Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
J Neurochem ; 159(5): 840-856, 2021 12.
Article in English | MEDLINE | ID: mdl-34133756

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a cytoprotective protein for the retina. We hypothesize that this protein acts on neuronal survival and differentiation of photoreceptor cells in culture. The purpose of the present study was to evaluate the neurotrophic effects of PEDF and its fragments in an in vitro model of cultured primary retinal neurons that die spontaneously in the absence of trophic factors. We used Wistar albino rats. Cell death was assayed by immunofluorescence and flow cytometry through TUNEL assay, propidium iodide, mitotracker, and annexin V. Immunofluorescence of cells for visualizing rhodopsin, CRX, and antisyntaxin under confocal microscopy was performed. Neurite outgrowth was also quantified. Results show that PEDF protected photoreceptor precursors from apoptosis, preserved mitochondrial function and promoted polarization of opsin enhancing their developmental process, as well as induced neurite outgrowth in amacrine neurons. These effects were abolished by an inhibitor of the PEDF receptor or receptor-derived peptides that block ligand/receptor interactions. While all the activities were specifically conferred by short peptide fragments (17 amino acid residues) derived from the PEDF neurotrophic domain, no effects were triggered by peptides from the PEDF antiangiogenic region. The observed effects on retinal neurons imply a specific activation of the PEDF receptor by a small neurotrophic region of PEDF. Our findings support the neurotrophic PEDF peptides as neuronal guardians for the retina, highlighting their potential as promoters of retinal differentiation, and inhibitors of retinal cell death and its blinding consequences. Cover Image for this issue: https://doi.org/10.1111/jnc.15089.


Subject(s)
Amacrine Cells/drug effects , Cell Differentiation/drug effects , Eye Proteins/pharmacology , Nerve Growth Factors/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Serpins/pharmacology , Amacrine Cells/physiology , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Eye Proteins/genetics , Female , Male , Nerve Growth Factors/genetics , Neuronal Outgrowth/physiology , Neurons/physiology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Photoreceptor Cells, Vertebrate/physiology , Rats , Rats, Wistar , Serpins/genetics
2.
Cell Mol Neurobiol ; 41(2): 229-245, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32323153

ABSTRACT

Gap junctions are ubiquitous within the retina, but in general, it remains to be determined whether gap junction coupling between specific cell types is sufficiently strong to mediate functionally relevant coupling via electrical synapses. From ultrastructural, tracer coupling and immunolabeling studies, there is clear evidence for gap junctions between cone bipolar cells, but it is not known if these gap junctions function as electrical synapses. Here, using whole-cell voltage-clamp recording in rat (male and female) retinal slices, we investigated whether the gap junctions of bipolar cells make a measurable contribution to the membrane properties of these cells. We measured the input resistance (RN) of bipolar cells before and after applying meclofenamic acid (MFA) to block gap junctions. In the presence of MFA, RN of ON-cone bipolar cells displayed a clear increase, paralleled by block of the electrical coupling between these cells and AII amacrine cells in recordings of coupled cell pairs. For OFF-cone and rod bipolar cells, RN did not increase in the presence of MFA. The results for rod bipolar cells are consistent with the lack of gap junctions in these cells. However, for OFF-cone bipolar cells, our results suggest that the morphologically identified gap junctions between these cells do not support a junctional conductance that is sufficient to mediate effective electrical coupling. Instead, these junctions might play a role in chemical and/or metabolic coupling between subcellular compartments.


Subject(s)
Cell Membrane/metabolism , Gap Junctions/metabolism , Retinal Bipolar Cells/metabolism , Amacrine Cells/drug effects , Amacrine Cells/metabolism , Animals , Cell Membrane/drug effects , Electrophysiological Phenomena/drug effects , Female , Gap Junctions/drug effects , Male , Meclofenamic Acid/pharmacology , Rats , Retinal Bipolar Cells/drug effects , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism
3.
Int J Mol Sci ; 22(12)2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34200611

ABSTRACT

One of the causes of nervous system degeneration is an excess of glutamate released upon several diseases. Glutamate analogs, like N-methyl-DL-aspartate (NMDA) and kainic acid (KA), have been shown to induce experimental retinal neurotoxicity. Previous results have shown that NMDA/KA neurotoxicity induces significant changes in the full field electroretinogram response, a thinning on the inner retinal layers, and retinal ganglion cell death. However, not all types of retinal neurons experience the same degree of injury in response to the excitotoxic stimulus. The goal of the present work is to address the effect of intraocular injection of different doses of NMDA/KA on the structure and function of several types of retinal cells and their functionality. To globally analyze the effect of glutamate receptor activation in the retina after the intraocular injection of excitotoxic agents, a combination of histological, electrophysiological, and functional tools has been employed to assess the changes in the retinal structure and function. Retinal excitotoxicity caused by the intraocular injection of a mixture of NMDA/KA causes a harmful effect characterized by a great loss of bipolar, amacrine, and retinal ganglion cells, as well as the degeneration of the inner retina. This process leads to a loss of retinal cell functionality characterized by an impairment of light sensitivity and visual acuity, with a strong effect on the retinal OFF pathway. The structural and functional injury suffered by the retina suggests the importance of the glutamate receptors expressed by different types of retinal cells. The effect of glutamate agonists on the OFF pathway represents one of the main findings of the study, as the evaluation of the retinal lesions caused by excitotoxicity could be specifically explored using tests that evaluate the OFF pathway.


Subject(s)
Amacrine Cells/pathology , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/metabolism , N-Methylaspartate/analogs & derivatives , Retinal Ganglion Cells/pathology , Vision Disorders/pathology , Amacrine Cells/drug effects , Amacrine Cells/metabolism , Animals , Apoptosis , Mice , Mice, Inbred C57BL , N-Methylaspartate/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Vision Disorders/chemically induced , Vision Disorders/metabolism
4.
J Neurosci ; 39(4): 627-650, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30459218

ABSTRACT

In the rod pathway of the mammalian retina, axon terminals of glutamatergic rod bipolar cells are presynaptic to AII and A17 amacrine cells in the inner plexiform layer. Recent evidence suggests that both amacrines express NMDA receptors, raising questions concerning molecular composition, localization, activation, and function of these receptors. Using dual patch-clamp recording from synaptically connected rod bipolar and AII or A17 amacrine cells in retinal slices from female rats, we found no evidence that NMDA receptors contribute to postsynaptic currents evoked in either amacrine. Instead, NMDA receptors on both amacrine cells were activated by ambient glutamate, and blocking glutamate uptake increased their level of activation. NMDA receptor activation also increased the frequency of GABAergic postsynaptic currents in rod bipolar cells, suggesting that NMDA receptors can drive release of GABA from A17 amacrines. A striking dichotomy was revealed by pharmacological and immunolabeling experiments, which found GluN2B-containing NMDA receptors on AII amacrines and GluN2A-containing NMDA receptors on A17 amacrines. Immunolabeling also revealed a clustered organization of NMDA receptors on both amacrines and a close spatial association between GluN2B subunits and connexin 36 on AII amacrines, suggesting that NMDA receptor modulation of gap junction coupling between these cells involves the GluN2B subunit. Using multiphoton Ca2+ imaging, we verified that activation of NMDA receptors evoked an increase of intracellular Ca2+ in dendrites of both amacrines. Our results suggest that AII and A17 amacrines express clustered, extrasynaptic NMDA receptors, with different and complementary subunits that are likely to contribute differentially to signal processing and plasticity.SIGNIFICANCE STATEMENT Glutamate is the most important excitatory neurotransmitter in the CNS, but not all glutamate receptors transmit fast excitatory signals at synapses. NMDA-type glutamate receptors act as voltage- and ligand-gated ion channels, with functional properties determined by their specific subunit composition. These receptors can be found at both synaptic and extrasynaptic sites on neurons, but the role of extrasynaptic NMDA receptors is unclear. Here, we demonstrate that retinal AII and A17 amacrine cells, postsynaptic partners at rod bipolar dyad synapses, express extrasynaptic (but not synaptic) NMDA receptors, with different and complementary GluN2 subunits. The localization of GluN2A-containing receptors to A17s and GluN2B-containing receptors to AIIs suggests a mechanism for differential modulation of excitability and signaling in this retinal microcircuit.


Subject(s)
Amacrine Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Amacrine Cells/drug effects , Amacrine Cells/ultrastructure , Animals , Calcium/metabolism , Connexins/metabolism , Dendrites/metabolism , Excitatory Postsynaptic Potentials/drug effects , Female , Gap Junctions/drug effects , In Vitro Techniques , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Rod Photoreceptor Cells/ultrastructure , Signal Transduction/drug effects , gamma-Aminobutyric Acid/physiology , Gap Junction delta-2 Protein
5.
J Cell Mol Med ; 24(7): 4312-4323, 2020 04.
Article in English | MEDLINE | ID: mdl-32130787

ABSTRACT

Nitrite oxide plays an important role in the pathogenesis of various retinal diseases, especially when hypoxic processes are involved. This degeneration can be simulated by incubating porcine retinal explants with CoCl2 . Here, the therapeutic potential of iNOS-inhibitor 1400W was evaluated. Degeneration through CoCl2 and treatment with the 1400W were applied simultaneously to porcine retinae explants. Three groups were compared: control, CoCl2 , and CoCl2  + iNOS-inhibitor (1400W). At days 4 and 8, retinal ganglion cells (RGCs), bipolar, and amacrine cells were analysed. Furthermore, the influence on the glia cells and different stress markers were evaluated. Treatment with CoCl2 resulted in a significant loss of RGCs already after 4 days, which was counteracted by the iNOS-inhibitor. Expression of HIF-1α and its downstream targets confirmed the effective treatment with 1400W. After 8 days, the CoCl2 group displayed a significant loss in amacrine cells and also a drastic reduction in bipolar cells was observed, which was prevented by 1400W. The decrease in microglia could not be prevented by the inhibitor. CoCl2 induces strong degeneration in porcine retinae by mimicking hypoxia, damaging certain retinal cell types. Treatment with the iNOS-inhibitor counteracted these effects to some extent, by preventing loss of retinal ganglion and bipolar cells. Hence, this inhibitor seems to be a very promising treatment for retinal diseases.


Subject(s)
Amidines/pharmacology , Benzylamines/pharmacology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Retinal Diseases/drug therapy , Amacrine Cells/drug effects , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Disease Models, Animal , Humans , Microglia/drug effects , Microglia/pathology , Neuroprotection/drug effects , Nitric Oxide Synthase Type II/genetics , Organ Culture Techniques , Retina/drug effects , Retina/pathology , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Swine
6.
Int J Mol Sci ; 21(5)2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32106602

ABSTRACT

Combined administration of N-Methyl-D-Aspartate (NMDA) and kainic acid (KA) on the inner retina was studied as a model of excitotoxicity. The right eye of C57BL6J mice was injected with 1 µL of PBS containing NMDA 30 mM and KA 10 mM. Only PBS was injected in the left eye. One week after intraocular injection, electroretinogram recordings and immunohistochemistry were performed on both eyes. Retinal ganglion cell (RGC) projections were studied by fluorescent-cholerotoxin anterograde labeling. A clear decrease of the retinal "b" wave amplitude, both in scotopic and photopic conditions, was observed in the eyes injected with NMDA/KA. No significant effect on the "a" wave amplitude was observed, indicating the preservation of photoreceptors. Immunocytochemical labeling showed no effects on the outer nuclear layer, but a significant thinning on the inner retinal layers, thus indicating that NMDA and KA induce a deleterious effect on bipolar, amacrine and ganglion cells. Anterograde tracing of the visual pathway after NMDA and KA injection showed the absence of RGC projections to the contralateral superior colliculus and lateral geniculate nucleus. We conclude that glutamate receptor agonists, NMDA and KA, induce a deleterious effect of the inner retina when injected together into the vitreous chamber.


Subject(s)
Amacrine Cells/drug effects , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , N-Methylaspartate/toxicity , Photoreceptor Cells/drug effects , Retinal Ganglion Cells/drug effects , Amacrine Cells/pathology , Amacrine Cells/physiology , Animals , Cells, Cultured , Membrane Potentials , Mice , Mice, Inbred C57BL , Photoreceptor Cells/pathology , Photoreceptor Cells/physiology , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Visual Pathways/drug effects , Visual Pathways/pathology , Visual Pathways/physiology
7.
J Neurophysiol ; 121(4): 1232-1243, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30726156

ABSTRACT

During adaptation from dim to bright environments, changes in retinal signaling are mediated, in part, by dopamine. Dopamine is released with light and can modulate retinal receptive fields, neuronal coupling, inhibitory receptors, and rod pathway inhibition. However, it is unclear how dopamine affects inner retinal inhibition to cone bipolar cells, which relay visual information from photoreceptors to ganglion cells and are important signal processing sites. We tested the hypothesis that dopamine (D)1 receptor activation is sufficient to elicit light-adapted inhibitory changes. Local light-evoked inhibition and spontaneous activity were measured from OFF cone bipolar cells in dark-adapted mouse retinas while stimulating D1 receptors, which are located on bipolar, horizontal, and inhibitory amacrine cells. The D1 agonist SKF38393 reduced local inhibitory light-evoked response magnitude and increased response transience, which mimicked changes measured with light adaptation. D1-mediated reductions in local inhibition were more pronounced for glycinergic than GABAergic inputs, comparable with light adaptation. The effects of D1 receptors on light-evoked input were similar to the effects on spontaneous input. D1 receptor activation primarily decreased glycinergic spontaneous current frequency, similar to light adaptation, suggesting mainly a presynaptic amacrine cell site of action. These results expand the role of dopamine to include signal modulation of cone bipolar cell local inhibition. In this role, D1 receptor activation, acting primarily through glycinergic amacrine cells, may be an important mechanism for the light-adapted reduction in OFF bipolar cell inhibition since the actions are similar and dopamine is released during light adaptation. NEW & NOTEWORTHY Retinal adaptation to different luminance conditions requires the adjustment of local circuits for accurate signaling of visual scenes. Understanding mechanisms behind luminance adaptation at different retinal levels is important for understanding how the retina functions in a dynamic environment. In the mouse, we show that dopamine pathways reduce inner retinal inhibition similar to increased background luminance, suggesting the two are linked and highlighting a possible mechanism for light adaptation at an early retinal processing center.


Subject(s)
Adaptation, Physiological , Amacrine Cells/physiology , Contrast Sensitivity , Neural Inhibition , Receptors, Dopamine D1/metabolism , Retinal Cone Photoreceptor Cells/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Amacrine Cells/drug effects , Amacrine Cells/metabolism , Animals , Dopamine Agonists/pharmacology , Glycine/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Dopamine D1/agonists , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/physiology , Synaptic Transmission , Vision, Ocular , gamma-Aminobutyric Acid/metabolism
8.
Mol Vis ; 25: 780-790, 2019.
Article in English | MEDLINE | ID: mdl-31819340

ABSTRACT

Purpose: The neuromodulator dopamine plays an important role in light adaptation for the visual system. Light can stimulate dopamine release from dopaminergic amacrine cells (DACs) by activating three classes of photosensitive retinal cells: rods, cones, and melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). However, the synaptic mechanisms by which these photoreceptors excite DACs remain poorly understood. Our previous work demonstrated that α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors contribute to light regulation of DAC activity. AMPA receptors are classified into Ca2+-permeable and Ca2+-impermeable subtypes. We sought to identify which subtype of AMPA receptors is involved in light regulation of DAC activity. Methods: AMPA receptor-mediated light responses and miniature excitatory postsynaptic currents were recorded from genetically labeled DACs in mouse retinas with the whole-cell voltage-clamp mode. Immunostaining with antibodies against tyrosine hydroxylase, GluA2 (GluR2), and PSD-95 was performed in vertical retinal slices. Results: The biophysical and pharmacological data showed that only Ca2+-impermeable AMPA receptors contribute to DAC light responses driven by ipRGCs or cones (via depolarizing bipolar cells). We further found that the same subtype of AMPA receptors mediates miniature excitatory postsynaptic currents of DACs. These findings are supported by the immunohistochemical results demonstrating that DACs express the PSD-95 with GluA2, a subunit that is essential for determining the impermeability of AMPA receptors to calcium. Conclusions: The results indicated that GluA2-containing Ca2+-impermeable AMPA receptors contribute to signal transmission from photosensitive retinal cells to DACs.


Subject(s)
Amacrine Cells/metabolism , Calcium/metabolism , Cell Membrane Permeability , Dopamine/metabolism , Receptors, AMPA/metabolism , Amacrine Cells/drug effects , Animals , Benzodiazepines/pharmacology , Biophysical Phenomena , Cell Membrane Permeability/drug effects , Disks Large Homolog 4 Protein/metabolism , Excitatory Postsynaptic Potentials , Female , Light , Male , Mice, Inbred C57BL , Protein Subunits/metabolism , Receptors, AMPA/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism
9.
J Physiol ; 596(16): 3709-3724, 2018 08.
Article in English | MEDLINE | ID: mdl-29758086

ABSTRACT

KEY POINTS: Starburst amacrine cells release GABA and ACh. This study explores the coordinated function of starburst-mediated cholinergic excitation and GABAergic inhibition to bistratified retinal ganglion cells, predominantly direction-selective ganglion cells (DSGCs). In rat retina, under our recording conditions, starbursts were found to provide the major excitatory drive to a sub-population of ganglion cells whose dendrites co-stratify with starburst dendrites (putative DSGCs). In mouse retina, recordings from genetically identified DSGCs at physiological temperatures reveal that ACh inputs dominate the response to small spot-high contrast light stimuli, with preferential addition of bipolar cell input shifting the balance towards glutamate for larger spot stimuli In addition, starbursts also appear to gate glutamatergic excitation to DSGCs by postsynaptic and possibly presynaptic inhibitory processes ABSTRACT: Starburst amacrine cells release both GABA and ACh, allowing them to simultaneously mediate inhibition and excitation. However, the precise pre- and postsynaptic targets for ACh and GABA remain under intense investigation. Most previous studies have focused on starburst-mediated postsynaptic GABAergic inhibition and its role in the formation of directional selectivity in ganglion cells. However, the significance of postsynaptic cholinergic excitation is only beginning to be appreciated. Here, we found that light-evoked responses measured in bi-stratified rat ganglion cells with dendrites that co-fasciculate with ON and OFF starburst dendrites (putative direction-selective ganglion cells, DSGCs) were abolished by the application of nicotinic receptor antagonists, suggesting ACh could act as the primary source of excitation. Recording from genetically labelled DSGCs in mouse retina at physiological temperatures revealed that cholinergic synaptic inputs dominated the excitation for high contrast stimuli only when the size of the stimulus was small. Canonical glutamatergic inputs mediated by bipolar cells were prominent when GABA/glycine receptors were blocked or when larger spot stimuli were utilized. In mouse DSGCs, bipolar cell excitation could also be unmasked through the activation of mGluR2,3 receptors, which we show suppresses starburst output, suggesting that GABA from starbursts serves to inhibit bipolar cell signals in DSGCs. Taken together, these results suggest that starbursts amplify excitatory signals traversing the retina, endowing DSGCs with the ability to encode fine spatial information without compromising their ability to encode direction.


Subject(s)
Acetylcholine/pharmacology , Amacrine Cells/physiology , Glutamic Acid/metabolism , Retinal Ganglion Cells/physiology , Synapses/physiology , Visual Pathways/physiology , Amacrine Cells/cytology , Amacrine Cells/drug effects , Animals , Cells, Cultured , Cholinergic Agonists/pharmacology , Mice , Neural Inhibition , Photic Stimulation , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Synapses/drug effects , Synaptic Transmission , Visual Pathways/drug effects , gamma-Aminobutyric Acid/metabolism
10.
J Neurosci ; 36(13): 3871-86, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27030771

ABSTRACT

Retinal waves are correlated bursts of spontaneous activity whose spatiotemporal patterns are critical for early activity-dependent circuit elaboration and refinement in the mammalian visual system. Three separate developmental wave epochs or stages have been described, but the mechanism(s) of pattern generation of each and their distinct roles in visual circuit development remain incompletely understood. We used neuroanatomical,in vitroandin vivoelectrophysiological, and optical imaging techniques in genetically manipulated mice to examine the mechanisms of wave initiation and propagation and the role of wave patterns in visual circuit development. Through deletion of ß2 subunits of nicotinic acetylcholine receptors (ß2-nAChRs) selectively from starburst amacrine cells (SACs), we show that mutual excitation among SACs is critical for Stage II (cholinergic) retinal wave propagation, supporting models of wave initiation and pattern generation from within a single retinal cell type. We also demonstrate that ß2-nAChRs in SACs, and normal wave patterns, are necessary for eye-specific segregation. Finally, we show that Stage III (glutamatergic) retinal waves are not themselves necessary for normal eye-specific segregation, but elimination of both Stage II and Stage III retinal waves dramatically disrupts eye-specific segregation. This suggests that persistent Stage II retinal waves can adequately compensate for Stage III retinal wave loss during the development and refinement of eye-specific segregation. These experiments confirm key features of the "recurrent network" model for retinal wave propagation and clarify the roles of Stage II and Stage III retinal wave patterns in visual circuit development. SIGNIFICANCE STATEMENT: Spontaneous activity drives early mammalian circuit development, but the initiation and patterning of activity vary across development and among modalities. Cholinergic "retinal waves" are initiated in starburst amacrine cells and propagate to retinal ganglion cells and higher-order visual areas, but the mechanism responsible for creating their unique and critical activity pattern is incompletely understood. We demonstrate that cholinergic wave patterns are dictated by recurrent connectivity within starburst amacrine cells, and retinal ganglion cells act as "readouts" of patterned activity. We also show that eye-specific segregation occurs normally without glutamatergic waves, but elimination of both cholinergic and glutamatergic waves completely disrupts visual circuit development. These results suggest that each retinal wave pattern during development is optimized for concurrently refining multiple visual circuits.


Subject(s)
Action Potentials/physiology , Amacrine Cells/physiology , Gene Expression Regulation, Developmental/genetics , Retina/cytology , Visual Pathways/physiology , Action Potentials/drug effects , Age Factors , Amacrine Cells/drug effects , Animals , Animals, Newborn , Calcium/metabolism , Cholera Toxin/metabolism , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Patch-Clamp Techniques , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Retina/drug effects , Retina/growth & development , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Visual Pathways/drug effects
11.
J Neurophysiol ; 117(3): 1185-1199, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28053242

ABSTRACT

Retinal amacrine cells express nitric oxide (NO) synthase and produce NO, making NO available to regulate the function of amacrine cells. Here we test the hypothesis that NO can alter the GABAergic synaptic output of amacrine cells. We investigate this using whole cell voltage clamp recordings and Ca2+ imaging of cultured chick retinal amacrine cells. When recording from amacrine cells receiving synaptic input from other amacrine cells, we find that NO increases GABAergic spontaneous postsynaptic current (sPSC) frequency. This increase in sPSC frequency does not require the canonical NO receptor, soluble guanylate cyclase, or presynaptic action potentials. However, removal of extracellular Ca2+ and buffering of cytosolic Ca2+ both inhibit the response to NO. In Ca2+ imaging experiments, we confirm that NO increases cytosolic Ca2+ in amacrine cell processes by activating a Ca2+ influx pathway. Neither the increase in sPSC frequency nor the cytosolic Ca2+ elevations are dependent upon Ca2+ release from stores. NO also enhances evoked GABAergic responses. Because voltage-gated Ca2+ channel function is not altered by NO, the increased evoked response is likely due to the combined effect of voltage-dependent Ca2+ influx adding to the NO-dependent, voltage-independent, Ca2+ influx. Insight into the identity of the Ca2+ influx pathway is provided by the transient receptor potential canonical (TRPC) channel inhibitor clemizole, which prevents the NO-dependent increase in sPSC frequency and cytosolic Ca2+ elevations. These data suggest that NO production in the inner retina will enhance Ca2+-dependent GABA release from amacrine cells by activating TRPC channel(s).NEW & NOTEWORTHY Our research provides evidence that nitric oxide (NO) promotes GABAergic output from retinal amacrine cells by activating a likely transient receptor potential canonical-mediated Ca2+ influx pathway. This NO-dependent mechanism promoting GABA release can be voltage independent, suggesting that, in the retina, local NO production can bypass the formal retinal circuitry and increase local inhibition.


Subject(s)
Amacrine Cells/drug effects , Calcium Signaling/drug effects , Calcium/metabolism , Neurotransmitter Agents/pharmacology , Nitric Oxide/pharmacology , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Animals , Benzimidazoles/pharmacology , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Nitric Oxide Donors/pharmacology , Patch-Clamp Techniques , Retina/cytology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Synaptic Potentials/drug effects
12.
Nature ; 469(7330): 402-6, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21131947

ABSTRACT

Establishing precise synaptic connections is crucial to the development of functional neural circuits. The direction-selective circuit in the retina relies upon highly selective wiring of inhibitory inputs from starburst amacrine cells (SACs) onto four subtypes of ON-OFF direction-selective ganglion cells (DSGCs), each preferring motion in one of four cardinal directions. It has been reported in rabbit that the SACs on the 'null' sides of DSGCs form functional GABA (γ-aminobutyric acid)-mediated synapses, whereas those on the preferred sides do not. However, it is not known how the asymmetric wiring between SACs and DSGCs is established during development. Here we report that in transgenic mice with cell-type-specific labelling, the synaptic connections from SACs to DSGCs were of equal strength during the first postnatal week, regardless of whether the SAC was located on the preferred or null side of the DSGC. However, by the end of the second postnatal week, the strength of the synapses made from SACs on the null side of a DSGC significantly increased whereas those made from SACs located on the preferred side remained constant. Blocking retinal activity by intraocular injections of muscimol or gabazine during this period did not alter the development of direction selectivity. Hence, the asymmetric inhibition between the SACs and DSGCs is achieved by a developmental program that specifically strengthens the GABA-mediated inputs from SACs located on the null side, in a manner not dependent on neural activity.


Subject(s)
Models, Neurological , Neural Inhibition/physiology , Retina/physiology , Action Potentials/drug effects , Action Potentials/physiology , Amacrine Cells/drug effects , Amacrine Cells/physiology , Animals , Dendrites/physiology , Electric Conductivity , Mice , Mice, Transgenic , Motion , Motion Perception/drug effects , Motion Perception/physiology , Muscimol/pharmacology , Neural Inhibition/drug effects , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Photic Stimulation , Pyridazines/pharmacology , Retina/cytology , Retina/drug effects , Retina/growth & development , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Synapses/drug effects , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
13.
J Neurosci ; 35(48): 15955-70, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26631476

ABSTRACT

An inner retinal microcircuit composed of dopamine (DA)-containing amacrine cells and melanopsin-containing, intrinsically photosensitive retinal ganglion cells (M1 ipRGCs) process information about the duration and intensity of light exposures, mediating light adaptation, circadian entrainment, pupillary reflexes, and other aspects of non-image-forming vision. The neural interaction is reciprocal: M1 ipRGCs excite DA amacrine cells, and these, in turn, feed inhibition back onto M1 ipRGCs. We found that the neuropeptide somatostatin [somatotropin release inhibiting factor (SRIF)] also inhibits the intrinsic light response of M1 ipRGCs and postulated that, to tune the bidirectional interaction of M1 ipRGCs and DA amacrine cells, SRIF amacrine cells would provide inhibitory modulation to both cell types. SRIF amacrine cells, DA amacrine cells, and M1 ipRGCs form numerous contacts. DA amacrine cells and M1 ipRGCs express the SRIF receptor subtypes sst(2A) and sst4 respectively. SRIF modulation of the microcircuit was investigated with targeted patch-clamp recordings of DA amacrine cells in TH-RFP mice and M1 ipRGCs in OPN4-EGFP mice. SRIF increases K(+) currents, decreases Ca(2+) currents, and inhibits spike activity in both cell types, actions reproduced by the selective sst(2A) agonist L-054,264 (N-[(1R)-2-[[[(1S*,3R*)-3-(aminomethyl)cyclohexyl]methyl]amino]-1-(1H-indol-3-ylmethyl)-2-oxoethyl]spiro[1H-indene-1,4'-piperidine]-1'-carboxamide) in DA amacrine cells and the selective sst4 agonist L-803,087 (N(2)-[4-(5,7-difluoro-2-phenyl-1H-indol-3-yl)-1-oxobutyl]-L-arginine methyl ester trifluoroacetate) in M1 ipRGCs. These parallel actions of SRIF may serve to counteract the disinhibition of M1 ipRGCs caused by SRIF inhibition of DA amacrine cells. This allows the actions of SRIF on DA amacrine cells to proceed with adjusting retinal DA levels without destabilizing light responses by M1 ipRGCs, which project to non-image-forming targets in the brain.


Subject(s)
Amacrine Cells/physiology , Dopamine/metabolism , Neural Inhibition/physiology , Retina/cytology , Retinal Ganglion Cells/physiology , Visual Pathways/physiology , Amacrine Cells/drug effects , Amides/pharmacology , Animals , Calcium/metabolism , Excitatory Amino Acid Agents/pharmacology , GABA Agents/pharmacology , In Vitro Techniques , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Inhibition/drug effects , Neural Inhibition/genetics , Photic Stimulation , Piperidines/pharmacology , Rod Opsins/genetics , Rod Opsins/metabolism , Somatostatin/agonists , Somatostatin/antagonists & inhibitors , Somatostatin/metabolism
14.
J Neurosci ; 35(13): 5422-33, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834065

ABSTRACT

Diabetes leads to dysfunction of the neural retina before and independent of classical microvascular diabetic retinopathy, but previous studies have failed to demonstrate which neurons and circuits are affected at the earliest stages. Here, using patch-clamp recording and two-photon Ca(2+) imaging in rat retinal slices, we investigated diabetes-evoked changes in a microcircuit consisting of rod bipolar cells and their dyad postsynaptic targets, AII and A17 amacrine cells, which play an essential role in processing scotopic visual signals. AII amacrines forward their signals to ON- and OFF-cone bipolar cells and A17 amacrines provide GABAergic feedback inhibition to rod bipolar cells. Whereas Ca(2+)-permeable AMPA receptors mediate input from rod bipolar cells to both AII and A17 amacrines, diabetes changes the synaptic receptors on A17, but not AII amacrine cells. This was expressed as a change in pharmacological properties and single-channel conductance of the synaptic receptors, consistent with an upregulation of the AMPA receptor GluA2 subunit and reduced Ca(2+) permeability. In addition, two-photon imaging revealed reduced agonist-evoked influx of Ca(2+) in dendritic varicosities of A17 amacrine cells from diabetic compared with normal animals. Because Ca(2+)-permeable receptors in A17 amacrine cells mediate synaptic release of GABA, the reduced Ca(2+) permeability of these receptors in diabetic animals leads to reduced release of GABA, followed by disinhibition and increased release of glutamate from rod bipolar cells. This perturbation of neuron and microcircuit dynamics can explain the decreased dynamic range and sensitivity of scotopic vision that has been observed in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Neural Pathways/pathology , Retinal Rod Photoreceptor Cells/pathology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Amacrine Cells/drug effects , Amacrine Cells/metabolism , Animals , Calcium/metabolism , Diabetes Mellitus, Experimental/metabolism , Excitatory Postsynaptic Potentials , Female , Glutamic Acid/metabolism , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/biosynthesis , Receptors, AMPA/metabolism , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Up-Regulation , gamma-Aminobutyric Acid/metabolism
15.
J Neurosci ; 35(4): 1675-86, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25632142

ABSTRACT

Spontaneous retinal activity mediated by glutamatergic neurotransmission-so-called "Stage 3" retinal waves-drives anti-correlated spiking in ON and OFF RGCs during the second week of postnatal development of the mouse. In the mature retina, the activity of a retinal interneuron called the AII amacrine cell is responsible for anti-correlated spiking in ON and OFF α-RGCs. In mature AIIs, membrane hyperpolarization elicits bursting behavior. Here, we postulated that bursting in AIIs underlies the initiation of glutamatergic retinal waves. We tested this hypothesis by using two-photon calcium imaging of spontaneous activity in populations of retinal neurons and by making whole-cell recordings from individual AIIs and α-RGCs in in vitro preparations of mouse retina. We found that AIIs participated in retinal waves, and that their activity was correlated with that of ON α-RGCs and anti-correlated with that of OFF α-RGCs. Though immature AIIs lacked the complement of membrane conductances necessary to generate bursting, pharmacological activation of the M-current, a conductance that modulates bursting in mature AIIs, blocked retinal wave generation. Interestingly, blockade of the pacemaker conductance Ih, a conductance absent in AIIs but present in both ON and OFF cone bipolar cells, caused a dramatic loss of spatial coherence of spontaneous activity. We conclude that during glutamatergic waves, AIIs act to coordinate and propagate activity generated by BCs rather than to initiate spontaneous activity.


Subject(s)
Amacrine Cells/physiology , Glutamic Acid/metabolism , Retina/cytology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Amacrine Cells/drug effects , Animals , Animals, Newborn , Calcium/metabolism , Cdh1 Proteins/genetics , Excitatory Amino Acid Antagonists/pharmacology , Green Fluorescent Proteins/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Patch-Clamp Techniques , Quinoxalines/pharmacology , Retina/growth & development , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/physiology , SKP Cullin F-Box Protein Ligases/genetics , Visual Pathways/drug effects , Visual Pathways/physiology
16.
J Neurosci ; 35(39): 13336-50, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424882

ABSTRACT

Much of the computational power of the retina derives from the activity of amacrine cells, a large and diverse group of GABAergic and glycinergic inhibitory interneurons. Here, we identify an ON-type orientation-selective, wide-field, polyaxonal amacrine cell (PAC) in the rabbit retina and demonstrate how its orientation selectivity arises from the structure of the dendritic arbor and the pattern of excitatory and inhibitory inputs. Excitation from ON bipolar cells and inhibition arising from the OFF pathway converge to generate a quasi-linear integration of visual signals in the receptive field center. This serves to suppress responses to high spatial frequencies, thereby improving sensitivity to larger objects and enhancing orientation selectivity. Inhibition also regulates the magnitude and time course of excitatory inputs to this PAC through serial inhibitory connections onto the presynaptic terminals of ON bipolar cells. This presynaptic inhibition is driven by graded potentials within local microcircuits, similar in extent to the size of single bipolar cell receptive fields. Additional presynaptic inhibition is generated by spiking amacrine cells on a larger spatial scale covering several hundred microns. The orientation selectivity of this PAC may be a substrate for the inhibition that mediates orientation selectivity in some types of ganglion cells. Significance statement: The retina comprises numerous excitatory and inhibitory circuits that encode specific features in the visual scene, such as orientation, contrast, or motion. Here, we identify a wide-field inhibitory neuron that responds to visual stimuli of a particular orientation, a feature selectivity that is primarily due to the elongated shape of the dendritic arbor. Integration of convergent excitatory and inhibitory inputs from the ON and OFF visual pathways suppress responses to small objects and fine textures, thus enhancing selectivity for larger objects. Feedback inhibition regulates the strength and speed of excitation on both local and wide-field spatial scales. This study demonstrates how different synaptic inputs are regulated to tune a neuron to respond to specific features in the visual scene.


Subject(s)
Amacrine Cells/physiology , Axons/physiology , Orientation/physiology , Retina/physiology , Synapses/physiology , Amacrine Cells/drug effects , Animals , Axons/drug effects , Dendrites/drug effects , Dendrites/physiology , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , GABA Agents/pharmacology , Nerve Net/drug effects , Nerve Net/physiology , Orientation/drug effects , Patch-Clamp Techniques , Photic Stimulation , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Rabbits , Receptors, GABA/drug effects , Retina/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Synapses/drug effects , Visual Fields/drug effects , Visual Fields/physiology
17.
J Neurophysiol ; 115(1): 389-403, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26561610

ABSTRACT

At many glutamatergic synapses, non-N-methyl-d-aspartate (NMDA) and NMDA receptors are coexpressed postsynaptically. In the mammalian retina, glutamatergic rod bipolar cells are presynaptic to two rod amacrine cells (AII and A17) that constitute dyad postsynaptic partners opposite each presynaptic active zone. Whereas there is strong evidence for expression of non-NMDA receptors by both AII and A17 amacrines, the expression of NMDA receptors by the pre- and postsynaptic neurons in this microcircuit has not been resolved. In this study, using patch-clamp recording from visually identified cells in rat retinal slices, we investigated the expression and functional properties of NMDA receptors in these cells with a combination of pharmacological and biophysical methods. Pressure application of NMDA did not evoke a response in rod bipolar cells, but for both AII and A17 amacrines, NMDA evoked responses that were blocked by a competitive antagonist (CPP) applied extracellularly and an open channel blocker (MK-801) applied intracellularly. NMDA-evoked responses also displayed strong Mg(2+)-dependent voltage block and were independent of gap junction coupling. With low-frequency application (60-s intervals), NMDA-evoked responses remained stable for up to 50 min, but with higher-frequency stimulation (10- to 20-s intervals), NMDA responses were strongly and reversibly suppressed. We observed strong potentiation when NMDA was applied in nominally Ca(2+)-free extracellular solution, potentially reflecting Ca(2+)-dependent NMDA receptor inactivation. These results indicate that expression of functional (i.e., conductance-increasing) NMDA receptors is common to both AII and A17 amacrine cells and suggest that these receptors could play an important role for synaptic signaling, integration, or plasticity in the rod pathway.


Subject(s)
Amacrine Cells/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Retinal Rod Photoreceptor Cells/physiology , Visual Pathways/physiology , Amacrine Cells/cytology , Amacrine Cells/drug effects , Animals , Dizocilpine Maleate/pharmacology , Female , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Retinal Bipolar Cells/physiology , Retinal Rod Photoreceptor Cells/drug effects , Visual Pathways/cytology , Visual Pathways/drug effects
18.
Exp Eye Res ; 136: 45-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25989217

ABSTRACT

Cannabinoids have been suggested to protect retinal ganglion cells in different models of toxicity, but their effects on other retinal neurons are poorly known. We investigated the neuroprotective actions of the endocannabinoid N-arachidonoyl ethanolamine (Anandamide/AEA) and the synthetic cannabinoids R1-Methanandamide (MethAEA) and HU-210, in an in vivo retinal model of AMPA excitotoxicity, and the mechanisms involved in the neuroprotection. Sprague-Dawley rats were intravitreally injected with PBS or AMPA in the absence or presence of the cannabinoid agonists. Brain nitric oxide synthase (bNOS) and choline acetyltransferase (ChAT) immunoreactivity (IR), as well as TUNEL staining, assessed the AMPA-induced retinal amacrine cell loss and the dose-dependent neuroprotection afforded by cannabinoids. The CB1 receptor selective antagonist AM251 and the PI3K/Akt inhibitor wortmannin reversed the cannabinoid-induced neuroprotection, suggesting the involvement of CB1 receptors and the PI3K/Akt pathway in cannabinoids' actions. Experiments with the CB2 agonist JWH015 and [(3)H]CP55940 radioligand binding suggested that the CB2 receptor is not involved in the neuroprotection. AEA and HU-210 induced phosphorylation of Akt but only AEA induced phosphorylation of ERK1/2 kinases, as revealed by western blot analysis. To investigate the role of caspase-3 in the AMPA-induced cell death, the caspase-3 inhibitor Z-DEVD-FMK was co-injected with AMPA. Z-DEVD-FMK had no effect on AMPA excitotoxicity. Moreover, no difference was observed in the phosphorylation of SAPK/JNK kinases between PBS- and AMPA-treated retinas. These results suggest that endogenous and synthetic cannabinoids protect retinal amacrine neurons from AMPA excitotoxicity in vivo via a mechanism involving the CB1 receptors, and the PI3K/Akt and/or MEK/ERK1/2 signaling pathways.


Subject(s)
Amacrine Cells/drug effects , MAP Kinase Signaling System/physiology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Cannabinoid, CB1/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Apoptosis , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Endocannabinoids/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/toxicity , Fluorescent Antibody Technique, Indirect , In Situ Nick-End Labeling , Intravitreal Injections , Male , Nitric Oxide Synthase Type I/metabolism , Phosphoinositide-3 Kinase Inhibitors , Polyunsaturated Alkamides/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors
19.
Exp Eye Res ; 135: 174-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25752698

ABSTRACT

Retinal ischemia is a common risk factor for visual impairment and blindness. Two common changes after retinal ischemia are retinal ganglion cell (RGC) loss and Müller glial cell (MGC)-mediated endogenous repair. Matrix metalloproteinase 9 (MMP-9) has been shown to be responsible to RGC death. However, the effects of MMP-9 on the loss of other neurons and the reactivity of MGCs after retinal injury remain unclear. Ouabain, a Na/K-ATPase inhibitor, was injected into the vitreous body of rat eyes to induce cell death in the inner nuclear layer (INL). MMP-9 expression and activation in the retinas were examined by gelatin zymography and immunohistochemistry. The role of MMP-9 inhibitor (MMP-9i) in ouabain-treated retinas was assessed. After ouabain injection, there was an upregulation of MMP-9 activity in the inner retinas, and the activation of MMP-9 reached a maximum at 2 day. Unexpectedly, MMP-9i enhanced the thinning of the INL, the loss of Calbindin D-28k-positive cells and Syntaxin-positive amacrine cells (ACs) in the INL and decreased levels of Calbindin D-28k protein, while leaving the outer nuclear layer (ONL) unchanged. In addition, MMP-9i led to a minor increase in the number of BrdU positive cells that did not express GS in the INL. Collectively, these results revealed that the inhibition of MMP-9 activity facilitated AC loss and promoted the generation of MGC-derived cells in ouabain-treated retinas, which indicates that treating retinal diseases with drugs that inhibit MMP-9 activity should be considered with caution.


Subject(s)
Amacrine Cells/pathology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Ouabain/pharmacology , Retina/drug effects , Retinal Degeneration , Amacrine Cells/drug effects , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Ischemia/complications , Matrix Metalloproteinase 9/physiology , Neuroglia/drug effects , Rats , Rats, Sprague-Dawley , Retina/enzymology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Vessels
20.
J Physiol ; 592(1): 49-65, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24042496

ABSTRACT

The centre-surround organisation of receptive fields is a feature of most retinal ganglion cells (RGCs) and is critical for spatial discrimination and contrast detection. Although lateral inhibitory processes are known to be important in generating the receptive field surround, the contribution of each of the two synaptic layers in the primate retina remains unclear. Here we studied the spatial organisation of excitatory and inhibitory synaptic inputs onto ON and OFF ganglion cells in the primate retina. All RGCs showed an increase in excitation in response to stimulus of preferred polarity. Inhibition onto RGCs comprised two types of responses to preferred polarity: some RGCs showed an increase in inhibition whilst others showed removal of tonic inhibition. Excitatory inputs were strongly spatially tuned but inhibitory inputs showed more variable organisation: in some neurons they were as strongly tuned as excitation, and in others inhibitory inputs showed no spatial tuning. We targeted one source of inner retinal inhibition by functionally ablating spiking amacrine cells with bath application of tetrodotoxin (TTX). TTX significantly reduced the spatial tuning of excitatory inputs. In addition, TTX reduced inhibition onto those RGCs where a stimulus of preferred polarity increased inhibition. Reconstruction of the spatial tuning properties by somatic injection of excitatory and inhibitory synaptic conductances verified that TTX-mediated inhibition onto bipolar cells increases the strength of the surround in RGC spiking output. These results indicate that in the primate retina inhibitory mechanisms in the inner plexiform layer sharpen the spatial tuning of ganglion cells.


Subject(s)
Inhibitory Postsynaptic Potentials , Retinal Ganglion Cells/physiology , Visual Fields , Amacrine Cells/drug effects , Amacrine Cells/physiology , Animals , Callithrix , Excitatory Postsynaptic Potentials , Female , Male , Retinal Ganglion Cells/drug effects , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL