Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(13): 2291-2304, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36828637

ABSTRACT

Increasing spike rates drive greater neuronal energy demand. In turn, mitochondrial ATP production leads to the generation of reactive oxygen species (ROS) that can modulate ion channel gating. Does ROS production autoregulate the excitability of a neuron? We investigated the links between retinal ganglion cell (RGC) excitability and spike activity-driven ROS production in male and female mice. Changes to the light-evoked and current-evoked spike patterns of functionally identified αRGC subtypes, along with their NaV channel-gating properties, were recorded during experimentally induced decreases and increases of intracellular ROS. During periods of highest spike rates (e.g., following light onset in ON sustained RGCs and light offset in OFF sustained RGCs), these αRGC subtypes responded to reductions of ROS (induced by catalase or glutathione monoethyl ester) with higher spike rates. Increases in ROS (induced by mercaptosuccinate, antimycin-A, or H2O2) lowered spike rates. In ON and OFF transient RGCs, there were no changes in spike rate during ROS decreases but increased ROS increased spiking. This suggests that endogenous ROS are intrinsic neuromodulators in RGCs having high metabolic demands but not in RGCs with lower energy needs. We identified ROS-induced shifts in the voltage-dependent gating of specific isoforms of NaV channels that account for the modulation of ON and OFF sustained RGC spike frequency by ROS-mediated feedback. ROS-induced changes to NaV channel gating, affecting activation and inactivation kinetics, are consistent with the differing spike pattern alterations observed in RGC subtypes. Cell-autonomous generation of ROS during spiking contributes to tuning the spike patterns of RGCs.SIGNIFICANCE STATEMENT Energy production within retinal ganglion cells (RGCs) is accompanied by metabolic by-products harmful to cellular function. How these by-products modulate the excitability of RGCs bears heavily on visual function and the etiology of optic neuropathies. A novel hypothesis of how RGC metabolism can produce automodulation of electrical signaling was tested by identifying the characteristics and biophysical origins of changes to the excitability of RGCs caused by oxidizing by-products in the retina. This impacts our understanding of the pathophysiology of RGC dysfunction, supporting an emerging model in which increases in oxidizing chemical species during energy production, but not necessarily bioenergetic failure, lead to preferential degeneration of specific subtypes of RGCs, yielding loss of different aspects of visual capacity.


Subject(s)
Hydrogen Peroxide , Retinal Ganglion Cells , Mice , Female , Male , Animals , Reactive Oxygen Species , Retinal Ganglion Cells/physiology , Retina , Signal Transduction
2.
Front Cell Neurosci ; 14: 595064, 2020.
Article in English | MEDLINE | ID: mdl-33328894

ABSTRACT

How neurons in the eye feed signals back to photoreceptors to optimize sensitivity to patterns of light appears to be mediated by one or more unconventional mechanisms. Via these mechanisms, horizontal cells control photoreceptor synaptic gain and enhance key aspects of temporal and spatial center-surround receptive field antagonism. After the transduction of light energy into an electrical signal in photoreceptors, the next key task in visual processing is the transmission of an optimized signal to the follower neurons in the retina. For this to happen, the release of the excitatory neurotransmitter glutamate from photoreceptors is carefully regulated via horizontal cell feedback, which acts as a thermostat to keep the synaptic transmission in an optimal range during changes to light patterns and intensities. Novel findings of a recently described model that casts a classical neurotransmitter system together with ion transport mechanisms to adjust the alkaline milieu outside the synapse are reviewed. This novel inter-neuronal messaging system carries feedback signals using two separate, but interwoven regulated systems. The complex interplay between these two signaling modalities, creating synaptic modulation-at-a-distance, has obscured it's being defined. The foundations of our understanding of the feedback mechanism from horizontal cells to photoreceptors have been long established: Horizontal cells have broad receptive fields, suitable for providing surround inhibition, their membrane potential, a function of stimulus intensity and size, regulates inhibition of photoreceptor voltage-gated Ca2+ channels, and strong artificial pH buffering eliminates this action. This review compares and contrasts models of how these foundations are linked, focusing on a recent report in mammals that shows tonic horizontal cell release of GABA activating Cl- and HCO3 - permeable GABA autoreceptors. The membrane potential of horizontal cells provides the driving force for GABAR-mediated HCO3 - efflux, alkalinizing the cleft when horizontal cells are hyperpolarized by light or adding to their depolarization in darkness and contributing to cleft acidification via NHE-mediated H+ efflux. This model challenges interpretations of earlier studies that were considered to rule out a role for GABA in feedback to cones.

3.
Front Cell Neurosci ; 14: 600777, 2020.
Article in English | MEDLINE | ID: mdl-33335476

ABSTRACT

Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.

4.
Neuron ; 108(1): 111-127.e6, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32795398

ABSTRACT

Cajal recognized that the elaborate shape of neurons is fundamental to their function in the brain. However, there are no simple and generalizable genetic methods to study neuronal or glial cell morphology in the mammalian brain. Here, we describe four mouse lines conferring Cre-dependent sparse cell labeling based on mononucleotide repeat frameshift (MORF) as a stochastic translational switch. Notably, the optimized MORF3 mice, with a membrane-bound multivalent immunoreporter, confer Cre-dependent sparse and bright labeling of thousands of neurons, astrocytes, or microglia in each brain, revealing their intricate morphologies. MORF3 mice are compatible with imaging in tissue-cleared thick brain sections and with immuno-EM. An analysis of 151 MORF3-labeled developing retinal horizontal cells reveals novel morphological cell clusters and axonal maturation patterns. Our study demonstrates a conceptually novel, simple, generalizable, and scalable mouse genetic solution to sparsely label and illuminate the morphology of genetically defined neurons and glia in the mammalian brain.


Subject(s)
Astrocytes/ultrastructure , Brain/ultrastructure , Microglia/ultrastructure , Neurons/ultrastructure , Retinal Horizontal Cells/ultrastructure , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Frameshift Mutation/genetics , Green Fluorescent Proteins/genetics , Integrases , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Microsatellite Repeats/genetics , Neurons/metabolism , Neurons/pathology , Retinal Horizontal Cells/metabolism , Retinal Horizontal Cells/pathology
5.
Elife ; 92020 05 07.
Article in English | MEDLINE | ID: mdl-32378514

ABSTRACT

Structural changes in pre and postsynaptic neurons that accompany synapse formation often temporally and spatially overlap. Thus, it has been difficult to resolve which processes drive patterned connectivity. To overcome this, we use the laminated outer murine retina. We identify the serine/threonine kinase LKB1 as a key driver of synapse layer emergence. The absence of LKB1 in the retina caused a marked mislocalization and delay in synapse layer formation. In parallel, LKB1 modulated postsynaptic horizontal cell refinement and presynaptic photoreceptor axon growth. Mislocalized horizontal cell processes contacted aberrant cone axons in LKB1 mutants. These defects coincided with altered synapse protein organization, and horizontal cell neurites were misdirected to ectopic synapse protein regions. Together, these data suggest that LKB1 instructs the timing and location of connectivity in the outer retina via coordinate regulation of pre and postsynaptic neuron structure and the localization of synapse-associated proteins.


Subject(s)
Neurites/enzymology , Neurogenesis , Photoreceptor Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Synapses/enzymology , AMP-Activated Protein Kinases , Animals , Female , Male , Mice, Knockout , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Transport , Vesicular Glutamate Transport Protein 1/metabolism
6.
Neuron ; 103(1): 39-51.e5, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31122676

ABSTRACT

Despite robust effects on immature neurons, growth factors minimally promote axon regeneration in the adult central nervous system (CNS). Attempting to improve growth-factor responsiveness in mature neurons by dedifferentiation, we overexpressed Lin28 in the retina. Lin28-treated retinas responded to insulin-like growth factor-1 (IGF1) by initiating retinal ganglion cell (RGC) axon regeneration after axotomy. Surprisingly, this effect was cell non-autonomous. Lin28 expression was required only in amacrine cells, inhibitory neurons that innervate RGCs. Ultimately, we found that optic-nerve crush pathologically upregulated activity in amacrine cells, which reduced RGC electrical activity and suppressed growth-factor signaling. Silencing amacrine cells or pharmacologically blocking inhibitory neurotransmission also induced IGF1 competence. Remarkably, RGCs regenerating across these manipulations localized IGF1 receptor to their primary cilia, which maintained their signaling competence and regenerative ability. Thus, our results reveal a circuit-based mechanism that regulates CNS axon regeneration and implicate primary cilia as a regenerative signaling hub.


Subject(s)
Axons/physiology , Nerve Growth Factor/physiology , Nerve Regeneration/physiology , Receptors, Presynaptic/physiology , Amacrine Cells/physiology , Animals , Cilia/metabolism , Cilia/ultrastructure , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Nerve Crush , Optic Nerve Injuries/pathology , RNA-Binding Proteins/genetics , Receptor, IGF Type 1/metabolism , Retina/metabolism , Retinal Ganglion Cells/drug 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.
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
9.
J Neurosci ; 38(3): 723-732, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29217689

ABSTRACT

The vertebrate retina has the remarkable ability to support visual function under conditions of limited illumination, including the processing of signals evoked by single photons. Dim-light vision is regulated by several adaptive mechanisms. The mechanism explored in this study is responsible for increasing the light sensitivity and operational range of rod bipolar cells, the retinal neurons operating immediately downstream of rod photoreceptors. This sensitization is achieved through the sustained dopamine-dependent GABA release from other retinal neurons. Our goals were to identify the cell type responsible for the GABA release and the site of its modulation by dopamine. Previous studies have suggested the involvement of amacrine and/or horizontal cells. We now demonstrate, using mice of both sexes, that horizontal cells do not participate in this mechanism. Instead, sustained GABA input is provided by a subpopulation of wide-field amacrine cells, which stimulate the GABAC receptors at rod bipolar cell axons. We also found that dopamine does not act directly on either of these cells. Rather, it suppresses inhibition imposed on these wide-field cells by another subpopulation of upstream GABAergic amacrine cells, thereby sustaining the GABAC receptor activation required for rod bipolar cell sensitization.SIGNIFICANCE STATEMENT The vertebrate retina has an exquisite ability to adjust information processing to ever-changing conditions of ambient illumination, from bright sunlight to single-photon counting under dim starlight. Operation under each of these functional regimes requires an engagement of specific adaptation mechanisms. Here, we describe a mechanism optimizing the performance of the dim-light channel of vision, which consists of sensitizing rod bipolar cells by a sustained GABAergic input originating from a population of wide-field amacrine cells. Wide-field amacrine cells span large segments of the retina, making them uniquely equipped to normalize and optimize response sensitivity across distant receptive fields and preclude any bias toward local light-intensity fluctuations.


Subject(s)
Amacrine Cells/metabolism , Dopamine/metabolism , Retinal Bipolar Cells/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL
10.
J Physiol ; 595(13): 4449-4465, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28374528

ABSTRACT

KEY POINTS: Large conductance, Ca2+ -activated K+ (BKCa ) channels play important roles in mammalian retinal neurons, including photoreceptors, bipolar cells, amacrine cells and ganglion cells, but they have not been identified in horizontal cells. BKCa channel blockers paxilline and iberiotoxin, as well as Ca2+ free solutions and divalent cation Cav channel blockers, eliminate the outwardly rectifying current, while NS1619 enhances it. In symmetrical 150 mm K+ , single channels had a conductance close to 250 pS, within the range of all known BKCa channels. In current clamped horizontal cells, BKCa channels subdue depolarizing membrane potential excursions, reduce the average resting potential and decrease oscillations. The results show that BKCa channel activation puts a ceiling on horizontal cell depolarization and regulates the temporal responsivity of the cells. ABSTRACT: Large conductance, calcium-activated potassium (BKCa ) channels have numerous roles in neurons including the regulation of membrane excitability, intracellular [Ca2+ ] regulation, and neurotransmitter release. In the retina, they have been identified in photoreceptors, bipolar cells, amacrine cells and ganglion cells, but have not been conclusively identified in mammalian horizontal cells. We found that outward current recorded between -30 and +60 mV is carried primarily in BKCa channels in isolated horizontal cells of rats and mice. Whole-cell outward currents were maximal at +50 mV and declined at membrane potentials positive to this value. This current was eliminated by the selective BKCa channel blocker paxilline (100 nm), iberiotoxin (10 µm), Ca2+ free solutions and divalent cation Cav channel blockers. It was activated by the BKCa channel activator NS1619 (30 µm). Single channel recordings revealed the conductance of the channels to be 244 ± 11 pS (n = 17; symmetrical 150 mm K+ ) with open probability being both voltage- and Ca2+ -dependent. The channels showed fast activation kinetics in response to Ca2+ influx and inactivation gating that could be modified by intracellular protease treatment, which suggests ß subunit involvement. Under current clamp, block of BKCa current increased depolarizing membrane potential excursions, raising the average resting potential and producing oscillations. BKCa current activation with NS1619 inhibited oscillations and hyperpolarized the resting potential. These effects underscore the functional role of BKCa current in limiting depolarization of the horizontal cell membrane potential and suggest actions of these channels in regulating the temporal responsivity of the cells.


Subject(s)
Action Potentials , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Retinal Neurons/metabolism , Animals , Benzimidazoles , Cells, Cultured , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Retinal Neurons/physiology
11.
J Neurophysiol ; 116(2): 686-97, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27193322

ABSTRACT

Horizontal cells form the first laterally interacting network of inhibitory interneurons in the retina. Dopamine released onto horizontal cells under photic and circadian control modulates horizontal cell function. Using isolated, identified horizontal cells from a connexin-57-iCre × ROSA26-tdTomato transgenic mouse line, we investigated dopaminergic modulation of calcium channel currents (ICa) with whole cell patch-clamp techniques. Dopamine (10 µM) blocked 27% of steady-state ICa, an action blunted to 9% in the presence of the L-type Ca channel blocker verapamil (50 µM). The dopamine type 1 receptor (D1R) agonist SKF38393 (20 µM) inhibited ICa by 24%. The D1R antagonist SCH23390 (20 µM) reduced dopamine and SKF38393 inhibition. Dopamine slowed ICa activation, blocking ICa by 38% early in a voltage step. Enhanced early inhibition of ICa was eliminated by applying voltage prepulses to +120 mV for 100 ms, increasing ICa by 31% and 11% for early and steady-state currents, respectively. Voltage-dependent facilitation of ICa and block of dopamine inhibition after preincubation with a Gßγ-blocking peptide suggested involvement of Gßγ proteins in the D1R-mediated modulation. When the G protein activator guanosine 5'-O-(3-thiotriphosphate) (GTPγS) was added intracellularly, ICa was smaller and showed the same slowed kinetics seen during D1R activation. With GTPγS in the pipette, additional block of ICa by dopamine was only 6%. Strong depolarizing voltage prepulses restored the GTPγS-reduced early ICa amplitude by 36% and steady-state ICa amplitude by 3%. These results suggest that dopaminergic inhibition of ICa via D1Rs is primarily mediated through the action of Gßγ proteins in horizontal cells.


Subject(s)
Calcium Channels/physiology , Membrane Potentials/physiology , Receptors, Dopamine D1/metabolism , Retinal Horizontal Cells/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Connexins/genetics , Connexins/metabolism , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plant Lectins/genetics , Plant Lectins/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Retina/cytology , Retinal Horizontal Cells/drug effects , Spiperone/pharmacology , omega-Conotoxin GVIA/pharmacology
12.
eNeuro ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27022629

ABSTRACT

The cellular mechanisms underlying feedback signaling from horizontal cells to photoreceptors, which are important for the formation of receptive field surrounds of early visual neurons, remain unsettled. Mammalian horizontal cells express a complement of synaptic proteins that are necessary and sufficient for calcium-dependent exocytosis of inhibitory neurotransmitters at their contacts with photoreceptor terminals, suggesting that they are capable of releasing GABA via vesicular release. To test whether horizontal cell vesicular release is involved in feedback signaling, we perturbed inhibitory neurotransmission in these cells by targeted deletion of the vesicular GABA transporter (VGAT), the protein responsible for the uptake of inhibitory transmitter by synaptic vesicles. To manipulate horizontal cells selectively, an iCre mouse line with Cre recombinase expression controlled by connexin57 (Cx57) regulatory elements was generated. In Cx57-iCre mouse retina, only horizontal cells expressed Cre protein, and its expression occurred in all retinal regions. After crossing with a VGAT(flox/flox) mouse line, VGAT was selectively eliminated from horizontal cells, which was confirmed immunohistochemically. Voltage-gated ion channel currents in horizontal cells of Cx57-VGAT(-/-) mice were the same as Cx57-VGAT(+/+) controls, as were the cell responses to the ionotropic glutamate receptor agonist kainate, but the response to the GABAA receptor agonist muscimol in Cx57-VGAT(-/-) mice was larger. In contrast, the feedback inhibition of photoreceptor calcium channels, which in control animals is induced by horizontal cell depolarization, was completely absent in Cx57-VGAT(-/-) mice. The results suggest that vesicular release of GABA from horizontal cells is required for feedback inhibition of photoreceptors.


Subject(s)
Calcium Channels/metabolism , Feedback, Physiological/physiology , Photoreceptor Cells/metabolism , Retinal Horizontal Cells/physiology , Sequence Deletion/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/deficiency , Animals , Connexins/genetics , Connexins/metabolism , Excitatory Amino Acid Agonists/pharmacology , Feedback, Physiological/drug effects , Female , GABA-A Receptor Agonists/pharmacology , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Kainic Acid/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Muscimol/pharmacology , Retina/cytology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Visual Pathways/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
13.
Proc Natl Acad Sci U S A ; 112(41): 12840-5, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26420868

ABSTRACT

Neuronal output is modulated by inhibition onto both dendrites and axons. It is unknown whether inhibitory synapses at these two cellular compartments of an individual neuron are regulated coordinately or separately during in vivo development. Because neurotransmission influences synapse maturation and circuit development, we determined how loss of inhibition affects the expression of diverse types of inhibitory receptors on the axon and dendrites of mouse retinal bipolar cells. We found that axonal GABA but not glycine receptor expression depends on neurotransmission. Importantly, axonal and dendritic GABAA receptors comprise distinct subunit compositions that are regulated differentially by GABA release: Axonal GABAA receptors are down-regulated but dendritic receptors are up-regulated in the absence of inhibition. The homeostatic increase in GABAA receptors on bipolar cell dendrites is pathway-specific: Cone but not rod bipolar cell dendrites maintain an up-regulation of receptors in the transmission deficient mutants. Furthermore, the bipolar cell GABAA receptor alterations are a consequence of impaired vesicular GABA release from amacrine but not horizontal interneurons. Thus, inhibitory neurotransmission regulates in vivo postsynaptic maturation of inhibitory synapses with contrasting modes of action specific to synapse type and location.


Subject(s)
Axons/metabolism , Dendrites/metabolism , Receptors, GABA-A/metabolism , Retinal Bipolar Cells/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Dendrites/genetics , Mice , Mice, Transgenic , Receptors, GABA-A/genetics , Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Synapses/genetics
14.
J Physiol ; 591(13): 3309-24, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23613534

ABSTRACT

Horizontal cells send inhibitory feedback to photoreceptors, helping form antagonistic receptive fields in the retina, but the neurotransmitter and the mechanisms underlying this signalling are not known. Since the proteins responsible for conventional Ca(2+)-dependent release of GABAergic synaptic vesicles are present in mammalian horizontal cells, we investigated this conventional mechanism as the means by which horizontal cells inhibit photoreceptors. Using Ca(2+) imaging in rat retinal slices, we confirm that horizontal cell depolarization with kainate inhibits and horizontal cell hyperpolarization with NBQX disinhibits the Ca(2+) signals produced by pH-sensitive activation of voltage-gated calcium channels (Ca channels) in photoreceptors. We show that while 100 µm Co(2+) reduces photoreceptor Ca(2+) signals, it disinhibits them at 10 µm, an effect reminiscent of earlier studies where low [Co(2+)] eliminated feedback. The low [Co(2+)] disinhibition is pH sensitive. We localized L-, N- and P/Q-type Ca channels in rat horizontal cells, and showed that both the N-type Ca channel blocker -conotoxin GVIA and the P/Q-type Ca channel blocker -agatoxin IVA increased Ca(2+) signals in photoreceptors in a pH-sensitive manner. Pronounced actions of GABAergic agents on feedback signals to photoreceptors were observed, and are pH sensitive, but are inconsistent with direct inhibition by GABA of photoreceptor [Ca(2+)]. Patch-clamp studies revealed that GABA activates a conductance having high bicarbonate permeability in isolated horizontal cells, suggesting that the commonality of pH sensitivity throughout the results could arise from a GABA autofeedback action in horizontal cells. This could change cleft pH with concomitant inhibitory influences on photoreceptor Ca channels.


Subject(s)
Calcium Channels/physiology , Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , gamma-Aminobutyric Acid/physiology , Animals , Feedback, Physiological , Female , Hydrogen-Ion Concentration , In Vitro Techniques , Male , Membrane Potentials , Rats , Rats, Sprague-Dawley , Receptors, GABA/physiology
15.
J Comp Neurol ; 519(5): 972-88, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21280047

ABSTRACT

Horizontal cells mediate inhibitory feedforward and feedback lateral interactions in the outer retina at photoreceptor terminals and bipolar cell dendrites; however, the mechanisms that underlie synaptic transmission from mammalian horizontal cells are poorly understood. The localization of a vesicular γ-aminobutyric acid (GABA) transporter (VGAT) to horizontal cell processes in primate and rodent retinae suggested that mammalian horizontal cells release transmitter in a vesicular manner. Toward determining whether the molecular machinery for vesicular transmitter release is present in horizontal cells, we investigated the expression of SNAP25 (synaptosomal-associated protein of 25 kDa), a key SNARE protein, by immunocytochemistry with cell type-specific markers in the retinae of mouse, rat, rabbit, and monkey. Different commercial antibodies to SNAP25 were tested on vertical sections of retina. We report the robust expression of SNAP25 in both plexiform layers. Double labeling with SNAP25 and calbindin antibodies demonstrated that horizontal cell processes and their endings in photoreceptor triad synapses were strongly labeled for both proteins in mouse, rat, rabbit, and monkey retinae. Double labeling with parvalbumin antibodies in monkey retina verified SNAP25 immunoreactivity in all horizontal cells. Pre-embedding immunoelectron microscopy in rabbit retina confirmed expression of SNAP25 in lateral elements within photoreceptor triad synapses. The SNAP25 immunoreactivity in the plexiform layers and outer nuclear layer fell into at least three patterns depending on the antibody, suggesting a differential distribution of SNAP25 isoforms. The presence of SNAP25a and SNAP25b isoforms in mouse retina was established by reverse transcriptase-polymerase chain reaction. SNAP25 expression in mammalian horizontal cells along with other SNARE proteins is consistent with vesicular exocytosis.


Subject(s)
Protein Isoforms/metabolism , Retinal Horizontal Cells/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Biomarkers/metabolism , Calbindins , Humans , Immunohistochemistry , Macaca fascicularis , Mice , Mice, Inbred C57BL , Parvalbumins/metabolism , Protein Isoforms/genetics , Rabbits , Rats , Rats, Sprague-Dawley , Retinal Horizontal Cells/cytology , S100 Calcium Binding Protein G/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptosomal-Associated Protein 25/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
16.
Mol Vis ; 16: 1854-63, 2010 Sep 12.
Article in English | MEDLINE | ID: mdl-21031018

ABSTRACT

PURPOSE: The retina has the demanding task of encoding all aspects of the visual scene within the space of one fixation period lasting only a few hundred milliseconds. To accomplish this feat, information is encoded in specialized parallel channels and passed on to numerous central nuclei via the optic nerve. These parallel channels achieve specialization in at least three ways: the synaptic networks in which they participate, the neurotransmitter receptors expressed and the types and locations of ion channels or transporters used. Subcellular localization of receptors, channels and transporters is made yet more complex in the retina by the double duty many retinal processes serve. In the present work, we show that the protein Caspr (Contactin Associated Protein), best known for its critical role in the localization of voltage-gated ion channels at the nodes of Ranvier, is present in several types of retinal neurons including amacrine, bipolar, horizontal, and ganglion cells. METHODS: Using standard double label immunofluorescence protocols, we characterized the pattern of Caspr expression in the rodent retina. RESULTS: Caspr labeling was observed through much of the retina, including horizontal, bipolar, amacrine, and ganglion cells. Among amacrine cells, Caspr was observed in AII amacrine cells through co-localization with Parvalbumin and Disabled-1 in rat and mouse retinas, respectively. An additional amacrine cell type containing Calretinin also co-localized with Caspr, but did not co-localize with choline-acetyltransferase. Nearly all cells in the ganglion cell layer contain Caspr, including both displaced amacrine and ganglion cells. In the outer retina, Caspr was co-localized with PKC labeling in rod bipolar cell dendrites. In addition, Caspr labeling was found inside syntaxin-4 'sandwiches' in the outer plexiform layer, most likely indicating its presence in cone bipolar cell dendrites. Finally, Caspr was co-localized in segments of horizontal cell dendrites labeled with Calbindin-D28k. CONCLUSIONS: Caspr is best known for its role in organizing the localization of different voltage-gated ion channels in and around nodes of Ranvier. As neuronal processes in the retina often play a dual role involving both input and output, it is possible that the localization of Caspr in the retina will help us decipher the way retinal cells localize ion channels in their processes to increase computational capacity.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Mammals/metabolism , Retina/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Biomarkers/metabolism , Mice , Protein Transport , Rats , Retina/cytology , Staining and Labeling
17.
J Comp Neurol ; 518(10): 1647-69, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20235161

ABSTRACT

Gamma-aminobutyric acid (GABA) is likely expressed in horizontal cells of all species, although conflicting physiological findings have led to considerable controversy regarding its role as a transmitter in the outer retina. This study has evaluated key components of the GABA system in the outer retina of guinea pig, an emerging retinal model system. The presence of GABA, its rate-limiting synthetic enzyme glutamic acid decarboxylase (GAD(65) and GAD(67) isoforms), the plasma membrane GABA transporters (GAT-1 and GAT-3), and the vesicular GABA transporter (VGAT) was evaluated by using immunohistochemistry with well-characterized antibodies. The presence of GAD(65) mRNA was also evaluated by using laser capture microdissection and reverse transcriptase-polymerase chain reaction. Specific GABA, GAD(65), and VGAT immunostaining was localized to horizontal cell bodies, as well as to their processes and tips in the outer plexiform layer. Furthermore, immunostaining of retinal whole mounts and acutely dissociated retinas showed GAD(65) and VGAT immunoreactivity in both A-type and B-type horizontal cells. However, these cells did not contain GAD(67), GAT-1, or GAT-3 immunoreactivity. GAD(65) mRNA was detected in horizontal cells, and sequencing of the amplified GAD(65) fragment showed approximately 85% identity with other mammalian GAD(65) mRNAs. These studies demonstrate the presence of GABA, GAD(65), and VGAT in horizontal cells of the guinea pig retina, and support the idea that GABA is synthesized from GAD(65), taken up into synaptic vesicles by VGAT, and likely released by a vesicular mechanism from horizontal cells.


Subject(s)
Glutamate Decarboxylase/metabolism , Isoenzymes/metabolism , Retinal Horizontal Cells/enzymology , Retinal Horizontal Cells/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Base Sequence , Calbindins , Female , Glutamate Decarboxylase/genetics , Guinea Pigs , Humans , Immunohistochemistry , Isoenzymes/genetics , Male , Mice , Microdissection/methods , Molecular Sequence Data , Neuroglia/cytology , Neuroglia/metabolism , Retinal Horizontal Cells/cytology , S100 Calcium Binding Protein G/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics
18.
J Comp Neurol ; 512(1): 6-26, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18975268

ABSTRACT

Plasmalemmal and vesicular gamma-aminobutyric acid (GABA) transporters influence neurotransmission by regulating high-affinity GABA uptake and GABA release into the synaptic cleft and extracellular space. Postnatal expression of the plasmalemmal GABA transporter-1 (GAT-1), GAT-3, and the vesicular GABA/glycine transporter (VGAT) were evaluated in the developing mouse retina by using immunohistochemistry with affinity-purified antibodies. Weak transporter immunoreactivity was observed in the inner retina at postnatal day 0 (P0). GAT-1 immunostaining at P0 and at older ages was in amacrine and displaced amacrine cells in the inner nuclear layer (INL) and ganglion cell layer (GCL), respectively, and in their processes in the inner plexiform layer (IPL). At P10, weak GAT-1 immunostaining was in Müller cell processes. GAT-3 immunostaining at P0 and older ages was in amacrine cells and their processes, as well as in Müller cells and their processes that extended radially across the retina. At P10, Müller cell somata were observed in the middle of the INL. VGAT immunostaining was present at P0 and older ages in amacrine cells in the INL as well as processes in the IPL. At P5, weak VGAT immunostaining was also observed in horizontal cell somata and processes. By P15, the GAT and VGAT immunostaining patterns appear similar to the adult immunostaining patterns; they reached adult levels by about P20. These findings demonstrate that GABA uptake and release are initially established in the inner retina during the first postnatal week and that these systems subsequently mature in the outer retina during the second postnatal week.


Subject(s)
Cell Membrane/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Retina/growth & development , Retina/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Retina/cytology
19.
Vis Neurosci ; 24(4): 489-502, 2007.
Article in English | MEDLINE | ID: mdl-17640443

ABSTRACT

Horizontal cells mediate inhibitory feed-forward and feedback communication in the outer retina; however, mechanisms that underlie transmitter release from mammalian horizontal cells are poorly understood. Toward determining whether the molecular machinery for exocytosis is present in horizontal cells, we investigated the localization of syntaxin-4, a SNARE protein involved in targeting vesicles to the plasma membrane, in mouse, rat, and rabbit retinae using immunocytochemistry. We report robust expression of syntaxin-4 in the outer plexiform layer of all three species. Syntaxin-4 occurred in processes and tips of horizontal cells, with regularly spaced, thicker sandwich-like structures along the processes. Double labeling with syntaxin-4 and calbindin antibodies, a horizontal cell marker, demonstrated syntaxin-4 localization to horizontal cell processes; whereas, double labeling with PKC antibodies, a rod bipolar cell (RBC) marker, showed a lack of co-localization, with syntaxin-4 immunolabeling occurring just distal to RBC dendritic tips. Syntaxin-4 immunolabeling occurred within VGLUT-1-immunoreactive photoreceptor terminals and underneath synaptic ribbons, labeled by CtBP2/RIBEYE antibodies, consistent with localization in invaginating horizontal cell tips at photoreceptor triad synapses. Vertical sections of retina immunostained for syntaxin-4 and peanut agglutinin (PNA) established that the prominent patches of syntaxin-4 immunoreactivity were adjacent to the base of cone pedicles. Horizontal sections through the OPL indicate a one-to-one co-localization of syntaxin-4 densities at likely all cone pedicles, with syntaxin-4 immunoreactivity interdigitating with PNA labeling. Pre-embedding immuno-electron microscopy confirmed the subcellular localization of syntaxin-4 labeling to lateral elements at both rod and cone triad synapses. Finally, co-localization with SNAP-25, a possible binding partner of syntaxin-4, indicated co-expression of these SNARE proteins in the same subcellular compartment of the horizontal cell. Taken together, the strong expression of these two SNARE proteins in the processes and endings of horizontal cells at rod and cone terminals suggests that horizontal cell axons and dendrites are likely sites of exocytotic activity.


Subject(s)
Qa-SNARE Proteins/metabolism , Retinal Horizontal Cells/metabolism , Animals , Female , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Neurotransmitter Agents/metabolism , Photoreceptor Cells, Vertebrate/physiology , Rabbits , Rats , Rats, Sprague-Dawley , Retinal Bipolar Cells/metabolism , Subcellular Fractions/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Synaptosomal-Associated Protein 25/metabolism
20.
Vis Neurosci ; 23(1): 11-24, 2006.
Article in English | MEDLINE | ID: mdl-16597347

ABSTRACT

Glutamate release from photoreceptor terminals is controlled by voltage-dependent calcium channels (VDCCs). In humans, mutations in the Cacna1f gene, encoding the alpha1F subunit of VDCCs, underlie the incomplete form of X-linked congenital stationary night blindness (CSNB2). These mutations impair synaptic transmission from rod and cone photoreceptors to bipolar cells. Here, we report anatomical and functional characterizations of the retina in the nob2 (no b-wave 2) mouse, a naturally occurring mutant caused by a null mutation in Cacna1f. Not surprisingly, the b-waves of both the light- and dark-adapted electroretinogram are abnormal in nob2 mice. The outer plexiform layer (OPL) is disorganized, with extension of ectopic neurites through the outer nuclear layer that originate from rod bipolar and horizontal cells, but not from hyperpolarizing bipolar cells. These ectopic neurites continue to express mGluR6, which is frequently associated with profiles that label with the presynaptic marker Ribeye, indicating potential points of ectopic synapse formation. However, the morphology of the presynaptic Ribeye-positive profiles is abnormal. While cone pedicles are present their morphology also appears compromised. Characterizations of visual responses in retinal ganglion cells in vivo, under photopic conditions, demonstrate that ON-center cells have a reduced dynamic range, although their basic center-surround organization is retained; no alteration in the responses of OFF-center cells was evident. These results indicate that nob2 mice are a valuable model in which to explore the pathophysiological mechanisms associated with Cacna1f mutations causing CSNB2, and the subsequent effects on visual information processing. Further, the nob2 mouse represents a model system in which to define the signals that guide synapse formation and/or maintenance in the OPL.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Mutation , Retina/physiopathology , Retinal Ganglion Cells/physiology , Visual Pathways , Action Potentials/genetics , Age Factors , Alcohol Oxidoreductases , Animals , Calbindins , Calcium Channels, L-Type , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Dark Adaptation/physiology , Dose-Response Relationship, Radiation , Electroretinography/methods , Immunohistochemistry/methods , Mice , Mice, Mutant Strains , Peanut Agglutinin , Phosphoproteins/metabolism , Photic Stimulation/methods , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Reaction Time/physiology , Receptors, Metabotropic Glutamate/metabolism , Receptors, Neurokinin-3/metabolism , Retina/metabolism , Retina/pathology , Reverse Transcriptase Polymerase Chain Reaction/methods , S100 Calcium Binding Protein G/metabolism , Synapses/metabolism , Synapses/pathology , Time Factors , Visual Pathways/metabolism , Visual Pathways/pathology , Visual Pathways/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...