Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Open Biol ; 14(7): 240140, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39079673

ABSTRACT

In the central nervous system of vertebrates, glutamate serves as the primary excitatory neurotransmitter. However, in the retina, glutamate released from photoreceptors causes hyperpolarization in post-synaptic ON-bipolar cells through a glutamate-gated chloride current, which seems paradoxical. Our research reveals that this current is modulated by two excitatory glutamate transporters, EAAT5b and EAAT7. In the zebrafish retina, these transporters are located at the dendritic tips of ON-bipolar cells and interact with all four types of cone photoreceptors. The absence of these transporters leads to a decrease in ON-bipolar cell responses, with eaat5b mutants being less severely affected than eaat5b/eaat7 double mutants, which also exhibit altered response kinetics. Biophysical investigations establish that EAAT7 is an active glutamate transporter with a predominant anion conductance. Our study is the first to demonstrate the direct involvement of post-synaptic glutamate transporters in inhibitory direct synaptic transmission at a central nervous system synapse.


Subject(s)
Amino Acid Transport System X-AG , Retina , Synaptic Transmission , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/metabolism , Retina/metabolism , Amino Acid Transport System X-AG/metabolism , Amino Acid Transport System X-AG/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Glutamic Acid/metabolism , Mutation , Retinal Bipolar Cells/metabolism
2.
Adv Exp Med Biol ; 775: 69-83, 2013.
Article in English | MEDLINE | ID: mdl-23392925

ABSTRACT

Retinal ganglion cells (RGCs) are spiking neurons, which send visual information to the brain, through the optic nerve. RGC degeneration occurs in retinal diseases, either as a primary process or secondary to photoreceptor loss. Mechanisms involved in this neuronal degeneration are still unclear and no drugs directly targeting RGC neuroprotection are yet available. Here, we show that taurine is one factor involved in preserving the RGC survival. Indeed, a taurine depletion induced by the antiepileptic drug, vigabatrin, was incriminated in its retinal toxicity leading to the RGC loss. Similarly, we showed that RGC degeneration can be induced by pharmacologically blocking the taurine-transporter with the chronic administration of a selective inhibitor, which results in a decrease in the taurine levels both in the plasma and in the retinal tissue. Finally, we found that taurine can directly prevent RGC degeneration, occurring either in serum-deprived pure RGC cultures or in animal models presenting an RGC loss (glaucomatous rats and the P23H rats, a model for retinitis pigmentosa). These data suggest that the retinal taurine level is a crucial marker to prevent RGC damage in major retinal diseases.


Subject(s)
Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Taurine/pharmacology , Animals , Cell Survival/drug effects , Disease Models, Animal , Glaucoma/complications , Glaucoma/drug therapy , Glaucoma/pathology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Neuroprotective Agents/therapeutic use , Rats , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/pathology , Taurine/analogs & derivatives , Taurine/therapeutic use , Time Factors , Vigabatrin/administration & dosage , Vigabatrin/pharmacology
3.
Eur J Neurosci ; 27(10): 2575-86, 2008 May.
Article in English | MEDLINE | ID: mdl-18547244

ABSTRACT

At the photoreceptor ribbon synapse, glutamate released from vesicles at different positions along the ribbon reaches the same postsynaptic receptors. Thus, vesicles may not exert entirely independent effects. We examined whether responses of salamander retinal horizontal cells evoked by light or direct depolarization during paired recordings could be predicted by summation of individual miniature excitatory postsynaptic currents (mEPSCs). For EPSCs evoked by depolarization of rods or cones, linear convolution of mEPSCs with photoreceptor release functions predicted EPSC waveforms and changes caused by inhibiting glutamate receptor desensitization. A low-affinity glutamate antagonist, kynurenic acid (KynA), preferentially reduced later components of rod-driven EPSCs, suggesting lower levels of glutamate are present during the later sustained component of the EPSC. A glutamate-scavenging enzyme, glutamic-pyruvic transaminase, did not inhibit mEPSCs or the initial component of rod-driven EPSCs, but reduced later components of the EPSC. Inhibiting glutamate uptake with a low concentration of DL-threo-beta-benzoyloxyaspartate (TBOA) also did not alter mEPSCs or the initial component of rod-driven EPSCs, but enhanced later components of the EPSC. Low concentrations of TBOA and KynA did not affect the kinetics of fast cone-driven EPSCs. Under both rod- and cone-dominated conditions, light-evoked currents (LECs) were enhanced considerably by TBOA. LECs were more strongly inhibited than EPSCs by KynA, suggesting the presence of lower glutamate levels. Collectively, these results indicate that the initial EPSC component can be largely predicted from a linear sum of individual mEPSCs, but with sustained release, residual amounts of glutamate from multiple vesicles pool together, influencing LECs and later components of EPSCs.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Alanine Transaminase/metabolism , Alanine Transaminase/pharmacology , Ambystoma , Animals , Aspartic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Light , Organ Culture Techniques , Patch-Clamp Techniques , Photic Stimulation , Photoreceptor Cells/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/physiology , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Horizontal Cells/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Vision, Ocular/drug effects , Vision, Ocular/physiology
4.
J Neurosci ; 26(9): 2555-63, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510733

ABSTRACT

Synaptic depression produced by repetitive stimulation is likely to be particularly important in shaping responses of second-order retinal neurons at the tonically active photoreceptor synapse. We analyzed the time course and mechanisms of synaptic depression at rod and cone synapses using paired-pulse protocols involving two complementary measurements of exocytosis: (1) paired whole-cell recordings of the postsynaptic current (PSC) in second-order retinal neurons and (2) capacitance measurements of vesicular membrane fusion in rods and cones. PSCs in ON bipolar, OFF bipolar, and horizontal cells evoked by stimulation of either rods or cones recovered from paired-pulse depression (PPD) at rates similar to the recovery of exocytotic capacitance changes in rods and cones. Correlation between presynaptic and postsynaptic measures of recovery from PPD suggests that 80-90% of the depression at these synapses is presynaptic in origin. Consistent with a predominantly presynaptic mechanism, inhibiting desensitization of postsynaptic glutamate receptors had little effect on PPD. The depression of exocytotic capacitance changes exceeded depression of the presynaptic calcium current, suggesting that it is primarily caused by a depletion of synaptic vesicles. In support of this idea, limiting Ca2+ influx by using weaker depolarizing stimuli promoted faster recovery from PPD. Although cones exhibit much faster exocytotic kinetics than rods, exocytotic capacitance changes recovered from PPD at similar rates in both cell types. Thus, depression of release is not likely to contribute to differences in the kinetics of transmission from rods and cones.


Subject(s)
Electric Stimulation , Neural Inhibition/radiation effects , Photoreceptor Cells/physiology , Synapses/physiology , Animals , Benzothiadiazines/pharmacology , Dose-Response Relationship, Radiation , Electric Capacitance , Exocytosis/drug effects , Exocytosis/physiology , Glutamates/pharmacology , In Vitro Techniques , Indoles/pharmacology , Patch-Clamp Techniques/methods , Photoreceptor Cells/radiation effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Presynaptic Terminals/radiation effects , Retinal Cone Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Rod Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/radiation effects , Synapses/radiation effects , Time Factors , Urodela , Visual Pathways/physiology , Visual Pathways/radiation effects
5.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28612046

ABSTRACT

Photoreceptor ribbon synapses tonically release glutamate. To ensure efficient signal transmission and prevent glutamate toxicity, a highly efficient glutamate removal system provided by members of the SLC1 gene family is required. By using a combination of biophysical and in vivo studies, we elucidate the role of excitatory amino acid transporter 2 (EAAT2) proteins in synaptic glutamate homeostasis at the zebrafish photoreceptor synapse. The main glutamate sink is provided by the glial EAAT2a, reflected by reduced electroretinographic responses in EAAT2a-depleted larvae. EAAT2b is located on the tips of cone pedicles and contributes little to glutamate reuptake. However, this transporter displays both a large chloride conductance and leak current, being important in stabilizing the cone resting potential. This work demonstrates not only how proteins originating from the same gene family can complement each other's expression profiles and biophysical properties, but also how presynaptic and glial transporters are coordinated to ensure efficient synaptic transmission at glutamatergic synapses of the central nervous system.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Photoreceptor Cells/classification , Photoreceptor Cells/metabolism , Presynaptic Terminals/metabolism , Animals , Animals, Genetically Modified , Excitatory Amino Acid Transporter 2/genetics , Eye/cytology , Gene Expression Regulation/genetics , Glutamic Acid/metabolism , In Vitro Techniques , Larva , Morpholinos/genetics , Morpholinos/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Retina/anatomy & histology , Signal Transduction/drug effects , Signal Transduction/genetics , Synapses , Synaptic Transmission/physiology , Visual Pathways/physiology , Xenopus , Zebrafish
6.
J Neurosci ; 25(18): 4633-40, 2005 May 04.
Article in English | MEDLINE | ID: mdl-15872111

ABSTRACT

Cone-driven responses of second-order retinal neurons are considerably faster than rod-driven responses. We examined whether differences in the kinetics of synaptic transmitter release from rods and cones may contribute to differences in postsynaptic response kinetics. Exocytosis from rods and cones was triggered by membrane depolarization and monitored in two ways: (1) by measuring EPSCs evoked in second-order neurons by depolarizing steps applied to presynaptic rods or cones during simultaneous paired whole-cell recordings or (2) by direct measurements of exocytotic increases in membrane capacitance. The kinetics of release was assessed by varying the length of the depolarizing test step. Both measures of release revealed two kinetic components to the increase in exocytosis as a function of the duration of a step depolarization. In addition to slow sustained components in both cell types, the initial fast component of exocytosis had a time constant of <5 ms in cones, >10-fold faster than that of rods. Rod/cone differences in the kinetics of release were substantiated by a linear correlation between depolarization-evoked capacitance increases and EPSC charge transfer. Experiments on isolated rods indicate that the slower kinetics of exocytosis from rods was not a result of rod-rod coupling. The initial rapid release of vesicles from cones can shape the postsynaptic response and may contribute to the faster responses of cone-driven cells observed at light offset.


Subject(s)
Exocytosis/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Synaptic Transmission/physiology , Animals , Benzothiadiazines/pharmacology , Dose-Response Relationship, Radiation , Electric Capacitance , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , In Vitro Techniques , Kinetics , Patch-Clamp Techniques/methods , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/radiation effects , Salamandra , Synaptic Transmission/drug effects , Time Factors
7.
Prog Retin Eye Res ; 41: 44-63, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24721186

ABSTRACT

Taurine is the most abundant amino acid in the retina. In the 1970s, it was thought to be involved in retinal diseases with photoreceptor degeneration, because cats on a taurine-free diet presented photoreceptor loss. However, with the exception of its introduction into baby milk and parenteral nutrition, taurine has not yet been incorporated into any commercial treatment with the aim of slowing photoreceptor degeneration. Our recent discovery that taurine depletion is involved in the retinal toxicity of the antiepileptic drug vigabatrin has returned taurine to the limelight in the field of neuroprotection. However, although the retinal toxicity of vigabatrin principally involves a deleterious effect on photoreceptors, retinal ganglion cells (RGCs) are also affected. These findings led us to investigate the possible role of taurine depletion in retinal diseases with RGC degeneration, such as glaucoma and diabetic retinopathy. The major antioxidant properties of taurine may influence disease processes. In addition, the efficacy of taurine is dependent on its uptake into retinal cells, microvascular endothelial cells and the retinal pigment epithelium. Disturbances of retinal vascular perfusion in these retinal diseases may therefore affect the retinal uptake of taurine, resulting in local depletion. The low plasma taurine concentrations observed in diabetic patients may further enhance such local decreases in taurine concentration. We here review the evidence for a role of taurine in retinal ganglion cell survival and studies suggesting that this compound may be involved in the pathophysiology of glaucoma or diabetic retinopathy. Along with other antioxidant molecules, taurine should therefore be seriously reconsidered as a potential treatment for such retinal diseases.


Subject(s)
Retinal Degeneration/prevention & control , Taurine/physiology , Animals , Humans , Neuroprotective Agents/therapeutic use , Retinal Degeneration/physiopathology , Taurine/chemistry , Taurine/therapeutic use
8.
PLoS One ; 7(10): e42017, 2012.
Article in English | MEDLINE | ID: mdl-23115615

ABSTRACT

Retinal ganglion cell (RGC) degeneration occurs in numerous retinal diseases leading to blindness, either as a primary process like in glaucoma, or secondary to photoreceptor loss. However, no commercial drug is yet directly targeting RGCs for their neuroprotection. In the 70s, taurine, a small sulfonic acid provided by nutrition, was found to be essential for the survival of photoreceptors, but this dependence was not related to any retinal disease. More recently, taurine deprivation was incriminated in the retinal toxicity of an antiepileptic drug. We demonstrate here that taurine can improve RGC survival in culture or in different animal models of RGC degeneration. Taurine effect on RGC survival was assessed in vitro on primary pure RCG cultures under serum-deprivation conditions, and on NMDA-treated retinal explants from adult rats. In vivo, taurine was administered through the drinking water in two glaucomatous animal models (DBA/2J mice and rats with vein occlusion) and in a model of Retinitis pigmentosa with secondary RGC degeneration (P23H rats). After a 6-day incubation, 1 mM taurine significantly enhanced RGCs survival (+68%), whereas control RGCs were cultured in a taurine-free medium, containing all natural amino-acids. This effect was found to rely on taurine-uptake by RGCs. Furthermore taurine (1 mM) partly prevented NMDA-induced RGC excitotoxicity. Finally, taurine supplementation increased RGC densities both in DBA/2J mice, in rats with vein occlusion and in P23H rats by contrast to controls drinking taurine-free water. This study indicates that enriched taurine nutrition can directly promote RGC survival through RGC intracellular pathways. It provides evidence that taurine can positively interfere with retinal degenerative diseases.


Subject(s)
Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Taurine/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Mice , Mice, Inbred DBA , N-Methylaspartate/pharmacology , Photoreceptor Cells, Vertebrate/metabolism , Retinal Ganglion Cells/cytology
9.
J Neurophysiol ; 95(3): 1992-5, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16371457

ABSTRACT

Horizontal cell (HC) to cone feedback helps establish the center-surround arrangement of visual receptive fields. It has been shown that HC activity influences cone synaptic output by altering the amplitude and voltage dependence of the calcium current (ICa) in cones. In this study, we obtained voltage-clamp recordings simultaneously from cones and HCs to directly control the membrane potential of HCs and thereby measure the influence of HC membrane potential changes on ICa in adjacent cones. Directly hyperpolarizing voltage clamped HCs produced a negative activation shift and increased the amplitude of ICa in cones. Both of these effects were abolished by enhancing extracellular pH buffering capacity with HEPES. In contrast, addition of the gap junction blocker, carbenoxolone, did not significantly alter the shifts or amplitude changes in cone ICa produced by changes in HC membrane potential. These results support the hypothesis that changes in the HC membrane potential alter the voltage dependence and amplitude of cone ICa by altering extracellular pH levels at the synapse.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Membrane Potentials/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Feedback/physiology , Hydrogen-Ion Concentration , Patch-Clamp Techniques , Urodela
10.
Eur J Neurosci ; 23(11): 2983-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16819987

ABSTRACT

We examined the contribution of calcium-induced calcium release (CICR) to synaptic transmission from rod photoreceptor terminals. Whole-cell recording and confocal calcium imaging experiments were conducted on rods with intact synaptic terminals in a retinal slice preparation from salamander. Low concentrations of ryanodine stimulated calcium increases in rod terminals, consistent with the presence of ryanodine receptors. Application of strong depolarizing steps (-70 to -10 mV) exceeding 200 ms or longer in duration evoked a wave of calcium that spread across the synaptic terminals of voltage-clamped rods. This secondary calcium increase was blocked by high concentrations of ryanodine, indicating it was due to CICR. Ryanodine (50 microm) had no significant effect on rod calcium current (I(ca)) although it slightly diminished rod light-evoked voltage responses. Bath application of 50 microm ryanodine strongly inhibited light-evoked currents in horizontal cells. Whether applied extracellularly or delivered into the rod cell through the patch pipette, ryanodine (50 microm) also inhibited excitatory post-synaptic currents (EPSCs) evoked in horizontal cells by depolarizing steps applied to rods. Ryanodine caused a preferential reduction in the later portions of EPSCs evoked by depolarizing steps of 200 ms or longer. These results indicate that CICR enhances calcium increases in rod terminals evoked by sustained depolarization, which in turn acts to boost synaptic exocytosis from rods.


Subject(s)
Calcium/metabolism , Calcium/pharmacology , Presynaptic Terminals/drug effects , Retinal Rod Photoreceptor Cells/cytology , Synaptic Transmission/drug effects , Animals , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , In Vitro Techniques , Light , Patch-Clamp Techniques/methods , Presynaptic Terminals/physiology , Presynaptic Terminals/radiation effects , Retina/cytology , Ryanodine/pharmacology , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , Time Factors , Urodela
11.
J Physiol ; 569(Pt 3): 773-88, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16223761

ABSTRACT

Synaptic transmission from cones is faster than transmission from rods. Using paired simultaneous recordings from photoreceptors and second-order neurones in the salamander retina, we studied the contributions of rod-cone differences in glutamate receptor properties and synaptic release rates to shaping postsynaptic responses. Depolarizing steps evoked sustained calcium currents in rods and cones that in turn produced transient excitatory postsynaptic currents (EPSCs) in horizontal and OFF bipolar cells. Cone-driven EPSCs rose and decayed faster than rod-driven EPSCs, even when comparing inputs from a rod and cone onto the same postsynaptic neurone. Thus, rod-cone differences in EPSCs reflect properties of individual rod and cone synapses. Experiments with selective AMPA and KA agonists and antagonists showed that rods and cones both contact pharmacologically similar AMPA receptors. Spontaneous miniature EPSCs (mEPSCs) exhibited unimodal distributions of amplitude and half-amplitude time width and there were no rod-cone differences in mEPSC properties. To examine how release kinetics shape the EPSC, we convolved mEPSC waveforms with empirically determined release rate functions for rods and cones. The predicted EPSC waveform closely matched the actual EPSC evoked by cones, supporting a quantal release model at the photoreceptor synapse. Convolution with the rod release function also produced a good match in rod-driven cells, although the actual EPSC was often somewhat slower than the predicted EPSC, a discrepancy partly explained by rod-rod coupling. Rod-cone differences in the rates of exocytosis are thus a major factor in producing faster cone-driven responses in second-order retinal neurones.


Subject(s)
Receptors, Glutamate/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Benzodiazepines/pharmacology , Benzothiadiazines/pharmacology , Calcium/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials , Kinetics , Models, Biological , Neurotransmitter Agents/metabolism , Quinoxalines/pharmacology , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, Glutamate/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/drug effects , Retinal Horizontal Cells/drug effects , Retinal Horizontal Cells/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism , Urodela
12.
J Neurophysiol ; 90(1): 165-74, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12843308

ABSTRACT

Adenosine is released from retina in darkness; photoreceptors possess A2 adenosine receptors, and A2 agonists inhibit L-type Ca2+ currents (ICa) in rods. We therefore investigated whether A2 agonists inhibit rod inputs into second-order neurons and whether selective antagonists to A1, A2A, or A3 receptors prevent Ca2+ influx through rod ICa. [Ca2+]i changes in rods were assessed with fura-2. ICa in rods and light responses of rods and second-order neurons were recorded using perforated patch-clamp techniques in the aquatic tiger salamander retinal slice preparation. Consistent with earlier results using the A2 agonist N6-[2-(3,5-dimethoxyphenyl)-2-(2-methylphenyl)-ethyl]adenosine (DPMA), the A2A agonist CGS-21680 significantly inhibited ICa and depolarization-evoked [Ca2+]i increases in rods. The A1 antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), and A2A antagonist, ZM-241385, but not the A3 antagonist, VUF-5574, inhibited effects of adenosine on Ca2+ influx in rods. DPCPX and ZM-241385 also inhibited effects of CGS-21680, suggesting they both act at A2A receptors. Both A2 agonists, CGS-21680 and DPMA, reduced light-evoked currents in second-order neurons but not light-evoked voltage responses of rods, suggesting that activation of A2 receptors inhibits transmitter release from rods. The inhibitory effects of CGS-21680 on both depolarization-evoked Ca2+ influx and light-evoked currents in second-order neurons were antagonized by ZM-241385. By itself, ZM-241385 enhanced the light-evoked currents in second-order neurons, suggesting that endogenous levels of adenosine inhibit transmitter release from rods. The effects of these drugs suggest that endogenous adenosine activates an A2-like adenosine receptor on rods leading to inhibition of ICa, which in turn inhibits l-glutamate release from rod photoreceptors.


Subject(s)
Adenosine/physiology , Calcium/physiology , Glutamic Acid/physiology , Neurons/physiology , Receptors, Purinergic P1/metabolism , Retinal Rod Photoreceptor Cells/physiology , Synaptic Transmission , Adenosine/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Electrophysiology , Glutamic Acid/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Retina/physiology , Retinal Rod Photoreceptor Cells/metabolism , Urodela
SELECTION OF CITATIONS
SEARCH DETAIL