1
|
Hellevik AM, Mardoum P, Hahn J, Kölsch Y, D'Orazi FD, Suzuki SC, Godinho L, Lawrence O, Rieke F, Shekhar K, Sanes JR, Baier H, Baden T, Wong RO, Yoshimatsu T. Ancient origin of the rod bipolar cell pathway in the vertebrate retina. Nat Ecol Evol 2024; 8:1165-1179. [PMID: 38627529 DOI: 10.1038/s41559-024-02404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/20/2024] [Indexed: 04/30/2024]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions. The specialized downstream circuit for rod signalling, called the primary rod pathway, is well characterized in mammals, but circuitry for rod signalling in non-mammals is largely unknown. Here we demonstrate that the mammalian primary rod pathway is conserved in zebrafish, which diverged from extant mammals ~400 million years ago. Using single-cell RNA sequencing, we identified two bipolar cell types in zebrafish that are related to mammalian rod bipolar cell (RBCs), the only bipolar type that directly carries rod signals from the outer to the inner retina in the primary rod pathway. By combining electrophysiology, histology and ultrastructural reconstruction of the zebrafish RBCs, we found that, similar to mammalian RBCs, both zebrafish RBC types connect with all rods in their dendritic territory and provide output largely onto amacrine cells. The wiring pattern of the amacrine cells postsynaptic to one RBC type is strikingly similar to that of mammalian RBCs and their amacrine partners, suggesting that the cell types and circuit design of the primary rod pathway emerged before the divergence of teleost fish and mammals. The second RBC type, which forms separate pathways, was either lost in mammals or emerged in fish.
Collapse
Affiliation(s)
- Ayana M Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Philip Mardoum
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA, USA
| | - Yvonne Kölsch
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Florence D D'Orazi
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sachihiro C Suzuki
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Owen Lawrence
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Vision Science Center, University of Washington, Seattle, WA, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Herwig Baier
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO, USA.
- BioRTC, Yobe State University, Damatsuru, Yobe, Nigeria.
| |
Collapse
|
2
|
Shrestha AP, Stiles M, Grambergs RC, Boff JM, Madireddy S, Mondal K, Rajmanna R, Porter H, Sherry DM, Proia RL, Vaithianathan T, Mandal N. The Role of Sphingosine-1-Phosphate Receptor 2 in Mouse Retina Light Responses. Biomolecules 2023; 13:1691. [PMID: 38136563 PMCID: PMC10741782 DOI: 10.3390/biom13121691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/09/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
The bioactive sphingolipid sphingosine-1-phosphate (S1P) acts as a ligand for a family of G protein-coupled S1P receptors (S1PR1-5) to participate in a variety of signaling pathways. However, their specific roles in the neural retina remain unclear. We previously showed that S1P receptor subtype 2 (S1PR2) is expressed in murine retinas, primarily in photoreceptors and bipolar cells, and its expression is altered by retinal stress. This study aims to elucidate the role of S1PR2 in the mouse retina. We examined light responses by electroretinography (ERG), structural differences by optical coherence tomography (OCT), and protein levels by immunohistochemistry (IHC) in wild-type (WT) and S1PR2 knockout (KO) mice at various ages between 3 and 6 months. We found that a- and b-wave responses significantly increased at flash intensities between 400~2000 and 4~2000 cd.s/m2, respectively, in S1PR2 KO mice relative to those of WT controls at baseline. S1PR2 KO mice also exhibited significantly increased retinal nerve fiber layer (RNFL) and outer plexiform layer (OPL) thickness by OCT relative to the WT. Finally, in S1PR2 KO mice, we observed differential labeling of synaptic markers by immunohistochemistry (IHC) and quantitative reverse transcription polymerase chain reaction (RT-qPCR). These results suggest a specific involvement of S1PR2 in the structure and synaptic organization of the retina and a potential role in light-mediated functioning of the retina.
Collapse
Affiliation(s)
- Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Megan Stiles
- Departments of Cell Biology, Neurosurgery, and Pharmacological Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Richard C. Grambergs
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johane M. Boff
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Saivikram Madireddy
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Koushik Mondal
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rhea Rajmanna
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Hunter Porter
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - David M. Sherry
- Departments of Cell Biology, Neurosurgery, and Pharmacological Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Richard L. Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Nawajes Mandal
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
3
|
Hellevik AM, Mardoum P, Hahn J, Kölsch Y, D’Orazi FD, Suzuki SC, Godinho L, Lawrence O, Rieke F, Shekhar K, Sanes JR, Baier H, Baden T, Wong RO, Yoshimatsu T. Ancient origin of the rod bipolar cell pathway in the vertebrate retina. RESEARCH SQUARE 2023:rs.3.rs-3411693. [PMID: 37886445 PMCID: PMC10602083 DOI: 10.21203/rs.3.rs-3411693/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions. Mammals have a specialized downstream circuit for rod signaling called the primary rod pathway, which comprises specific cell types and wiring patterns that are thought to be unique to this lineage. Thus, it has been long assumed that the primary rod pathway evolved in mammals. Here, we challenge this view by demonstrating that the mammalian primary rod pathway is conserved in zebrafish, which diverged from extant mammals ~400 million years ago. Using single-cell RNA-sequencing, we identified two bipolar cell (BC) types in zebrafish that are related to mammalian rod BCs (RBCs) of the primary rod pathway. By combining electrophysiology, histology, and ultrastructural reconstruction of the zebrafish RBCs, we found that, like mammalian RBCs, both zebrafish RBC types connect with all rods in their dendritic territory, and provide output largely onto amacrine cells. The wiring pattern of the amacrine cells post-synaptic to one RBC type is strikingly similar to that of mammalian RBCs, suggesting that the cell types and circuit design of the primary rod pathway have emerged before the divergence of teleost fish and amniotes. The second RBC type, which forms separate pathways, is either lost in mammals or emerged in fish.
Collapse
Affiliation(s)
- Ayana M Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Philip Mardoum
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
| | - Yvonne Kölsch
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Florence D D’Orazi
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Sachihiro C. Suzuki
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, 80802 Munich, Germany
| | - Owen Lawrence
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
- Vision Science Center, University of Washington, Seattle, WA 98195, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Joshua R Sanes
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology & Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
- BioRTC, Yobe State University, Damatsuru, Yobe 620101, Nigeria
| |
Collapse
|
4
|
Shrestha AP, Stiles M, Grambergs RC, Boff JM, Madireddy S, Mondal K, Rajmanna R, Porter H, Sherry D, Proia RL, Vaithianathan T, Mandal N. The role of sphingosine-1-phosphate receptor 2 in mouse retina light responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555709. [PMID: 37732206 PMCID: PMC10508730 DOI: 10.1101/2023.09.01.555709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
The bioactive sphingolipid sphingosine-1-phosphate (S1P) acts as a ligand for a family of G protein-coupled S1P receptors (S1PR1-5) to participate in a variety of signaling pathways. However, their specific roles in the neural retina remain unclear. We previously showed that S1P receptor subtype 2 (S1PR2) is expressed in murine retinas, primarily in photoreceptors and bipolar cells, and its expression is altered by retinal stress. This study aims to elucidate the role of S1PR2 in the mouse retina. We examined light responses by electroretinography (ERG), structural differences by optical coherence tomography (OCT), and protein levels by immunohistochemistry (IHC) in wild-type (WT) and S1PR2 knockout (KO) mice at various ages between 3 and 6 months. We found that a- and b-wave responses significantly increased at flash intensities between 400∼2000 and 4∼2,000 cd.s/m 2 respectively, in S1PR2 KO mice relative to those of WT controls at baseline. S1PR2 KO mice also exhibited significantly increased retinal nerve fiber layer (RNFL) and outer plexiform layer (OPL) thickness by OCT relative to the WT. Finally, in S1PR2 KO mice, we observed differential labeling of synaptic markers by immunohistochemistry (IHC) and quantitative reverse transcription polymerase chain reaction (RT-qPCR). These results suggest a specific involvement of S1PR2 in the structure and synaptic organization of the retina and a potential role in light-mediated functioning of the retina.
Collapse
|
5
|
Orexin-A differentially modulates inhibitory and excitatory synaptic transmission in rat inner retina. Neuropharmacology 2021; 187:108492. [PMID: 33582153 DOI: 10.1016/j.neuropharm.2021.108492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/08/2021] [Accepted: 02/06/2021] [Indexed: 11/21/2022]
Abstract
In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.
Collapse
|
6
|
Veruki ML, Zhou Y, Castilho Á, Morgans CW, Hartveit E. Extrasynaptic NMDA Receptors on Rod Pathway Amacrine Cells: Molecular Composition, Activation, and Signaling. J Neurosci 2019; 39:627-650. [PMID: 30459218 PMCID: PMC6343648 DOI: 10.1523/jneurosci.2267-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/29/2018] [Accepted: 11/14/2018] [Indexed: 12/15/2022] Open
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.
Collapse
Affiliation(s)
- Margaret L Veruki
- University of Bergen, Department of Biomedicine, N-5009 Bergen, Norway, and
| | - Yifan Zhou
- University of Bergen, Department of Biomedicine, N-5009 Bergen, Norway, and
| | - Áurea Castilho
- University of Bergen, Department of Biomedicine, N-5009 Bergen, Norway, and
| | - Catherine W Morgans
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239
| | - Espen Hartveit
- University of Bergen, Department of Biomedicine, N-5009 Bergen, Norway, and
| |
Collapse
|
7
|
Dunn VK, Gleason E. Inhibition of endocytosis suppresses the nitric oxide-dependent release of Cl- in retinal amacrine cells. PLoS One 2018; 13:e0201184. [PMID: 30044876 PMCID: PMC6059450 DOI: 10.1371/journal.pone.0201184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/10/2018] [Indexed: 11/18/2022] Open
Abstract
Our lab has previously shown that nitric oxide (NO) can alter the synaptic response properties of amacrine cells by releasing Cl- from internal acidic compartments. This alteration in the Cl- gradient brings about a positive shift in the reversal potential of the GABA-gated current, which can convert inhibitory synapses into excitatory synapses. Recently, we have shown that the cystic fibrosis transmembrane regulator (CFTR) Cl- channel is involved in the Cl- release. Here, we test the hypothesis that (acidic) synaptic vesicles are a source of NO-releasable Cl- in chick retinal amacrine cells. If SVs are a source of Cl-, then depleting synaptic vesicles should decrease the nitric oxide-dependent shift in the reversal potential of the GABA-gated current. The efficacy of four inhibitors of dynamin (dynasore, Dyngo 4a, Dynole 34-2, and MiTMAB) were evaluated. In order to deplete synaptic vesicles, voltage-steps were used to activate V-gated Ca2+ channels and stimulate the synaptic vesicle cycle either under control conditions or after treatment with the dynamin inhibitors. Voltage-ramps were used to measure the NO-dependent shift in the reversal potential of the GABA-gated currents under both conditions. Our results reveal that activating the synaptic vesicle cycle in the presence of dynasore or Dyngo 4a blocked the NO-dependent shift in EGABA. However, we also discovered that some dynamin inhibitors reduced Ca2+ signaling and L-type Ca2+ currents. Conversely, dynasore also increased neurotransmitter release at autaptic sites. To further resolve the mechanism underlying the inhibition of the NO-dependent shift in the reversal potential for the GABA-gated currents, we also tested the effects of the clathrin assembly inhibitor Pitstop 2 and found that this compound also inhibited the shift. These data provide evidence that dynamin inhibitors have multiple effects on amacrine cell synaptic transmission. These data also suggest that inhibition of endocytosis disrupts the ability of NO to elicit Cl- release from internal stores which may in part be due to depletion of synaptic vesicles.
Collapse
Affiliation(s)
- Vernon K. Dunn
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
8
|
Lipin MY, Vigh J. Quantifying the effect of light activated outer and inner retinal inhibitory pathways on glutamate release from mixed bipolar cells. Synapse 2018; 72:e22028. [PMID: 29360185 DOI: 10.1002/syn.22028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 11/12/2022]
Abstract
Inhibition mediated by horizontal and amacrine cells in the outer and inner retina, respectively, are fundamental components of visual processing. Here, our purpose was to determine how these different inhibitory processes affect glutamate release from ON bipolar cells when the retina is stimulated with full-field light of various intensities. Light-evoked membrane potential changes (ΔVm ) were recorded directly from axon terminals of intact bipolar cells receiving mixed rod and cone inputs (Mbs) in slices of dark-adapted goldfish retina. Inner and outer retinal inhibition to Mbs was blocked with bath applied picrotoxin (PTX) and NBQX, respectively. Then, control and pharmacologically modified light responses were injected into axotomized Mb terminals as command potentials to induce voltage-gated Ca2+ influx (QCa ) and consequent glutamate release. Stimulus-evoked glutamate release was quantified by the increase in membrane capacitance (ΔCm ). Increasing depolarization of Mb terminals upon removal of inner and outer retinal inhibition enhanced the ΔVm /QCa ratio equally at a given light intensity and inhibition did not alter the overall relation between QCa and ΔCm . However, relative to control, light responses recorded in the presence of PTX and PTX + NBQX increased ΔCm unevenly across different stimulus intensities: at dim stimulus intensities predominantly the inner retinal GABAergic inhibition controlled release from Mbs, whereas the inner and outer retinal inhibition affected release equally in response to bright stimuli. Furthermore, our results suggest that non-linear relationship between QCa and glutamate release can influence the efficacy of inner and outer retinal inhibitory pathways to mediate Mb output at different light intensities.
Collapse
Affiliation(s)
- Mikhail Y Lipin
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, Colorado, 80523-1617
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, Colorado, 80523-1617
| |
Collapse
|
9
|
Travis AM, Heflin SJ, Hirano AA, Brecha NC, Arshavsky VY. Dopamine-Dependent Sensitization of Rod Bipolar Cells by GABA Is Conveyed through Wide-Field Amacrine Cells. J Neurosci 2018; 38:723-732. [PMID: 29217689 PMCID: PMC5777116 DOI: 10.1523/jneurosci.1994-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 11/21/2022] Open
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.
Collapse
Affiliation(s)
| | - Stephanie J Heflin
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710
| | - Arlene A Hirano
- Department of Neurobiology
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073
| | - Nicholas C Brecha
- Department of Neurobiology
- Department of Medicine, and
- Department of Ophthalmology, Stein Eye Institute David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, and
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073
| | - Vadim Y Arshavsky
- Department of Pharmacology and
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710
| |
Collapse
|
10
|
Albanna W, Neumaier F, Lüke JN, Kotliar K, Conzen C, Lindauer U, Hescheler J, Clusmann H, Schneider T, Schubert GA. Unconjugated bilirubin modulates neuronal signaling only in wild-type mice, but not after ablation of the R-type/Ca v 2.3 voltage-gated calcium channel. CNS Neurosci Ther 2017; 24:222-230. [PMID: 29274300 DOI: 10.1111/cns.12791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/04/2017] [Accepted: 12/04/2017] [Indexed: 01/30/2023] Open
Abstract
INTRODUCTION The relationship between blood metabolites and hemoglobin degradation products (BMHDPs) formed in the cerebrospinal fluid and the development of vasospasm and delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (aSAH) has been the focus of several previous studies, but their molecular and cellular targets remain to be elucidated. METHODS Because BMHDP-induced changes in Cav 2.3 channel function are thought to contribute to DCI after aSAH, we studied their modulation by unconjugated bilirubin (UCB) in an organotypical neuronal network from wild-type (WT) and Cav 2.3-deficient animals (KO). Murine retinae were isolated from WT and KO and superfused with nutrient solution. Electroretinograms were recorded before, during, and after superfusion with UCB. Transretinal signaling was analyzed as b-wave, implicit time, and area under the curve (AUC). RESULTS Superfusion of UCB significantly attenuated the b-wave amplitude in the isolated retina from wild-type mice by 14.9% (P < 0.05), followed by gradual partial recovery (P = 0.09). Correspondingly, AUC decreased significantly with superfusion of UCB (P < 0.05). During washout, the b-wave amplitude returned to baseline (P = 0.2839). The effects of UCB were absent in Cav 2.3-deficient mice, lacking the expression of Cav 2.3 as proofed on the biochemical level. CONCLUSIONS Ex vivo neuronal recording in the murine retina is able to detect transient impairment of transretinal signaling by UCB in WT, but not in KO. This new model may be useful to further clarify the role of calcium channels in neuronal signal alteration in the presence of BHMDPs.
Collapse
Affiliation(s)
- Walid Albanna
- Institute for Neurophysiology, University of Cologne, Cologne, Germany.,Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Felix Neumaier
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Jan Niklas Lüke
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Aachen, Germany
| | - Catharina Conzen
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Ute Lindauer
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Hans Clusmann
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
11
|
Opere CA, Heruye S, Njie-Mbye YF, Ohia SE, Sharif NA. Regulation of Excitatory Amino Acid Transmission in the Retina: Studies on Neuroprotection. J Ocul Pharmacol Ther 2017; 34:107-118. [PMID: 29267132 DOI: 10.1089/jop.2017.0085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Excitotoxicity occurs in neurons due to the accumulation of excitatory amino acids such as glutamate in the synaptic and extrasynaptic locations. In the retina, excessive glutamate concentrations trigger a neurotoxic cascade involving several mechanisms, including the elevation of intracellular calcium (Ca2+) and the activation of α-amino-3-hydroxy 5-methyl-4-iso-xazole-propionic acid/kainate (AMPA/KA) and N-methyl-d-aspartate (NMDA) receptors leading to retinal degeneration. Both ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs) are present in the mammalian retina. Indeed, due to the abundant expression of GluRs, the mammalian retina is highly susceptible to excitotoxic neurodegeneration. Excitotoxicity has been postulated to present a common downstream mechanism for several stimuli, including hypoglycemia, hypoxia, ischemia, and chronic neurodegenerative diseases. Experimental approaches to the study of neuroprotection in the retina have utilized insults that trigger hypoxia, hypoglycemia, or excitotoxicity. Using these experimental approaches, the neuroprotective potential of GluR agents, including the NMDA receptor modulators (MK801, ifenprodil, memantine); AMPA/KA receptor antagonist (CNQX); Group II and III mGluR agonists (LY354740, quisqualate); and Ca2+-channel blockers (diltiazem, lomerizine, verapamil, ω-conotoxin), and others (pituitary adenylate cyclase activating polypeptide, neuropeptide Y, acetylcholine receptor agonists) have been elucidated. In addition to corroborating the exocytotic role of excitatory amino acids in retinal degeneration, these studies affirm that multiple mechanism/s contribute to the prevention of damage caused by excitotoxicity in the retina. Therefore, it is feasible that several pathways are involved in protecting the retina from toxic insults in ocular neurodegenerative conditions such as glaucoma and retinal ischemia. Furthermore, these experimental models are viable tools for evaluating therapeutic candidates in ocular neuropathies.
Collapse
Affiliation(s)
- Catherine A Opere
- 1 Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University , Omaha, Nebraska
| | - Segewkal Heruye
- 1 Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University , Omaha, Nebraska
| | - Ya-Fatou Njie-Mbye
- 2 Department of Environmental and Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| | - Sunny E Ohia
- 2 Department of Environmental and Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas
| | - Najam A Sharif
- 2 Department of Environmental and Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University , Houston, Texas.,3 Santen Incorporated , Emeryville, California
| |
Collapse
|
12
|
Mohammadi E, Shamsizadeh A, Salari E, Fatemi I, Allahtavakoli M, Roohbakhsh A. Effect of TPMPA (GABACreceptor antagonist) on neuronal response properties in rat barrel cortex. Somatosens Mot Res 2017; 34:108-115. [DOI: 10.1080/08990220.2017.1317240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Elham Mohammadi
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Shamsizadeh
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Salari
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Iman Fatemi
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Kim MH, von Gersdorff H. Postsynaptic Plasticity Triggered by Ca²⁺-Permeable AMPA Receptor Activation in Retinal Amacrine Cells. Neuron 2016; 89:507-20. [PMID: 26804991 DOI: 10.1016/j.neuron.2015.12.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/18/2015] [Accepted: 12/15/2015] [Indexed: 01/04/2023]
Abstract
Amacrine cells are thought to be a major locus for mechanisms of light adaptation and contrast enhancement in the retina. However, the potential for plasticity in their AMPA receptor currents remains largely unknown. Using paired patch-clamp recordings between bipolar cell terminals and amacrine cells, we have simultaneously measured presynaptic membrane capacitance changes and EPSCs. Repetitive bipolar cell depolarizations, designed to maintain the same amount of exocytosis, nevertheless significantly potentiated evoked EPSCs in a subpopulation of amacrine cells. Likewise, repetitive iontophoresis (or puffs) of glutamate (or AMPA) onto the dendrites of amacrine cells also significantly potentiated evoked currents and [Ca(2+)]i rises. However, strong postsynaptic Ca(2+) buffering with BAPTA abolished the potentiation and selective antagonists of Ca(2+)-permeable AMPA receptors also blocked the potentiation of AMPA-mediated currents. Together these results suggest that Ca(2+) influx via Ca(2+)-permeable AMPA receptors can elicit a rapid form of postsynaptic plasticity in a subgroup of amacrine cell dendrites.
Collapse
Affiliation(s)
- Mean-Hwan Kim
- The Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Henrique von Gersdorff
- The Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
14
|
Zhou Y, Tencerová B, Hartveit E, Veruki ML. Functional NMDA receptors are expressed by both AII and A17 amacrine cells in the rod pathway of the mammalian retina. J Neurophysiol 2016; 115:389-403. [PMID: 26561610 PMCID: PMC4760463 DOI: 10.1152/jn.00947.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/08/2015] [Indexed: 11/22/2022] Open
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.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
15
|
The Synaptic and Morphological Basis of Orientation Selectivity in a Polyaxonal Amacrine Cell of the Rabbit Retina. J Neurosci 2015; 35:13336-50. [PMID: 26424882 DOI: 10.1523/jneurosci.1712-15.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
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.
Collapse
|
16
|
Balakrishnan V, Puthussery T, Kim MH, Taylor WR, von Gersdorff H. Synaptic Vesicle Exocytosis at the Dendritic Lobules of an Inhibitory Interneuron in the Mammalian Retina. Neuron 2015; 87:563-75. [PMID: 26247863 DOI: 10.1016/j.neuron.2015.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/07/2015] [Accepted: 07/20/2015] [Indexed: 11/16/2022]
Abstract
Ribbon synapses convey sustained and phasic excitatory drive within retinal microcircuits. However, the properties of retinal inhibitory synapses are less well known. AII-amacrine cells are interneurons in the retina that exhibit large glycinergic synapses at their dendritic lobular appendages. Using membrane capacitance measurements, we observe robust exocytosis elicited by the opening of L-type Ca(2+) channels located on the lobular appendages. Two pools of synaptic vesicles were detected: a small, rapidly releasable pool and a larger and more slowly releasable pool. Depending on the stimulus, either paired-pulse depression or facilitation could be elicited. During early postnatal maturation, the coupling of the exocytosis Ca(2+)-sensor to Ca(2+) channel becomes tighter. Light-evoked depolarizations of the AII-amacrine cell elicited exocytosis that was graded to light intensity. Our results suggest that AII-amacrine cell synapses are capable of providing both phasic and sustained inhibitory input to their postsynaptic partners without the benefit of synaptic ribbons.
Collapse
Affiliation(s)
| | - Theresa Puthussery
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Mean-Hwan Kim
- The Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - W Rowland Taylor
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Henrique von Gersdorff
- The Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
17
|
The ubiquitous nature of multivesicular release. Trends Neurosci 2015; 38:428-38. [PMID: 26100141 DOI: 10.1016/j.tins.2015.05.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/20/2015] [Accepted: 05/24/2015] [Indexed: 11/21/2022]
Abstract
'Simplicity is prerequisite for reliability' (E.W. Dijkstra [1]) Presynaptic action potentials trigger the fusion of vesicles to release neurotransmitter onto postsynaptic neurons. Each release site was originally thought to liberate at most one vesicle per action potential in a probabilistic fashion, rendering synaptic transmission unreliable. However, the simultaneous release of several vesicles, or multivesicular release (MVR), represents a simple mechanism to overcome the intrinsic unreliability of synaptic transmission. MVR was initially identified at specialized synapses but is now known to be common throughout the brain. MVR determines the temporal and spatial dispersion of transmitter, controls the extent of receptor activation, and contributes to adapting synaptic strength during plasticity and neuromodulation. MVR consequently represents a widespread mechanism that extends the dynamic range of synaptic processing.
Collapse
|
18
|
Popova E. GABAergic neurotransmission and retinal ganglion cell function. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2015; 201:261-83. [PMID: 25656810 DOI: 10.1007/s00359-015-0981-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 01/13/2023]
Abstract
Ganglion cells are the output retinal neurons that convey visual information to the brain. There are ~20 different types of ganglion cells, each encoding a specific aspect of the visual scene as spatial and temporal contrast, orientation, direction of movement, presence of looming stimuli; etc. Ganglion cell functioning depends on the intrinsic properties of ganglion cell's membrane as well as on the excitatory and inhibitory inputs that these cells receive from other retinal neurons. GABA is one of the most abundant inhibitory neurotransmitters in the retina. How it modulates the activity of different types of ganglion cells and what is its significance in extracting the basic features from visual scene are questions with fundamental importance in visual neuroscience. The present review summarizes current data concerning the types of membrane receptors that mediate GABA action in proximal retina; the effects of GABA and its antagonists on the ganglion cell light-evoked postsynaptic potentials and spike discharges; the action of GABAergic agents on centre-surround organization of the receptive fields and feature related ganglion cell activity. Special emphasis is put on the GABA action regarding the ON-OFF and sustained-transient ganglion cell dichotomy in both nonmammalian and mammalian retina.
Collapse
Affiliation(s)
- E Popova
- Department of Physiology, Medical Faculty, Medical University, 1431, Sofia, Bulgaria,
| |
Collapse
|
19
|
Lipin MY, Vigh J. Calcium spike-mediated digital signaling increases glutamate output at the visual threshold of retinal bipolar cells. J Neurophysiol 2014; 113:550-66. [PMID: 25339710 DOI: 10.1152/jn.00378.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Most retinal bipolar cells (BCs) transmit visual input from photoreceptors to ganglion cells using graded potentials, but some also generate calcium or sodium spikes. Sodium spikes are thought to increase temporal precision of light-evoked BC signaling; however, the role of calcium spikes in BCs is not fully understood. Here we studied how calcium spikes and graded responses mediate neurotransmitter release from Mb-type BCs, known to produce both. In dark-adapted goldfish retinal slices, light induced spikes in 40% of the axon terminals of intact Mbs; in the rest, light generated graded responses. These light-evoked membrane potentials were used to depolarize axotomized Mb terminals where depolarization-evoked calcium current (ICa) and consequent exocytosis-associated membrane capacitance increases (ΔCm) could be precisely measured. When evoked by identical dim light intensities, spiking responses transferred more calcium (Q(Ca)) and triggered larger exocytosis with higher efficiency (ΔCm/Q(Ca)) than graded potentials. Q(Ca) was translated into exocytosis linearly when transferred with spikes and supralinearly when transferred with graded responses. At the Mb output (ΔCm), spiking responses coded light intensity with numbers and amplitude whereas graded responses coded with amplitude, duration, and steepness. Importantly, spiking responses saturated exocytosis within scotopic range but graded potentials did not. We propose that calcium spikes in Mbs increase signal input-output ratio by boosting Mb glutamate release at threshold intensities. Therefore, spiking Mb responses are suitable to transfer low-light-intensity signals to ganglion cells with higher gain, whereas graded potentials signal for light over a wider range of intensities at the Mb output.
Collapse
Affiliation(s)
- Mikhail Y Lipin
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
20
|
Wang XH, Wu Y, Yang XF, Miao Y, Zhang CQ, Dong LD, Yang XL, Wang Z. Cannabinoid CB1 receptor signaling dichotomously modulates inhibitory and excitatory synaptic transmission in rat inner retina. Brain Struct Funct 2014; 221:301-16. [DOI: 10.1007/s00429-014-0908-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/26/2014] [Indexed: 12/01/2022]
|
21
|
Popova E. Ionotropic GABA Receptors and Distal Retinal ON and OFF Responses. SCIENTIFICA 2014; 2014:149187. [PMID: 25143858 PMCID: PMC4131092 DOI: 10.1155/2014/149187] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/24/2014] [Accepted: 05/27/2014] [Indexed: 05/27/2023]
Abstract
In the vertebrate retina, visual signals are segregated into parallel ON and OFF pathways, which provide information for light increments and decrements. The segregation is first evident at the level of the ON and OFF bipolar cells in distal retina. The activity of large populations of ON and OFF bipolar cells is reflected in the b- and d-waves of the diffuse electroretinogram (ERG). The role of gamma-aminobutyric acid (GABA), acting through ionotropic GABA receptors in shaping the ON and OFF responses in distal retina, is a matter of debate. This review summarized current knowledge about the types of the GABAergic neurons and ionotropic GABA receptors in the retina as well as the effects of GABA and specific GABAA and GABAC receptor antagonists on the activity of the ON and OFF bipolar cells in both nonmammalian and mammalian retina. Special emphasis is put on the effects on b- and d-waves of the ERG as a useful tool for assessment of the overall function of distal retinal ON and OFF channels. The role of GABAergic system in establishing the ON-OFF asymmetry concerning the time course and absolute and relative sensitivity of the ERG responses under different conditions of light adaptation in amphibian retina is also discussed.
Collapse
Affiliation(s)
- E. Popova
- Department of Physiology, Medical Faculty, Medical University, 1431 Sofia, Bulgaria
| |
Collapse
|
22
|
Popova E. Effects of picrotoxin on light adapted frog electroretinogram are not due entirely to its action in proximal retina. Vision Res 2014; 101:138-50. [PMID: 24999030 DOI: 10.1016/j.visres.2014.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/20/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
In order to evaluate the site of action of picrotoxin (antagonist of ionotropic GABA receptors) on the electroretinographic (ERG) b- and d-waves, in this study we compared its effects on the intensity-response function of the ERG waves in intact light adapted frog eyecup preparations with its effects in eyecups, where the activity of proximal neurons was blocked by 1 mMN-methyl-d-aspartate (MNDA). Picrotoxin markedly enhanced the b- and d-wave amplitude and slowed the time course of the responses at all stimulus intensities in the intact eyecups. Perfusion with NMDA alone caused significant enhancement of the b-wave amplitude and diminution of the d-wave amplitude without altering their time course in the entire intensity range. When picrotoxin was applied in combination with NMDA, an enhancement of the b-wave amplitude and slowing of its time course were observed at all stimulus intensities. The increase of the b-wave amplitude was significantly higher than that seen in NMDA group. Combined application of picrotoxin and NMDA caused an enhancement of the d-wave amplitude at the lower stimulus intensities and its diminution at the higher ones, while the d-wave time course was delayed over the entire intensity range. The results obtained indicate that a part of picrotoxin effects on the amplitude and time course of the photopic ERG b- and d-waves are due to its action in the distal frog retina.
Collapse
Affiliation(s)
- E Popova
- Department of Physiology, Medical Faculty, Medical University, 1431 Sofia, Bulgaria.
| |
Collapse
|
23
|
Tooker RE, Lipin MY, Leuranguer V, Rozsa E, Bramley JR, Harding JL, Reynolds MM, Vigh J. Nitric oxide mediates activity-dependent plasticity of retinal bipolar cell output via S-nitrosylation. J Neurosci 2013; 33:19176-93. [PMID: 24305814 PMCID: PMC3850041 DOI: 10.1523/jneurosci.2792-13.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/23/2013] [Accepted: 10/31/2013] [Indexed: 11/21/2022] Open
Abstract
Coding a wide range of light intensities in natural scenes poses a challenge for the retina: adaptation to bright light should not compromise sensitivity to dim light. Here we report a novel form of activity-dependent synaptic plasticity, specifically, a "weighted potentiation" that selectively increases output of Mb-type bipolar cells in the goldfish retina in response to weak inputs but leaves the input-output ratio for strong stimuli unaffected. In retinal slice preparation, strong depolarization of bipolar terminals significantly lowered the threshold for calcium spike initiation, which originated from a shift in activation of voltage-gated calcium currents (ICa) to more negative potentials. The process depended upon glutamate-evoked retrograde nitric oxide (NO) signaling as it was eliminated by pretreatment with an NO synthase blocker, TRIM. The NO-dependent ICa modulation was cGMP independent but could be blocked by N-ethylmaleimide (NEM), indicating that NO acted via an S-nitrosylation mechanism. Importantly, the NO action resulted in a weighted potentiation of Mb output in response to small (≤-30 mV) depolarizations. Coincidentally, light flashes with intensity ≥ 2.4 × 10(8) photons/cm(2)/s lowered the latency of scotopic (≤ 2.4 × 10(8) photons/cm(2)/s) light-evoked calcium spikes in Mb axon terminals in an NEM-sensitive manner, but light responses above cone threshold (≥ 3.5 × 10(9) photons/cm(2)/s) were unaltered. Under bright scotopic/mesopic conditions, this novel form of Mb output potentiation selectively amplifies dim retinal inputs at Mb → ganglion cell synapses. We propose that this process might counteract decreases in retinal sensitivity during light adaptation by preventing the loss of visual information carried by dim scotopic signals.
Collapse
Affiliation(s)
| | | | | | - Eva Rozsa
- Department of Biomedical Sciences and
| | | | | | - Melissa M. Reynolds
- Department of Chemistry, Colorado State University, Ft. Collins, Colorado 80523
| | | |
Collapse
|
24
|
Two-photon imaging of nonlinear glutamate release dynamics at bipolar cell synapses in the mouse retina. J Neurosci 2013; 33:10972-85. [PMID: 23825403 DOI: 10.1523/jneurosci.1241-13.2013] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alpha/Y-type retinal ganglion cells encode visual information with a receptive field composed of nonlinear subunits. This nonlinear subunit structure enhances sensitivity to patterns composed of high spatial frequencies. The Y-cell's subunits are the presynaptic bipolar cells, but the mechanism for the nonlinearity remains incompletely understood. We investigated the synaptic basis of the subunit nonlinearity by combining whole-cell recording of mouse Y-type ganglion cells with two-photon fluorescence imaging of a glutamate sensor (iGluSnFR) expressed on their dendrites and throughout the inner plexiform layer. A control experiment designed to assess iGluSnFR's dynamic range showed that fluorescence responses from Y-cell dendrites increased proportionally with simultaneously recorded excitatory current. Spatial resolution was sufficient to readily resolve independent release at intermingled ON and OFF bipolar terminals. iGluSnFR responses at Y-cell dendrites showed strong surround inhibition, reflecting receptive field properties of presynaptic release sites. Responses to spatial patterns located the origin of the Y-cell nonlinearity to the bipolar cell output, after the stage of spatial integration. The underlying mechanism differed between OFF and ON pathways: OFF synapses showed transient release and strong rectification, whereas ON synapses showed relatively sustained release and weak rectification. At ON synapses, the combination of fast release onset with slower release offset explained the nonlinear response of the postsynaptic ganglion cell. Imaging throughout the inner plexiform layer, we found transient, rectified release at the central-most levels, with increasingly sustained release near the borders. By visualizing glutamate release in real time, iGluSnFR provides a powerful tool for characterizing glutamate synapses in intact neural circuits.
Collapse
|
25
|
Amperometric resolution of a prespike stammer and evoked phases of fast release from retinal bipolar cells. J Neurosci 2013; 33:8144-58. [PMID: 23658155 DOI: 10.1523/jneurosci.5062-12.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The neurotransmitter glutamate is used by most neurons in the brain to activate a multitude of different types of glutamate receptors and transporters involved in fast and relatively slower signaling. Synaptic ribbons are large presynaptic structures found in neurons involved in vision, balance, and hearing, which use a large number of glutamate-filled synaptic vesicles to meet their signaling demands. To directly measure synaptic vesicle release events, the ribbon-type presynaptic terminals of goldfish retinal bipolar cells were coaxed to release a false transmitter that could be monitored with amperometry by placing the carbon fiber directly on the larger synaptic terminal. Spontaneous secretion events formed a unimodal charge distribution, but single spike properties were heterogeneous. Larger events rose exponentially without interruption (τ ∼ 30 μs), and smaller events exhibited a stammer in their rising phase that is interpreted as a brief pause in pore dilation, a characteristic commonly associated with large dense core granule fusion pores. These events were entirely Ca(2+)-dependent. Holding the cells at -60 mV halted spontaneous release; and when the voltage was stepped to >-40 mV, secretion ensued. When stepping the voltage to 0 mV, novel kinetic phases of vesicle recruitment were revealed. Approximately 14 vesicles were released per ribbon in two kinetic phases with time constants of 1.5 and 16 ms, which are proposed to represent different primed states within the population of docked vesicles.
Collapse
|
26
|
Rozsa E, Vigh J. Glycine transporter 1 modulates GABA release from amacrine cells by controlling occupancy of coagonist binding site of NMDA receptors. J Neurophysiol 2013; 110:1393-403. [PMID: 23803324 DOI: 10.1152/jn.00193.2013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The occupancy of coagonist binding sites of NMDA receptors (NMDARs) by glycine or d-serine has been thought to mediate NMDAR-dependent excitatory signaling, as simultaneous binding of glutamate and a coagonist is obligatory for NMDAR activation. Amacrine cells (ACs) mediating GABAergic feedback inhibition of mixed bipolar cells (Mbs) in the goldfish retina have been shown to express NMDARs. Here we studied whether NMDAR-mediated GABAergic inhibitory currents (IGABA) recorded from the axon terminals of Mbs are influenced by experimental manipulations altering retinal glycine and d-serine levels. Feedback IGABA in Mb axon terminals was triggered by focal NMDA application or by synaptically released glutamate from depolarized Mb terminals. In both cases, blocking the coagonist binding sites of NMDARs eliminated the NMDAR-dependent IGABA, demonstrating that coagonist binding is critical in mediating NMDAR activity-triggered GABA release. Glycine transporter 1 (GLYT1) inhibition increased IGABA, indicating that coagonist binding sites of NMDARs on ACs providing GABAergic feedback inhibition to Mbs were not saturated. Focal glycine application, in the presence of the ionotropic glycine receptor blocker strychnine, triggered a GLYT1-dependent current in ACs, suggesting that GLYT1 expressed by putative glycinergic ACs controls the saturation level of NMDARs' coagonist sites. External d-serine also increased NMDAR activation-triggered IGABA in Mbs, further substantiating that the coagonist sites were unsaturated. Together, our findings demonstrate that coagonist modulation of glutamatergic input to GABAergic ACs via NMDARs is strongly reflected in the AC neuronal output (i.e., transmitter release) and thus is critical in GABAergic signal transfer function in the inner retina.
Collapse
Affiliation(s)
- Eva Rozsa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | | |
Collapse
|
27
|
Tanaka M, Tachibana M. Independent control of reciprocal and lateral inhibition at the axon terminal of retinal bipolar cells. J Physiol 2013; 591:3833-51. [PMID: 23690563 DOI: 10.1113/jphysiol.2013.253179] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Bipolar cells (BCs), the second order neurons in the vertebrate retina, receive two types of GABAergic feedback inhibition at their axon terminal: reciprocal and lateral inhibition. It has been suggested that two types of inhibition may be mediated by different pathways. However, how each inhibition is controlled by excitatory BC output remains to be clarified. Here, we applied single/dual whole cell recording techniques to the axon terminal of electrically coupled BCs in slice preparation of the goldfish retina, and found that each inhibition was regulated independently. Activation voltage of each inhibition was different: strong output from a single BC activated reciprocal inhibition, but could not activate lateral inhibition. Outputs from multiple BCs were essential for activation of lateral inhibition. Pharmacological examinations revealed that composition of transmitter receptors and localization of Na(+) channels were different between two inhibitory pathways, suggesting that different amacrine cells may mediate each inhibition. Depending on visual inputs, each inhibition could be driven independently. Model simulation showed that reciprocal and lateral inhibition cooperatively reduced BC outputs as well as background noise, thereby preserving high signal-to-noise ratio. Therefore, we conclude that excitatory BC output is efficiently regulated by the dual operating mechanisms of feedback inhibition without deteriorating the quality of visual signals.
Collapse
Affiliation(s)
- Masashi Tanaka
- Department of Psychology, Graduate School of Humanities and Sociology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | |
Collapse
|
28
|
Paired-pulse plasticity in the strength and latency of light-evoked lateral inhibition to retinal bipolar cell terminals. J Neurosci 2012; 32:11688-99. [PMID: 22915111 DOI: 10.1523/jneurosci.0547-12.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Synapses in the inner plexiform layer of the retina undergo short-term plasticity that may mediate different forms of adaptation to regularities in light stimuli. Using patch-clamp recordings from axotomized goldfish Mb bipolar cell (BC) terminals with paired-pulse light stimulation, we isolated and quantified the short-term plasticity of GABAergic lateral IPSCs (L-IPSCs). Bright light stimulation evoked ON and OFF L-IPSCs in axotomized BCs, which had distinct onset latencies (∼50-80 and ∼70-150 ms, respectively) that depended on background light adaptation. We observed plasticity in both the synaptic strength and latency of the L-IPSCs. With paired light stimulation, latencies of ON L-IPSCs increased at paired-pulse intervals (PPIs) of 50 and 300 ms, whereas OFF L-IPSC latencies decreased at the 300 ms PPI. ON L-IPSCs showed paired-pulse depression at intervals <1 s, whereas OFF L-IPSCs showed depression at intervals ≤1 s and amplitude facilitation at longer intervals (1-2 s). This biphasic form of L-IPSC plasticity may underlie adaptation and sensitization to surround temporal contrast over multiple timescales. Block of retinal signaling at GABA(A)Rs and AMPARs differentially affected ON and OFF L-IPSCs, confirming that these two types of feedback inhibition are mediated by distinct and convergent retinal pathways with different mechanisms of plasticity. We propose that these plastic changes in the strength and timing of L-IPSCs help to dynamically shape the time course of glutamate release from ON-type BC terminals. Short-term plasticity of L-IPSCs may thus influence the strength, timing, and spatial extent of amacrine and ganglion cell inhibitory surrounds.
Collapse
|
29
|
Abstract
Excitatory amino acid transporters (EAATs) terminate signaling in the CNS by clearing released glutamate. Glutamate also evokes an EAAT-mediated Cl(-) current, but its role in CNS signaling is poorly understood. We show in mouse retina that EAAT-mediated Cl(-) currents that were evoked by light inhibit rod pathway signaling. EAATs reside on rod bipolar cell axon terminals where GABA and glycine receptors also mediate light-evoked inhibition. We found that the mode of inhibition depended on light intensity. Dim light evoked GABAergic and glycinergic inhibition with rapid kinetics and a large spatial extent. Bright light evoked predominantly EAAT-mediated inhibition with slow kinetics and a small spatial extent. The switch to EAAT-mediated signaling in bright light supplements receptor-mediated signaling to expand the dynamic range of inhibition and contributes to the transition from rod to cone signaling by suppressing rod pathway signaling in bright light conditions.
Collapse
|
30
|
Light-evoked lateral GABAergic inhibition at single bipolar cell synaptic terminals is driven by distinct retinal microcircuits. J Neurosci 2011; 31:15884-93. [PMID: 22049431 DOI: 10.1523/jneurosci.2959-11.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inhibitory amacrine cells (ACs) filter visual signals crossing the retina by modulating the excitatory, glutamatergic output of bipolar cells (BCs) on multiple temporal and spatial scales. Reciprocal feedback from ACs provides focal inhibition that is temporally locked to the activity of presynaptic BC activity, whereas lateral feedback originates from ACs excited by distant BCs. These distinct feedback mechanisms permit temporal and spatial computation at BC terminals. Here, we used a unique preparation to study light-evoked IPSCs recorded from axotomized terminals of ON-type mixed rod/cone BCs (Mb) in goldfish retinal slices. In this preparation, light-evoked IPSCs could only reach axotomized BC terminals via the lateral feedback pathway, allowing us to study lateral feedback in the absence of overlapping reciprocal feedback components. We found that light evokes ON and OFF lateral IPSCs (L-IPSCs) in Mb terminals having different temporal patterns and conveyed via distinct retinal pathways. The relative contribution of rods versus cones to ON and OFF L-IPSCs was light intensity dependent. ACs presynaptic to Mb BC terminals received inputs via AMPA/KA- and NMDA-type receptors in both the ON and OFF pathways, and used TTX-sensitive sodium channels to boost signal transfer along their processes. ON and OFF L-IPSCs, like reciprocal feedback IPSCs, were mediated by both GABA(A) and GABA(C) receptors. However, our results suggest that lateral and reciprocal feedback do not cross-depress each other, and are therefore mediated by distinct populations of ACs. These findings demonstrate that retinal inhibitory circuits are highly specialized to modulate BC output at different light intensities.
Collapse
|
31
|
Alnawaiseh M, Albanna W, Chen C, Campbell KP, Hescheler J, Lüke M, Schneider T. Two separate Ni(2+) -sensitive voltage-gated Ca(2+) channels modulate transretinal signalling in the isolated murine retina. Acta Ophthalmol 2011; 89:e579-90. [PMID: 21883984 PMCID: PMC3274955 DOI: 10.1111/j.1755-3768.2011.02167.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Light-evoked responses from vertebrate retinas were recorded as an electroretinogram (ERG). The b-wave is the most prominent component of the ERG, and in the bovine retina its NiCl(2) -sensitive component was attributed to reciprocal signalling by pharmacoresistant R-type voltage-gated Ca(2+) channels, which similar to other voltage-dependent Ca(2+) channels trigger and control neurotransmitter release. The murine retina has the great advantage that the effect of gene inactivation for Ni(2+) -sensitive Ca(2+) channels can be analysed to prove or disprove that any of these Ca(2+) channels is involved in retinal signalling. METHODS Superfused retinas from different murine genotypes lacking either one or both highly Ni(2+) -sensitive voltage-gated Ca(2+) channels were used to record their ex vivo ERGs. RESULTS The isolated retinas from mice lacking Ca(v)2.3 R-type or Ca(v)3.2 T-type or both voltage-gated Ca(2+) channels were superfused with a NiCl(2) (15 μm) containing nutrient solution. The change in the b-wave amplitude and implicit time, caused by NiCl(2), was calculated as a difference spectrum and compared to data from control animals. From the results, it can be deduced that Ca(v)2.3 contributes rather to a later component in the b-wave response, while in the absence of Ca(v)3.2 the gain of Ni(2+) -mediated increase in the b-wave amplitude is significantly increased, probably due to a loss of reciprocal inhibition to photoreceptors. Thus, each of the Ni(2+)-sensitive Ca(2+) channels contributes to specific features of the b-wave response. CONCLUSION Both high-affinity Ni(2+)-sensitive Ca(2+) channels contribute to transretinal signalling. Based on the results from the double knockout mice, additional targets for NiCl(2) must contribute to transretinal signalling, which will be most important for the structurally similar physiologically more important heavy metal cation Zn(2+).
Collapse
Affiliation(s)
- Maged Alnawaiseh
- Institute of Neurophysiology, University of Cologne, Koeln, Germany
| | - Walid Albanna
- Institute of Neurophysiology, University of Cologne, Koeln, Germany
| | - Chien‐Chang Chen
- Molecular Physiology and Biophysics, The University of Iowa College of Medicine, Iowa City, Iowa, USA
| | - Kevin P. Campbell
- Departments of Molecular Physiology and Biophysics, Neurology, and Internal Medicine, and the Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, USA
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Koeln, Germany
- Center of Molecular Medicine Cologne (CMMC), University of Cologne, Koeln, Germany
| | - Matthias Lüke
- Institute of Neurophysiology, University of Cologne, Koeln, Germany
- University Eye Hospital, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Toni Schneider
- Institute of Neurophysiology, University of Cologne, Koeln, Germany
- Departments of Molecular Physiology and Biophysics, Neurology, and Internal Medicine, and the Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
32
|
Jones SM, Palmer MJ. Pharmacological analysis of the activation and receptor properties of the tonic GABA(C)R current in retinal bipolar cell terminals. PLoS One 2011; 6:e24892. [PMID: 21949779 PMCID: PMC3174224 DOI: 10.1371/journal.pone.0024892] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/22/2011] [Indexed: 11/29/2022] Open
Abstract
GABAergic inhibition in the central nervous system (CNS) can occur via rapid, transient postsynaptic currents and via a tonic increase in membrane conductance, mediated by synaptic and extrasynaptic GABAA receptors (GABAARs) respectively. Retinal bipolar cells (BCs) exhibit a tonic current mediated by GABACRs in their axon terminal, in addition to synaptic GABAAR and GABACR currents, which strongly regulate BC output. The tonic GABACR current in BC terminals (BCTs) is not dependent on vesicular GABA release, but properties such as the alternative source of GABA and the identity of the GABACRs remain unknown. Following a recent report that tonic GABA release from cerebellar glial cells is mediated by Bestrophin 1 anion channels, we have investigated their role in non-vesicular GABA release in the retina. Using patch-clamp recordings from BCTs in goldfish retinal slices, we find that the tonic GABACR current is not reduced by the anion channel inhibitors NPPB or flufenamic acid but is reduced by DIDS, which decreases the tonic current without directly affecting GABACRs. All three drugs also exhibit non-specific effects including inhibition of GABA transporters. GABACR ρ subunits can form homomeric and heteromeric receptors that differ in their properties, but BC GABACRs are thought to be ρ1-ρ2 heteromers. To investigate whether GABACRs mediating tonic and synaptic currents may differ in their subunit composition, as is the case for GABAARs, we have examined the effects of two antagonists that show partial ρ subunit selectivity: picrotoxin and cyclothiazide. Tonic and synaptic GABACR currents were differentially affected by both drugs, suggesting that a population of homomeric ρ1 receptors contributes to the tonic current. These results extend our understanding of the multiple forms of GABAergic inhibition that exist in the CNS and contribute to visual signal processing in the retina.
Collapse
Affiliation(s)
- Stefanie M. Jones
- Neuroscience Group, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Mary J. Palmer
- Neuroscience Group, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Abstract
Ionotropic GABA receptors (GABA(A) and GABA(C)) belong to the Cys-loop receptor family of ligand-gated ion channels. GABA(C) receptors are highly expressed in the retina, mainly localized at the axon terminals of bipolar cells. Ascorbic acid, an endogenous redox agent, modulates the function of diverse proteins, and basal levels of ascorbic acid in the retina are very high. However, the effect of ascorbic acid on retinal GABA receptors has not been studied. Here we show that the function of GABA(C) and GABA(A) receptors is regulated by ascorbic acid. Patch-clamp recordings from bipolar cell terminals in goldfish retinal slices revealed that GABA(C) receptor-mediated currents activated by tonic background levels of extracellular GABA, and GABA(C) currents elicited by local GABA puffs, are both significantly enhanced by ascorbic acid. In addition, a significant rundown of GABA puff-evoked currents was observed in the absence of ascorbic acid. GABA-evoked Cl(-) currents mediated by homomeric ρ(1) GABA(C) receptors expressed in Xenopus laevis oocytes were also potentiated by ascorbic acid in a concentration-dependent, stereo-specific, reversible, and voltage-independent manner. Studies involving the chemical modification of sulfhydryl groups showed that the two Cys-loop cysteines and histidine 141, all located in the ρ(1) subunit extracellular domain, each play a key role in the modulation of GABA(C) receptors by ascorbic acid. Additionally, we show that retinal GABA(A) IPSCs and heterologously expressed GABA(A) receptor currents are similarly augmented by ascorbic acid. Our results suggest that ascorbic acid may act as an endogenous agent capable of potentiating GABAergic neurotransmission in the CNS.
Collapse
|
34
|
Abstract
PURPOSE To investigate changes in cytokine levels in tears of type 2 diabetics with or without retinopathy. METHODS Tears were collected from 15 type 2 diabetics without retinopathy (DNR), 15 patients with retinopathy (DR), and 15 age and gender matched non-diabetic controls. Tear concentrations of 27 cytokines were measured by multiplex bead immunoassay. Cytokine differences between groups, ratios of type-1 T helper (Th1)/type-2 T helper (Th2) cytokines and anti-angiogenic/pro-angiogenic cytokines were analyzed statistically. RESULTS The most abundant cytokine detected in tears was interferon-induced protein-10 (IP-10). In comparison with controls, IP-10 and monocyte chemoattracant protein-1 (MCP-1) levels were significantly elevated in DR (p=0.016 and 0.036, respectively) and DNR groups (p=0.021 and 0.026, respectively). Interleukin-1 (IL-1) receptor antagonist (IL-1ra) levels were significantly increased in DNR (p=0.016). Th1/Th2 cytokines interferon-gamma (IFN-γ)/IL-5 and IL-2/IL-5 ratios were significantly increased in DR compared to controls (p=0.037 and 0.031, respectively). Anti-angiogenic/angiogenic cytokines IFN-γ/MCP-1 and IL-4/MCP-1 ratios in DR and DNR were significantly decreased compared to controls (p<0.05). IL-4/IL-8 and IL-12p70/IL-8 ratios were also significantly decreased in DR compared to controls (p=0.02 and 0.045, respectively). No significant correlation was demonstrated between tear cytokine concentrations and glycosylated hemoglobin (HbA1c) or fasting plasma glucose (FPG). CONCLUSIONS Diabetic tears exhibited elevated levels of IP-10 and MCP-1. The Th1/Th2 cytokine balance may shift to a predominantly Th1 state in DR patients. Pro-angiogenic cytokines are more highly represented than anti-angiogenic cytokines in the tears of diabetic patients.
Collapse
|
35
|
Abstract
AbstractZebrafish are an existing model for genetic and developmental studies due to their rapid external development and transparent embryos, which allow easy manipulation and observation of early developmental stages. The application of the zebrafish model to vision research has allowed for examination of retinal development and the characteristics of different retinal cell types, including bipolar cells. In particular, bipolar cell development, including differentiation, maturation, and gene expression, has been documented, as has physiological properties, such as voltage- and ligand-gated currents, and neurotransmitter receptor and ion channel expression. Mutant strains and transgenic lines have been used to document how bipolar cell connections and/or development may be altered, and toxicological studies examining how environmental factors may impact bipolar cell activity have been performed. The purpose of this paper was to review the existing literature on zebrafish bipolar cells, to provide a comprehensive overview of current information pertaining to this retinal cell type.
Collapse
|
36
|
Abstract
Gap junctions are frequently observed in the adult vertebrate retina. It has been shown that gap junctions function as passive electrotonic pathways and play various roles, such as noise reduction, synchronization of electrical activities, regulation of the receptive field size, and transmission of rod signals to cone pathways. The presence of gap junctions between bipolar cells has been reported in various species but their functions are not known. In the present study, we applied dual whole-cell clamp techniques to the adult goldfish retina to elucidate the functions of gap junctions between ON-type bipolar cells with a giant axon terminal (Mb1-BCs). Electrophysiological and immunohistochemical experiments revealed that Mb1-BCs were coupled with each other through gap junctions that were located at the distal dendrites. The coupling conductance between Mb1-BCs under light-adapted conditions was larger than that under dark-adapted conditions. The gap junctions showed neither rectification nor voltage dependence, and behaved as a low-pass filter. Mb1-BCs could generate Ca(2+) spikes in response to depolarization, especially under dark-adapted conditions. The Ca(2+) spike evoked electrotonic depolarization through gap junctions in neighboring Mb1-BCs, and the depolarization in turn could trigger Ca(2+) spikes with a time lag. A brief depolarizing pulse applied to an Mb1-BC evoked a long-lasting EPSC in the postsynaptic ganglion cell. The EPSC was shortened in duration when gap junctions were pharmacologically or mechanically impaired. These results suggest that the spread of Ca(2+) spikes through gap junctions between bipolar cells may play a key role in lateral interactions in the adult retina.
Collapse
|
37
|
Tekmen M, Gleason E. Multiple Ca2+-dependent mechanisms regulate L-type Ca2+ current in retinal amacrine cells. J Neurophysiol 2010; 104:1849-66. [PMID: 20685929 DOI: 10.1152/jn.00031.2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Understanding the regulation of L-type voltage-gated Ca(2+) current is an important component of elucidating the signaling capabilities of retinal amacrine cells. Here we ask how the cytosolic Ca(2+) environment and the balance of Ca(2+)-dependent effectors shape native L-type Ca(2+) channel function in these cells. To achieve this, whole cell voltage clamp recordings were made from cultured amacrine cells under conditions that address the contribution of mitochondrial Ca(2+) uptake (MCU), Ca(2+)/calmodulin (CaM)-dependent channel inactivation (CDI), protein kinase A (PKA), and Ca(2+)-induced Ca(2+) release (CICR). Under control conditions, repeated activation of the L-type channels produces a progressive enhancement of the current. Inhibition of MCU causes a reduction in the Ca(2+) current amplitude that is dependent on Ca(2+) influx as well as cytosolic Ca(2+) buffering, consistent with CDI. Including the Ca(2+) buffer bis-(o-aminophenoxy)-N,N,N',N'-tetraacetic acid (BAPTA) internally can shift the balance between enhancement and inhibition such that inhibition of MCU can enhance the current. Inhibition of PKA can remove the enhancing effect of BAPTA suggesting that cyclic AMP-dependent phosphorylation is involved. Inhibition of CaM suppresses CDI but spares the enhancement, consistent with the substantially higher sensitivity of the Ca(2+)-sensitive adenylate cyclase 1 (AC1) to Ca(2+)/CaM. Inhibition of the ryanodine receptor reduces the current amplitude, suggesting that CICR might normally amplify the activation of AC1 and stimulation of PKA activity. These experiments reveal that the amplitude of L-type Ca(2+) currents in retinal amacrine cells are both positively and negatively regulated by Ca(2+)-dependent mechanisms.
Collapse
Affiliation(s)
- Merve Tekmen
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | |
Collapse
|
38
|
Chávez AE, Grimes WN, Diamond JS. Mechanisms underlying lateral GABAergic feedback onto rod bipolar cells in rat retina. J Neurosci 2010; 30:2330-9. [PMID: 20147559 PMCID: PMC2836865 DOI: 10.1523/jneurosci.5574-09.2010] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 12/18/2009] [Accepted: 12/24/2009] [Indexed: 11/21/2022] Open
Abstract
GABAergic feedback inhibition from amacrine cells shapes visual signaling in the inner retina. Rod bipolar cells (RBCs), ON-sensitive cells that depolarize in response to light increments, receive reciprocal GABAergic feedback from A17 amacrine cells and additional GABAergic inputs from other amacrine cells located laterally in the inner plexiform layer. The circuitry and synaptic mechanisms underlying lateral GABAergic inhibition of RBCs are poorly understood. A-type and rho-subunit-containing (C-type) GABA receptors (GABA(A)Rs and GABA(C)Rs) mediate both forms of inhibition, but their relative activation during synaptic transmission is unclear, and potential interactions between adjacent reciprocal and lateral synapses have not been explored. Here, we recorded from RBCs in acute slices of rat retina and isolated lateral GABAergic inhibition by pharmacologically ablating A17 amacrine cells. We found that amacrine cells providing lateral GABAergic inhibition to RBCs receive excitatory synaptic input mostly from ON bipolar cells via activation of both Ca(2+)-impermeable and Ca(2+)-permeable AMPA receptors (CP-AMPARs) but not NMDA receptors (NMDARs). Voltage-gated Ca(2+) (Ca(v)) channels mediate the majority of Ca(2+) influx that triggers GABA release, although CP-AMPARs contribute a small component. The intracellular Ca(2+) signal contributing to transmitter release is amplified by Ca(2+)-induced Ca(2+) release from intracellular stores via activation of ryanodine receptors. Furthermore, lateral nonreciprocal feedback is mediated primarily by GABA(C)Rs that are activated independently from receptors mediating reciprocal feedback inhibition. These results illustrate numerous physiological differences that distinguish GABA release at reciprocal and lateral synapses, indicating complex, pathway-specific modulation of RBC signaling.
Collapse
Affiliation(s)
- Andrés E. Chávez
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701
| | - William N. Grimes
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701
| | - Jeffrey S. Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701
| |
Collapse
|
39
|
Siapich SA, Banat M, Albanna W, Hescheler J, Lüke M, Schneider T. Antagonists of ionotropic gamma-aminobutyric acid receptors impair the NiCl2-mediated stimulation of the electroretinogram b-wave amplitude from the isolated superfused vertebrate retina. Acta Ophthalmol 2009; 87:854-65. [PMID: 20002018 DOI: 10.1111/j.1755-3768.2008.01387.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE NiCl(2) (15 microM) stimulates the electroretinogram (ERG) b-wave amplitude of vertebrate retina up to 1.5-fold through its blocking of E/R-type voltage-gated Ca(2+) channels. Assuming that such an increase is mediated by blocking the release of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) via ionotropic GABA receptors, we tested the effect of both GABA itself and GABA-receptor antagonists such as (-)bicuculline (1.51-fold increase) and (1,2,5,6-tetrahydropyridin-4-yl)methylphosphinic acid (TPMPA; 1.46-fold increase) on the b-wave amplitude. METHODS Recording of the transretinal potentials from the isolated bovine retina. RESULTS GABA (100 microM) reduced the b-wave amplitude only when NiCl(2) (15 microM) was applied first. Each antagonist applied on its own stimulated the b-wave amplitude only partially: subsequent NiCl(2) superfusion caused a small but additional increase, leading to a 1.69- and a 1.88-fold total increase of the amplitude by Ni(2+) plus (-)bicuculline or Ni(2+) plus TPMPA, respectively. Only the application of both antagonists in combination, before superfusing low NiCl(2) (15 microM), completely prevented subsequent stimulation by NiCl(2) with a similar 1.90-fold total increase of b-wave amplitude. Those retina segments that did not respond to NiCl(2) could not be stimulated by (-)bicuculline and vice versa. CONCLUSION The stimulatory effect of NiCl(2) on the ERG b-wave amplitude is mainly, but not only, mediated by a NiCl(2)-sensitive, Ca(v)2.3-triggered GABA release acting through ionotropic GABA-A and GABA-C receptors.
Collapse
|
40
|
Crousillac S, Colonna J, McMains E, Dewey JS, Gleason E. Sphingosine-1-phosphate elicits receptor-dependent calcium signaling in retinal amacrine cells. J Neurophysiol 2009; 102:3295-309. [PMID: 19776367 DOI: 10.1152/jn.00119.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Evidence is emerging indicating that sphingosine-1-phosphate (S1P) participates in signaling in the retina. To determine whether S1P might be involved in signaling in the inner retina specifically, we examine the effects of this sphingolipid on cultured retinal amacrine cells. Whole cell voltage-clamp recordings reveal that S1P activates a cation current that is dependent on signaling through G(i) and phospholipase C. These observations are consistent with the involvement of members of the S1P receptor family of G-protein-coupled receptors in the production of the current. Immunocytochemistry and PCR amplification provide evidence for the expression of S1P1R and S1P3R in amacrine cells. The receptor-mediated channel activity is shown to be highly sensitive to blockade by lanthanides consistent with the behavior of transient receptor potential canonical (TRPC) channels. PCR products amplified from amacrine cells reveal that TRPCs 1 and 3-7 channel subunits have the potential to be expressed. Because TRPC channels provide a Ca(2+) entry pathway, we asked whether S1P caused cytosolic Ca(2+) elevations in amacrine cells. We show that S1P-dependent Ca(2+) elevations do occur in these cells and that they might be mediated by S1P1R and S1P3R. The Ca(2+) elevations are partially due to release from internal stores, but the largest contribution is from influx across the plasma membrane. The effect of inhibition of sphingosine kinase suggests that the production of cytosolic S1P underlies the sustained nature of the Ca(2+) elevations. Elucidation of the downstream effects of these signals will provide clues to the role of S1P in regulating inner retinal function.
Collapse
Affiliation(s)
- Scott Crousillac
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | |
Collapse
|
41
|
Fahrenfort I, Steijaert M, Sjoerdsma T, Vickers E, Ripps H, van Asselt J, Endeman D, Klooster J, Numan R, ten Eikelder H, von Gersdorff H, Kamermans M. Hemichannel-mediated and pH-based feedback from horizontal cells to cones in the vertebrate retina. PLoS One 2009; 4:e6090. [PMID: 19564917 PMCID: PMC2699542 DOI: 10.1371/journal.pone.0006090] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 05/20/2009] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Recent studies designed to identify the mechanism by which retinal horizontal cells communicate with cones have implicated two processes. According to one account, horizontal cell hyperpolarization induces an increase in pH within the synaptic cleft that activates the calcium current (Ca(2+)-current) in cones, enhancing transmitter release. An alternative account suggests that horizontal cell hyperpolarization increases the Ca(2+)-current to promote transmitter release through a hemichannel-mediated ephaptic mechanism. METHODOLOGY/PRINCIPAL FINDINGS To distinguish between these mechanisms, we interfered with the pH regulating systems in the retina and studied the effects on the feedback responses of cones and horizontal cells. We found that the pH buffers HEPES and Tris partially inhibit feedback responses in cones and horizontal cells and lead to intracellular acidification of neurons. Application of 25 mM acetate, which does not change the extracellular pH buffer capacity, does lead to both intracellular acidification and inhibition of feedback. Because intracellular acidification is known to inhibit hemichannels, the key experiment used to test the pH hypothesis, i.e. increasing the extracellular pH buffer capacity, does not discriminate between a pH-based feedback system and a hemichannel-mediated feedback system. To test the pH hypothesis in a manner independent of artificial pH-buffer systems, we studied the effect of interfering with the endogenous pH buffer, the bicarbonate/carbonic anhydrase system. Inhibition of carbonic anhydrase allowed for large changes in pH in the synaptic cleft of bipolar cell terminals and cone terminals, but the predicted enhancement of the cone feedback responses, according to the pH-hypothesis, was not observed. These experiments thus failed to support a proton mediated feedback mechanism. The alternative hypothesis, the hemichannel-mediated ephaptic feedback mechanism, was therefore studied experimentally, and its feasibility was buttressed by means of a quantitative computer model of the cone/horizontal cell synapse. CONCLUSION We conclude that the data presented in this paper offers further support for physiologically relevant ephaptic interactions in the retina.
Collapse
Affiliation(s)
- Iris Fahrenfort
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Marvin Steijaert
- Department of Biomedical Engineering, Biomodeling and Bioinformatics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Trijntje Sjoerdsma
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Evan Vickers
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Harris Ripps
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Jorrit van Asselt
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Duco Endeman
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Jan Klooster
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Robert Numan
- Department of Medical Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Huub ten Eikelder
- Department of Biomedical Engineering, Biomodeling and Bioinformatics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Henrique von Gersdorff
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Maarten Kamermans
- Research Unit Retinal Signal Processing, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Department of Neurogenetics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
42
|
Thiers FA, Nadol JB, Liberman MC. Reciprocal synapses between outer hair cells and their afferent terminals: evidence for a local neural network in the mammalian cochlea. J Assoc Res Otolaryngol 2008; 9:477-89. [PMID: 18688678 PMCID: PMC2580814 DOI: 10.1007/s10162-008-0135-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 07/14/2008] [Indexed: 10/21/2022] Open
Abstract
Cochlear outer hair cells (OHCs) serve both as sensory receptors and biological motors. Their sensory function is poorly understood because their afferent innervation, the type-II spiral ganglion cell, has small unmyelinated axons and constitutes only 5% of the cochlear nerve. Reciprocal synapses between OHCs and their type-II terminals, consisting of paired afferent and efferent specialization, have been described in the primate cochlea. Here, we use serial and semi-serial-section transmission electron microscopy to quantify the nature and number of synaptic interactions in the OHC area of adult cats. Reciprocal synapses were found in all OHC rows and all cochlear frequency regions. They were more common among third-row OHCs and in the apical half of the cochlea, where 86% of synapses were reciprocal. The relative frequency of reciprocal synapses was unchanged following surgical transection of the olivocochlear bundle in one cat, confirming that reciprocal synapses were not formed by efferent fibers. In the normal ear, axo-dendritic synapses between olivocochlear terminals and type-II terminals and/or dendrites were as common as synapses between olivocochlear terminals and OHCs, especially in the first row, where, on average, almost 30 such synapses were seen in the region under a single OHC. The results suggest that a complex local neuronal circuitry in the OHC area, formed by the dendrites of type-II neurons and modulated by the olivocochlear system, may be a fundamental property of the mammalian cochlea, rather than a curiosity of the primate ear. This network may mediate local feedback control of, and bidirectional communication among, OHCs throughout the cochlear spiral.
Collapse
Affiliation(s)
- Fabio A. Thiers
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, 243 Charles Street, Boston, MA USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02114 USA
- Program in Speech and Hearing Bioscience and Technology, Division of Health Science and Technology, Harvard & MIT, Cambridge, MA USA
| | - Joseph B. Nadol
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02114 USA
- Program in Speech and Hearing Bioscience and Technology, Division of Health Science and Technology, Harvard & MIT, Cambridge, MA USA
| | - M. Charles Liberman
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, 243 Charles Street, Boston, MA USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02114 USA
- Program in Speech and Hearing Bioscience and Technology, Division of Health Science and Technology, Harvard & MIT, Cambridge, MA USA
| |
Collapse
|
43
|
Xu Y, Sulaiman P, Feddersen RM, Liu J, Smith RG, Vardi N. Retinal ON bipolar cells express a new PCP2 splice variant that accelerates the light response. J Neurosci 2008; 28:8873-84. [PMID: 18768681 PMCID: PMC2590633 DOI: 10.1523/jneurosci.0812-08.2008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2008] [Revised: 07/02/2008] [Accepted: 07/21/2008] [Indexed: 11/21/2022] Open
Abstract
PCP2, a member of the GoLoco domain-containing family, is present exclusively in cerebellar Purkinje cells and retinal ON bipolar cells. Its function in these tissues is unknown. Biochemical and expression system studies suggest that PCP2 is a guanine nucleotide dissociation inhibitor, although a guanine nucleotide exchange factor has also been suggested. Here, we studied the function of PCP2 in ON bipolar cells because their light response depends on Galpha(o1), which is known to interact with PCP2. We identified a new splice variant of PCP2 (Ret-PCP2) and localized it to rod bipolar and ON cone bipolar cells. Electroretinogram recordings from PCP2-null mice showed a normal a-wave but a slower falling phase of the b-wave (generated by the activity of ON bipolar cells) relative to the wild type. Whole-cell recordings from rod bipolar cells showed, both under Ames medium and after blocking GABA(A/C) and glycine receptors, that PCP2-null rod bipolar cells were more depolarized than wild-type cells with greater inward current when clamped to -60 mV. Also under both conditions, the rise time of the response to intense light was slower by 28% (Ames) and 44% (inhibitory blockers) in the null cells. Under Ames medium, we also observed >30% longer decay time in the PCP2-null rod bipolar cells. We conclude that PCP2 facilitates cation channels closure in the dark, shortens the rise time of the light response directly, and accelerates the decay time indirectly via the inhibitory network. These data can most easily be explained if PCP2 serves as a guanine nucleotide exchange factor.
Collapse
Affiliation(s)
- Ying Xu
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Chávez AE, Diamond JS. Diverse mechanisms underlie glycinergic feedback transmission onto rod bipolar cells in rat retina. J Neurosci 2008; 28:7919-28. [PMID: 18667624 PMCID: PMC2575372 DOI: 10.1523/jneurosci.0784-08.2008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 06/18/2008] [Accepted: 06/20/2008] [Indexed: 11/21/2022] Open
Abstract
Synaptic inhibition shapes visual signaling in the inner retina, but the physiology of most amacrine cells, the interneurons that mediate this inhibition, is poorly understood. Discerning the function of most individual amacrine cell types is a daunting task, because few molecular or morphological markers specifically distinguish between approximately two dozen different amacrine cell types. Here, we examine a functional subset of amacrine cells by pharmacologically isolating glycinergic inhibition and evoking feedback IPSCs in a single cell type, the rod bipolar cell (RBC), with brief glutamate applications in the inner plexiform layer. We find that glycinergic amacrine cells innervating RBCs receive excitatory inputs from ON and OFF bipolar cells primarily via NMDA receptors (NMDARs) and Ca2+-impermeable AMPA-type glutamate receptors. Glycine release from amacrine cells is triggered by Ca2+ influx through both voltage-gated Ca2+ (Ca(v)) channels and NMDARs. These intracellular Ca2+signals are amplified by Ca2+-induced Ca2+ release via both ryanodine and IP3 receptors, which are activated independently by Ca2+ influx through Ca(v) channels and NMDARs, respectively. Glycinergic feedback signaling depends strongly, although not completely, on voltage-gated Na+ channels, and the spatial extent of feedback inhibition is expanded by gap junction connections between glycinergic amacrine cells. These results indicate that a diversity of mechanisms underlie glycinergic feedback inhibition onto RBCs, yet they highlight several physiological themes that appear to distinguish amacrine cell function.
Collapse
Affiliation(s)
- Andrés E Chávez
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701, USA.
| | | |
Collapse
|
45
|
Sen M, McMains E, Gleason E. Local influence of mitochondrial calcium transport in retinal amacrine cells. Vis Neurosci 2007; 24:663-78. [PMID: 17697441 DOI: 10.1017/s0952523807070551] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 05/26/2007] [Indexed: 11/07/2022]
Abstract
Ca2+-dependent synaptic transmission from retinal amacrine cells is thought to be initiated locally at dendritic processes. Hence, understanding the spatial and temporal impact of Ca2+ transport is fundamental to understanding how amacrine cells operate. Here, we provide the first examination of the local effects of mitochondrial Ca2+ transport in neuronal processes. By combining mitochondrial localization with measurements of cytosolic Ca2+, the local impacts of mitochondrial Ca2+ transport for two types of Ca2+ signals were investigated. Disruption of mitochondrial Ca2+ uptake with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) produces cytosolic Ca2+ elevations. The amplitudes of these elevations decline with distance from mitochondria suggesting that they are related to mitochondrial Ca2+ transport. The time course of the FCCP-dependent Ca2+ elevations depend on the availability of ER Ca2+ and we provide evidence that Ca2+ is released primarily via nearby ryanodine receptors. These results indicate that interactions between the ER and mitochondria influence cytosolic Ca2+ in amacrine cell processes and cell bodies. We also demonstrate that the durations of glutamate-dependent Ca2+ elevations are dependent on their proximity to mitochondria in amacrine cell processes. Consistent with this observation, disruption of mitochondrial Ca2+ transport alters the duration of glutamate-dependent Ca2+ elevations near mitochondria but not at sites more than 10 microm away. These results indicate that mitochondria influence local Ca2+-dependent signaling in amacrine cell processes.
Collapse
Affiliation(s)
- Madhumita Sen
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | | | |
Collapse
|
46
|
Li GL, Vigh J, von Gersdorff H. Short-term depression at the reciprocal synapses between a retinal bipolar cell terminal and amacrine cells. J Neurosci 2007; 27:7377-85. [PMID: 17626198 PMCID: PMC6672600 DOI: 10.1523/jneurosci.0410-07.2007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Visual adaptation is thought to occur partly at retinal synapses that are subject to plastic changes. However, the locus and properties of this plasticity are not well known. Here, we studied short-term plasticity at the reciprocal synapse between bipolar cell terminals and amacrine cells in goldfish retinal slices. Depolarization of a single bipolar cell terminal for 100 ms triggers the release of glutamate onto amacrine cell processes, which in turn leads to GABAergic feedback from amacrine cells onto the same terminal. We find that this release of GABA undergoes paired-pulse depression (PPD) that recovers in <1 min (single exponential time constant, tau approximately = 12 s). This disynaptic PPD is independent of mGluR-mediated plasticity and depletion of glutamatergic synaptic vesicle pools, because exocytosis assayed via capacitance jumps (deltaC(m)) recovered completely after 10 s (tau approximately = 2 s). Fast application of GABA (10 mM) onto outside-out patches excised from bipolar cell terminals showed that the recovery of GABA(A) receptors from desensitization depends on the duration of the application [fast recovery (<2 s) for short applications; slow (tau approximately = 12 s) for prolonged applications]. We thus blocked GABA(A) receptors and retested the GABAergic response mediated by nondesensitizing GABA(C) receptors to two rapid glutamate puffs onto the bipolar cell terminal. These responses consistently displayed PPD. Furthermore, blocking AMPA-receptor desensitization with cyclothiazide, or evoking GABA release with NMDA receptors, did not reduce PPD. We thus suggest that depletion of synaptic vesicle pools in GABAergic amacrine cells is a major contributor to PPD.
Collapse
Affiliation(s)
- Geng-Lin Li
- The Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Jozsef Vigh
- The Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | | |
Collapse
|
47
|
Palmer MJ. Functional segregation of synaptic GABAA and GABAC receptors in goldfish bipolar cell terminals. J Physiol 2006; 577:45-53. [PMID: 17008372 PMCID: PMC2000669 DOI: 10.1113/jphysiol.2006.119560] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 09/25/2006] [Indexed: 11/08/2022] Open
Abstract
The transmission of light responses to retinal ganglion cells is regulated by inhibitory input from amacrine cells to bipolar cell (BC) synaptic terminals. GABA(A) and GABA(C) receptors in BC terminals mediate currents with different kinetics and are likely to have distinct functions in limiting BC output; however, the synaptic properties and localization of the receptors are currently poorly understood. By recording endogenous GABA receptor currents directly from BC terminals in goldfish retinal slices, I show that spontaneous GABA release activates rapid GABA(A) receptor miniature inhibitory postsynaptic currents (mIPSCs) (predominant decay time constant (tau(decay)), 1.0 ms) in addition to a tonic GABA(C) receptor current. The GABA(C) receptor antagonist (1,2,5,6-tetrahydropyridin-4-yl)methylphosphinic acid (TPMPA) has no effect on the amplitude or kinetics of the rapid GABA(A) mIPSCs. In addition, inhibition of the GAT-1 GABA transporter, which strongly regulates GABA(C) receptor currents in BC terminals, fails to reveal a GABA(C) component in the mIPSCs. These data suggest that GABA(A) and GABA(C) receptors are highly unlikely to be synaptically colocalized. Using non-stationary noise analysis of the mIPSCs, I estimate that GABA(A) receptors in BC terminals have a single-channel conductance (gamma) of 17 pS and that an average of just seven receptors mediates a quantal event. From noise analysis of the tonic current, GABA(C) receptor gamma is estimated to be 4 pS. Identified GABA(C) receptor mIPSCs exhibit a slow decay (tau(decay), 54 ms) and are mediated by approximately 42 receptors. The distinct properties and localization of synaptic GABA(A) and GABA(C) receptors in BC terminals are likely to facilitate their specific roles in regulating the transmission of light responses in the retina.
Collapse
Affiliation(s)
- Mary J Palmer
- Neuroscience Group, Institute for Science and Technology in Medicine, Keele University, Keele, ST5 5BG, UK.
| |
Collapse
|
48
|
Chávez AE, Singer JH, Diamond JS. Fast neurotransmitter release triggered by Ca influx through AMPA-type glutamate receptors. Nature 2006; 443:705-8. [PMID: 17036006 DOI: 10.1038/nature05123] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 07/31/2006] [Indexed: 11/08/2022]
Abstract
Feedback inhibition at reciprocal synapses between A17 amacrine cells and rod bipolar cells (RBCs) shapes light-evoked responses in the retina. Glutamate-mediated excitation of A17 cells elicits GABA (gamma-aminobutyric acid)-mediated inhibitory feedback onto RBCs, but the mechanisms that underlie GABA release from the dendrites of A17 cells are unknown. If, as observed at all other synapses studied, voltage-gated calcium channels (VGCCs) couple membrane depolarization to neurotransmitter release, feedforward excitatory postsynaptic potentials could spread through A17 dendrites to elicit 'surround' feedback inhibitory transmission at neighbouring synapses. Here we show, however, that GABA release from A17 cells in the rat retina does not depend on VGCCs or membrane depolarization. Instead, calcium-permeable AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors (AMPARs), activated by glutamate released from RBCs, provide the calcium influx necessary to trigger GABA release from A17 cells. The AMPAR-mediated calcium signal is amplified by calcium-induced calcium release (CICR) from intracellular calcium stores. These results describe a fast synapse that operates independently of VGCCs and membrane depolarization and reveal a previously unknown form of feedback inhibition within a neural circuit.
Collapse
Affiliation(s)
- Andrés E Chávez
- Synaptic Physiology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-3701, USA
| | | | | |
Collapse
|
49
|
Wersinger E, Schwab Y, Sahel JA, Rendon A, Pow DV, Picaud S, Roux MJ. The glutamate transporter EAAT5 works as a presynaptic receptor in mouse rod bipolar cells. J Physiol 2006; 577:221-34. [PMID: 16973698 PMCID: PMC2000664 DOI: 10.1113/jphysiol.2006.118281] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Membrane neurotransmitter transporters control the concentration of their substrate in the synaptic clefts, through the thermodynamic coupling of uptake to the movement of Na(+) and other ions. In addition, excitatory amino acid transporters (EAAT) have a Cl(-) conductance which is gated by the joint binding of Na(+) and glutamate, but thermodynamically uncoupled to the flux of glutamate. This conductance is particularly large in the retina-specific EAAT5 isoform. In the mouse retina, we located EAAT5 in both cone and rod photoreceptor terminals and in axon terminals of rod bipolar cells. In these later cells, application of glutamate on the axon terminal evoked a current that reversed at E(Cl), was insensitive to bicuculline, TPMPA, strychnine, dl-AP5, CNQX and MCPG, but blocked by the glutamate transporter inhibitor dl-tBOA. Furthermore, short depolarizations of the bipolar cells evoked a dl-tBOA and Cd(2+)-sensitive current whose amplitude was comparable to the glutamate-evoked current. Its kinetics indicated that EAAT5 was located close to the glutamate release site. For 2 ms depolarizations evoking maximal responses, the EAAT5-mediated current carried between 2 and 8 times more charge as an average inhibitory GABA or glycine postsynaptic current received spontaneously from amacrine cells, with 10 mm or 0.5 mm intracellular EGTA, respectively. In conditions for which reciprocal inhibition could be monitored, the charge carried by the EAAT5 current was 1.5 times larger than the one carried by the inhibitory postsynaptic currents received from amacrine cells. These results indicate that EAAT5 acts as a major inhibitory presynaptic receptor at mammalian rod bipolar cell axon terminals. This feedback mechanism could control glutamate release at the ribbon synapses of a non-spiking neuron and increase the temporal contrast in the rod photoreceptor pathway.
Collapse
Affiliation(s)
- Eric Wersinger
- Laboratory of Cellular and Molecular Physiopathology of the Retina, National Institute for Health and Medical Research (INSERM Unité 592), Université Pierre et Marie Curie-Paris6, Paris, France
| | | | | | | | | | | | | |
Collapse
|