photon, and phase imaging techniques to enhance GC contrast. Here, we show that singly scattered light actually carries substantial information that reveals GC somas, axons, and other retinal neurons and permits their quantitative analysis. We perform morphometry on GC layer somas, including projection of GCs onto photoreceptors and identification of the primary GC subtypes, even beneath nerve fibers. We obtained singly scattered images by: (i) marrying adaptive optics to optical coherence tomography to avoid optical blurring of the eye; (ii) performing 3D subcellular image registration to avoid motion blur; and (iii) using organelle motility inside somas as an intrinsic contrast agent. Moreover, through-focus imaging offers the potential to spatially map individual GCs to underlying amacrine, bipolar, horizontal, photoreceptor, and retinal pigment epithelium cells, thus exposing the anatomical substrate for neural processing of visual information. This imaging modality is also a tool for improving clinical diagnosis and assessing treatment of retinal disease.


Subject(s)
Amacrine Cells/ultrastructure , Optics and Photonics/methods , Retinal Bipolar Cells/ultrastructure , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Ganglion Cells/ultrastructure , Retinal Horizontal Cells/ultrastructure , Tomography, Optical Coherence/methods , Adult , Amacrine Cells/physiology , Cell Count , Healthy Volunteers , Humans , Middle Aged , Nerve Fibers/physiology , Nerve Fibers/ultrastructure , Optics and Photonics/instrumentation , Retinal Bipolar Cells/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Ganglion Cells/physiology , Retinal Horizontal Cells/physiology , Tomography, Optical Coherence/instrumentation , Vision, Ocular/physiology
14.
Sci Rep ; 7(1): 5540, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28717219

ABSTRACT

In the retinal circuit, environmental light signals are converted into electrical signals that can be decoded properly by the brain. At the first synapse of the visual system, information flow from photoreceptors to bipolar cells is modulated by horizontal cells (HCs), however, their functional contribution to retinal output and individual visual function is not fully understood. In the current study, we investigated functional roles for HCs in retinal ganglion cell (RGC) response properties and optokinetic responses by establishing a HC-depleted mouse line. We observed that HC depletion impairs the antagonistic center-surround receptive field formation of RGCs, supporting a previously reported HC function revealed by pharmacological approaches. In addition, we found that HC loss reduces both the ON and OFF response diversities of RGCs, impairs adjustment of the sensitivity to ambient light at the retinal output level, and alters spatial frequency tuning at an individual level. Taken together, our current study suggests multiple functional aspects of HCs crucial for visual processing.


Subject(s)
Retina/cytology , Retinal Horizontal Cells/physiology , Animals , Connexins/genetics , Electrophysiology/methods , Light , Mice, Transgenic , Retina/physiology , Retinal Ganglion Cells/physiology , Retinal Photoreceptor Cell Outer Segment/physiology , Synapses/physiology , Vision, Ocular/genetics
15.
J Neurosci ; 37(17): 4618-4634, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363980

ABSTRACT

G-protein ßγ subunits (Gßγ) interact with presynaptic proteins and regulate neurotransmitter release downstream of Ca2+ influx. To accomplish their roles in sensory signaling, photoreceptor synapses use specialized presynaptic proteins that support neurotransmission at active zone structures known as ribbons. While several G-protein coupled receptors (GPCRs) influence synaptic transmission at ribbon synapses of cones and other retinal neurons, it is unknown whether Gßγ contributes to these effects. We tested whether activation of one particular GPCR, a metabotropic glutamate receptor (mGluR), can reduce cone synaptic transmission via Gßγ in tiger salamander retinas. In recordings from horizontal cells, we found that an mGluR agonist (L-AP4) reduced cone-driven light responses and mEPSC frequency. In paired recordings of cones and horizontal cells, L-AP4 slightly reduced cone ICa (∼10%) and caused a larger reduction in cone-driven EPSCs (∼30%). Proximity ligation assay revealed direct interactions between SNAP-25 and Gßγ subunits in retinal synaptic layers. Pretreatment with the SNAP-25 cleaving protease BoNT/A inhibited L-AP4 effects on synaptic transmission, as did introduction of a peptide derived from the SNAP-25 C terminus. Introducing Gßγ subunits directly into cones reduced EPSC amplitude. This effect was inhibited by BoNT/A, supporting a role for Gßγ/SNAP-25 interactions. However, the mGluR-dependent reduction in ICa was not mimicked by Gßγ, indicating that this effect was independent of Gßγ. The finding that synaptic transmission at cone ribbon synapses is regulated by Gßγ/SNAP-25 interactions indicates that these mechanisms are shared by conventional and ribbon-type synapses. Gßγ liberated from other photoreceptor GPCRs is also likely to regulate synaptic transmission.SIGNIFICANCE STATEMENT Dynamic regulation of synaptic transmission by presynaptic G-protein coupled receptors shapes information flow through neural circuits. At the first synapse in the visual system, presynaptic metabotropic glutamate receptors (mGluRs) regulate cone photoreceptor synaptic transmission, although the mechanisms and functional impact of this are unclear. We show that mGluRs regulate light response encoding across the cone synapse, accomplished in part by triggering G-protein ßγ subunits (Gßγ) interactions with SNAP-25, a core component of the synaptic vesicle fusion machinery. In addition to revealing a role in visual processing, this provides the first demonstration that Gßγ/SNAP-25 interactions regulate synaptic function at a ribbon-type synapse, contributing to an emerging picture of the ubiquity of Gßγ/SNARE interactions in regulating synaptic transmission throughout the nervous system.


Subject(s)
Ambystoma/physiology , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Cone Photoreceptor Cells/physiology , SNARE Proteins/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Receptors, Metabotropic Glutamate/drug effects , Retinal Horizontal Cells/metabolism , Retinal Horizontal Cells/physiology
16.
J Physiol ; 595(16): 5495-5506, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28378516

ABSTRACT

The first synapse of the retina plays a fundamental role in the visual system. Due to its importance, it is critical that it encodes information from the outside world with the greatest accuracy and precision possible. Cone photoreceptor axon terminals contain many individual synaptic sites, each represented by a presynaptic structure called a 'ribbon'. These synapses are both highly sophisticated and conserved. Each ribbon relays the light signal to one ON cone bipolar cell and several OFF cone bipolar cells, while two dendritic processes from a GABAergic interneuron, the horizontal cell, modulate the cone output via parallel feedback mechanisms. The presence of these three partners within a single synapse has raised numerous questions, and its anatomical and functional complexity is still only partially understood. However, the understanding of this synapse has recently evolved, as a consequence of progress in understanding dendritic signal processing and its role in facilitating global versus local signalling. Indeed, for the downstream retinal network, dendritic processing in horizontal cells may be essential, as they must support important functional operations such as contrast enhancement, which requires spatial averaging of the photoreceptor array, while at the same time preserving accurate spatial information. Here, we review recent progress made towards a better understanding of the cone synapse, with an emphasis on horizontal cell function, and discuss why such complexity might be necessary for early visual processing.


Subject(s)
Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology , Animals , Interneurons/physiology
17.
J Neurophysiol ; 117(2): 523-536, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27832601

ABSTRACT

Horizontal cells (HCs) are inhibitory interneurons of the vertebrate retina. Unlike typical neurons, HCs are chronically depolarized in the dark, leading to a constant influx of Ca2+ Therefore, mechanisms of Ca2+ homeostasis in HCs must differ from neurons elsewhere in the central nervous system, which undergo excitotoxicity when they are chronically depolarized or stressed with Ca2+ HCs are especially well characterized in teleost fish and have been used to unlock mysteries of the vertebrate retina for over one century. More recently, mammalian models of the retina have been increasingly informative for HC physiology. We draw from both teleost and mammalian models in this review, using a comparative approach to examine what is known about Ca2+ pathways in vertebrate HCs. We begin with a survey of Ca2+-permeable ion channels, exchangers, and pumps and summarize Ca2+ influx and efflux pathways, buffering, and intracellular stores. This includes evidence for Ca2+-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors and N-methyl-d-aspartate receptors and for voltage-gated Ca2+ channels. Special attention is given to interactions between ion channels, to differences among species, and in which subtypes of HCs these channels have been found. We then discuss a number of unresolved issues pertaining to Ca2+ dynamics in HCs, including a potential role for Ca2+ in feedback to photoreceptors, the role for Ca2+-induced Ca2+ release, and the properties and functions of Ca2+-based action potentials. This review aims to highlight the unique Ca2+ dynamics in HCs, as these are inextricably tied to retinal function.


Subject(s)
Calcium/metabolism , Nonlinear Dynamics , Retina/cytology , Retinal Horizontal Cells/physiology , Animals , Fishes , Vertebrates
18.
PLoS One ; 11(11): e0163925, 2016.
Article in English | MEDLINE | ID: mdl-27861495

ABSTRACT

Opsin family genes encode G protein-coupled seven-transmembrane proteins that bind a retinaldehyde chromophore in photoreception. Here, we sought potential as yet undescribed avian retinal photoreceptors, focusing on Opsin 3 homologs in the chicken. We found two Opsin 3-related genes in the chicken genome: one corresponding to encephalopsin/panopsin (Opn3) in mammals, and the other belonging to the teleost multiple tissue opsin (TMT) 2 group. Bioluminescence imaging and G protein activation assays demonstrated that the chicken TMT opsin (cTMT) functions as a blue light sensor when forced-expressed in mammalian cultured cells. We did not detect evidence of light sensitivity for the chicken Opn3 (cOpn3). In situ hybridization demonstrated expression of cTMT in subsets of differentiating cells in the inner retina and, as development progressed, predominant localization to retinal horizontal cells (HCs). Immunohistochemistry (IHC) revealed cTMT in HCs as well as in small numbers of cells in the ganglion and inner nuclear layers of the post-hatch chicken retina. In contrast, cOpn3-IR cells were found in distinct subsets of cells in the inner nuclear layer. cTMT-IR cells were also found in subsets of cells in the hypothalamus. Finally, we found differential distribution of cOpn3 and cTMT proteins in specific cells of the cerebellum. The present results suggest that a novel TMT-type opsin 3 may function as a photoreceptor in the chicken retina and brain.


Subject(s)
Brain/metabolism , Retina/metabolism , Rod Opsins/metabolism , Animals , Brain/cytology , Calcium/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Chickens , Exons , Gene Expression , Genomics , Hypothalamus/cytology , Hypothalamus/metabolism , Introns , Light , Multigene Family , Phylogeny , Protein Transport , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Retinal Horizontal Cells/physiology , Rod Opsins/genetics
19.
Proc Natl Acad Sci U S A ; 113(46): 13215-13220, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27789727

ABSTRACT

In the vertebrate retina, three types of photoreceptors-visual photoreceptor cones and rods and the intrinsically photosensitive retinal ganglion cells (ipRGCs)-converged through evolution to detect light and regulate image- and nonimage-forming activities such as photic entrainment of circadian rhythms, pupillary light reflexes, etc. ipRGCs express the nonvisual photopigment melanopsin (OPN4), encoded by two genes: the Xenopus (Opn4x) and mammalian (Opn4m) orthologs. In the chicken retina, both OPN4 proteins are found in ipRGCs, and Opn4x is also present in retinal horizontal cells (HCs), which connect with visual photoreceptors. Here we investigate the intrinsic photosensitivity and functioning of HCs from primary cultures of embryonic retinas at day 15 by using calcium fluorescent fluo4 imaging, pharmacological inhibitory treatments, and Opn4x knockdown. Results show that HCs are avian photoreceptors with a retinal-based OPN4X photopigment conferring intrinsic photosensitivity. Light responses in HCs appear to be driven through an ancient type of phototransduction cascade similar to that in rhabdomeric photoreceptors involving a G-protein q, the activation of phospholipase C, calcium mobilization, and the release of the inhibitory neurotransmitter GABA. Based on their intrinsic photosensitivity, HCs may have a key dual function in the retina of vertebrates, potentially regulating nonvisual tasks together with their sister cells, ipRGCs, and with visual photoreceptors, modulating lateral interactions and retinal processing.


Subject(s)
Photoreceptor Cells, Vertebrate/physiology , Retinal Horizontal Cells/physiology , Rod Opsins/physiology , Animals , Calcium/physiology , Cells, Cultured , Chickens , Embryo, Nonmammalian , Light , Retinaldehyde/physiology , Rod Opsins/genetics , gamma-Aminobutyric Acid/physiology
20.
J Neurophysiol ; 116(6): 2799-2814, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27707811

ABSTRACT

The functional and morphological connectivity between various horizontal cell (HC) types (H1, H2, H3, and H4) and photoreceptors was studied in zebrafish retina. Since HCs are strongly coupled by gap junctions and feedback from HCs to photoreceptors depends strongly on connexin (Cx) hemichannels, we characterized the various HC Cxs (Cx52.6, Cx52.7, Cx52.9, and Cx55.5) in Xenopus oocytes. All Cxs formed hemichannels that were conducting at physiological membrane potentials. The Cx hemichannels differed in kinetic properties and voltage dependence, allowing for specific tuning of the coupling of HCs and the feedback signal from HCs to cones. The morphological connectivity between HC layers and cones was determined next. We used zebrafish expressing green fluorescent protein under the control of Cx promoters. We found that all HCs showed Cx55.5 promoter activity. Cx52.7 promoter activity was exclusively present in H4 cells, while Cx52.9 promoter activity occurred only in H1 cells. Cx52.6 promoter activity was present in H4 cells and in the ventral quadrant of the retina also in H1 cells. Finally, we determined the spectral sensitivities of the HC layers. Three response types were found. Monophasic responses were generated by HCs that contacted all cones (H1 cells), biphasic responses were generated by HCs that contacted M, S, and UV cones (H2 cells), and triphasic responses were generated by HCs that contacted either S and UV cones (H3 cells) or rods and UV cones (H4 cells). Electron microscopy confirms that H4 cells innervate cones. This indicates that rod-driven HCs process spectral information during photopic and luminance information during scotopic conditions.


Subject(s)
Gap Junctions/physiology , Green Fluorescent Proteins/metabolism , Membrane Potentials/physiology , Photoreceptor Cells, Vertebrate/physiology , Retina/cytology , Retinal Horizontal Cells/physiology , Analysis of Variance , Animals , Animals, Genetically Modified , Biophysics , Biotin/analogs & derivatives , Biotin/metabolism , Connexins/genetics , Connexins/metabolism , Electric Stimulation , Feedback, Physiological/physiology , Gap Junctions/ultrastructure , Green Fluorescent Proteins/genetics , Microinjections , Microscopy, Confocal , Microscopy, Electron , Oocytes , Patch-Clamp Techniques , Photoreceptor Cells, Vertebrate/classification , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Horizontal Cells/classification , Retinal Horizontal Cells/ultrastructure , Transduction, Genetic , Xenopus laevis , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...