1
|
Stevens-Sostre WA, Hoon M. Cellular and Molecular Mechanisms Regulating Retinal Synapse Development. Annu Rev Vis Sci 2024; 10:377-402. [PMID: 39292551 PMCID: PMC12022667 DOI: 10.1146/annurev-vision-102122-105721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Synapse formation within the retinal circuit ensures that distinct neuronal types can communicate efficiently to process visual signals. Synapses thus form the core of the visual computations performed by the retinal circuit. Retinal synapses are diverse but can be broadly categorized into multipartner ribbon synapses and 1:1 conventional synapses. In this article, we review our current understanding of the cellular and molecular mechanisms that regulate the functional establishment of mammalian retinal synapses, including the role of adhesion proteins, synaptic proteins, extracellular matrix and cytoskeletal-associated proteins, and activity-dependent cues. We outline future directions and areas of research that will expand our knowledge of these mechanisms. Understanding the regulators moderating synapse formation and function not only reveals the integrated developmental processes that establish retinal circuits, but also divulges the identity of mechanisms that could be engaged during disease and degeneration.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Sigulinsky CL, Pfeiffer RL, Jones BW. Retinal Connectomics: A Review. Annu Rev Vis Sci 2024; 10:263-291. [PMID: 39292552 DOI: 10.1146/annurev-vision-102122-110414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
The retina is an ideal model for understanding the fundamental rules for how neural networks are constructed. The compact neural networks of the retina perform all of the initial processing of visual information before transmission to higher visual centers in the brain. The field of retinal connectomics uses high-resolution electron microscopy datasets to map the intricate organization of these networks and further our understanding of how these computations are performed by revealing the fundamental topologies and allowable networks behind retinal computations. In this article, we review some of the notable advances that retinal connectomics has provided in our understanding of the specific cells and the organization of their connectivities within the retina, as well as how these are shaped in development and break down in disease. Using these anatomical maps to inform modeling has been, and will continue to be, instrumental in understanding how the retina processes visual signals.
Collapse
Affiliation(s)
- Crystal L Sigulinsky
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, USA;
| | - Rebecca L Pfeiffer
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, USA;
| | - Bryan William Jones
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, 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. 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
|
4
|
Park SJ, Lei W, Pisano J, Orpia A, Minehart J, Pottackal J, Hanke-Gogokhia C, Zapadka TE, Clarkson-Paredes C, Popratiloff A, Ross SE, Singer JH, Demb JB. Molecular identification of wide-field amacrine cells in mouse retina that encode stimulus orientation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.28.573580. [PMID: 38234775 PMCID: PMC10793454 DOI: 10.1101/2023.12.28.573580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Visual information processing is sculpted by a diverse group of inhibitory interneurons in the retina called amacrine cells. Yet, for most of the >60 amacrine cell types, molecular identities and specialized functional attributes remain elusive. Here, we developed an intersectional genetic strategy to target a group of wide-field amacrine cells (WACs) in mouse retina that co-express the transcription factor Bhlhe22 and the Kappa Opioid Receptor (KOR; B/K WACs). B/K WACs feature straight, unbranched dendrites spanning over 0.5 mm (∼15° visual angle) and produce non-spiking responses to either light increments or decrements. Two-photon dendritic population imaging reveals Ca 2+ signals tuned to the physical orientations of B/K WAC dendrites, signifying a robust structure-function alignment. B/K WACs establish divergent connections with multiple retinal neurons, including unexpected connections with non-orientation-tuned ganglion cells and bipolar cells. Our work sets the stage for future comprehensive investigations of the most enigmatic group of retinal neurons: WACs.
Collapse
|
5
|
Keeley PW, Trod S, Gamboa BN, Coffey PJ, Reese BE. Nfia Is Critical for AII Amacrine Cell Production: Selective Bipolar Cell Dependencies and Diminished ERG. J Neurosci 2023; 43:8367-8384. [PMID: 37775301 PMCID: PMC10711738 DOI: 10.1523/jneurosci.1099-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
The nuclear factor one (NFI) transcription factor genes Nfia, Nfib, and Nfix are all enriched in late-stage retinal progenitor cells, and their loss has been shown to retain these progenitors at the expense of later-generated retinal cell types. Whether they play any role in the specification of those later-generated fates is unknown, but the expression of one of these, Nfia, in a specific amacrine cell type may intimate such a role. Here, Nfia conditional knockout (Nfia-CKO) mice (both sexes) were assessed, finding a massive and largely selective absence of AII amacrine cells. There was, however, a partial reduction in type 2 cone bipolar cells (CBCs), being richly interconnected to AII cells. Counts of dying cells showed a significant increase in Nfia-CKO retinas at postnatal day (P)7, after AII cell numbers were already reduced but in advance of the loss of type 2 CBCs detected by P10. Those results suggest a role for Nfia in the specification of the AII amacrine cell fate and a dependency of the type 2 CBCs on them. Delaying the conditional loss of Nfia to the first postnatal week did not alter AII cell number nor differentiation, further suggesting that its role in AII cells is solely associated with their production. The physiological consequences of their loss were assessed using the ERG, finding the oscillatory potentials to be profoundly diminished. A slight reduction in the b-wave was also detected, attributed to an altered distribution of the terminals of rod bipolar cells, implicating a role of the AII amacrine cells in constraining their stratification.SIGNIFICANCE STATEMENT The transcription factor NFIA is shown to play a critical role in the specification of a single type of retinal amacrine cell, the AII cell. Using an Nfia-conditional knockout mouse to eliminate this population of retinal neurons, we demonstrate two selective bipolar cell dependencies on the AII cells; the terminals of rod bipolar cells become mis-stratified in the inner plexiform layer, and one type of cone bipolar cell undergoes enhanced cell death. The physiological consequence of this loss of the AII cells was also assessed, finding the cells to be a major contributor to the oscillatory potentials in the electroretinogram.
Collapse
Affiliation(s)
- Patrick W Keeley
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106-5060
| | - Stephanie Trod
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106-5060
| | - Bruno N Gamboa
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106-5060
| | - Pete J Coffey
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106-5060
| | - Benjamin E Reese
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106-5060
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, California 93106-5060
| |
Collapse
|
6
|
Saha A, Zuniga J, Mian K, Zhai H, Derr PJ, Hoon M, Sinha R. Regional variation in the organization and connectivity of the first synapse in the primate night vision pathway. iScience 2023; 26:108113. [PMID: 37915604 PMCID: PMC10616377 DOI: 10.1016/j.isci.2023.108113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/25/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Sensitivity of primate daylight vision varies across the visual field. This is attributed to regional variations in cone photoreceptor density and synaptic connectivity of the underlying circuitry. In contrast, we have limited understanding of how synapse organization of the primate night vision pathway changes across space. Using serial electron microscopy, we reconstructed the first synapse of the night vision pathway between rod photoreceptors and second-order neurons, at multiple locations from the central part of the primate retina, fovea, to the periphery. We find that most facets of the rod synapse connectivity vary across retinal regions. However, rod synaptic divergence and convergence patterns do not change in the same manner across locations. Moreover, patterns of rod synapse organization are tightly correlated with photoreceptor density. Such regional heterogeneities revise the connectivity diagram of the primate rod synapse which will shape synapse function and sensitivity of the night vision pathway across visual space.
Collapse
Affiliation(s)
- Aindrila Saha
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - Juan Zuniga
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Kainat Mian
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Haoshen Zhai
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Paul J. Derr
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Mrinalini Hoon
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| | - Raunak Sinha
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
7
|
Zhang K, Su A, Wang Y, Crair M. Acetylcholine Promotes Directionally Biased Glutamatergic Retinal Waves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566639. [PMID: 38014271 PMCID: PMC10680594 DOI: 10.1101/2023.11.10.566639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Spontaneous retinal waves are a critical driving force for the self-organization of the mouse visual system prior to eye-opening. Classically characterized as taking place in three distinct stages defined by their primary excitatory drive, Stage II waves during the first postnatal week are propagated through the volume transmission of acetylcholine while Stage III retinal waves during the second postnatal week depend on glutamatergic transmission from bipolar cells. However, both late Stage II and early Stage III retinal waves share a defining propagation bias toward the temporal-to-nasal direction despite developmental changes in the underlying cholinergic and glutamatergic retinal networks. Here, we leverage genetic and pharmacological manipulations to investigate the relationship between cholinergic and glutamatergic neurotransmission during the transition between Stage II and Stage III waves in vivo. We find that the cholinergic network continues to play a vital role in the propagation of waves during Stage III after the primary mode of neurotransmission changes to glutamate. In the absence of glutamatergic waves, compensatory cholinergic activity persists but lacks the propagation bias typically observed in Stage III waves. In the absence of cholinergic waves, gap junction-mediated activity typically associated with Stage I waves persists throughout the developmental window in which Stage III waves usually emerge and lacks the spatiotemporal profile of normal Stage III waves, including a temporal-to-nasal propagation bias. Finally, we show that cholinergic signaling through β2 subunit-containing nicotinic acetylcholine receptors, essential for Stage II wave propagation, is also critical for Stage III wave directionality.
Collapse
Affiliation(s)
- Kathy Zhang
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, United States
- Present address: Department of Ecology and Evolutionary Biology, Yale University, United States
| | - Ashley Su
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, United States
| | - Yixiang Wang
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, United States
| | - Michael Crair
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, United States
| |
Collapse
|
8
|
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
|
9
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557433. [PMID: 37771914 PMCID: PMC10525478 DOI: 10.1101/2023.09.12.557433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions1. 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 lineage2-6. Thus, it has been long assumed that the primary rod pathway evolved in mammals3,5-7. 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 RBCs8, both zebrafish RBC types connect with all rods and red-cones 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. This suggests that the cell types and circuit design of the primary rod pathway may have emerged before the divergence of teleost fish and amniotes (mammals, bird, reptiles). The second RBC type in zebrafish, which forms separate pathways from the first RBC type, is either lost in mammals or emerged in fish to serve yet unknown roles.
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
|
10
|
Keeley PW, Patel SS, Reese BE. Cell numbers, cell ratios, and developmental plasticity in the rod pathway of the mouse retina. J Anat 2023; 243:204-222. [PMID: 35292986 PMCID: PMC10335380 DOI: 10.1111/joa.13653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
The precise specification of cellular fate is thought to ensure the production of the correct number of neurons within a population. Programmed cell death may be an additional mechanism controlling cell number, believed to refine the proper ratio of pre- to post-synaptic neurons for a given species. Here, we consider the size of three different neuronal populations in the rod pathway of the mouse retina: rod photoreceptors, rod bipolar cells, and AII amacrine cells. Across a collection of 28 different strains of mice, large variation in the numbers of all three cell types is present. The variation in their numbers is not correlated, so that the ratio of rods to rod bipolar cells, as well as rod bipolar cells to AII amacrine cells, varies as well. Establishing connectivity between such variable pre- and post-synaptic populations relies upon plasticity that modulates process outgrowth and morphological differentiation, which we explore experimentally for both rod bipolar and AII amacrine cells in a mouse retina with elevated numbers of each cell type. While both rod bipolar dendritic and axonal arbors, along with AII lobular arbors, modulate their areal size in relation to local homotypic cell densities, the dendritic appendages of the AII amacrine cells do not. Rather, these processes exhibit a different form of plasticity, regulating the branching density of their overlapping arbors. Each form of plasticity should ensure uniformity in retinal coverage in the presence of the independent specification of afferent and target cell number.
Collapse
Affiliation(s)
- Patrick W. Keeley
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Shivam S. Patel
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Benjamin E. Reese
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
- Department of Psychological & Brain SciencesUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| |
Collapse
|
11
|
Sawant A, Saha A, Khoussine J, Sinha R, Hoon M. New insights into retinal circuits through EM connectomics: what we have learnt and what remains to be learned. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1168548. [PMID: 38983069 PMCID: PMC11182165 DOI: 10.3389/fopht.2023.1168548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/05/2023] [Indexed: 07/11/2024]
Abstract
The retinal neural circuit is intricately wired for efficient processing of visual signals. This is well-supported by the specialized connections between retinal neurons at both the functional and ultrastructural levels. Through 3D electron microscopic (EM) reconstructions of retinal neurons and circuits we have learnt much about the specificities of connections within the retinal layers including new insights into how retinal neurons establish connections and perform sophisticated visual computations. This mini-review will summarize the retinal circuitry and provide details about the novel insights EM connectomics has brought into our understanding of the retinal circuitry. We will also discuss unresolved questions about the retinal circuitry that can be addressed by EM connectomics in the future.
Collapse
Affiliation(s)
- Abhilash Sawant
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Aindrila Saha
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Jacob Khoussine
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Raunak Sinha
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
12
|
Kulesh B, Bozadjian R, Parisi RJ, Leong SA, Kautzman AG, Reese BE, Keeley PW. Quantitative trait loci on chromosomes 9 and 19 modulate AII amacrine cell number in the mouse retina. Front Neurosci 2023; 17:1078168. [PMID: 36816119 PMCID: PMC9932814 DOI: 10.3389/fnins.2023.1078168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
Sequence variants modulating gene function or expression affect various heritable traits, including the number of neurons within a population. The present study employed a forward-genetic approach to identify candidate causal genes and their sequence variants controlling the number of one type of retinal neuron, the AII amacrine cell. Data from twenty-six recombinant inbred (RI) strains of mice derived from the parental C57BL/6J (B6/J) and A/J laboratory strains were used to identify genomic loci regulating cell number. Large variation in cell number is present across the RI strains, from a low of ∼57,000 cells to a high of ∼87,000 cells. Quantitative trait locus (QTL) analysis revealed three prospective controlling genomic loci, on Chromosomes (Chrs) 9, 11, and 19, each contributing additive effects that together approach the range of variation observed. Composite interval mapping validated two of these loci, and chromosome substitution strains, in which the A/J genome for Chr 9 or 19 was introgressed on a B6/J genetic background, showed increased numbers of AII amacrine cells as predicted by those two QTL effects. Analysis of the respective genomic loci identified candidate controlling genes defined by their retinal expression, their established biological functions, and by the presence of sequence variants expected to modulate gene function or expression. Two candidate genes, Dtx4 on Chr 19, being a regulator of Notch signaling, and Dixdc1 on Chr 9, a modulator of the WNT-β-catenin signaling pathway, were explored in further detail. Postnatal overexpression of Dtx4 was found to reduce the frequency of amacrine cells, while Dixdc1 knockout retinas contained an excess of AII amacrine cells. Sequence variants in each gene were identified, being the likely sources of variation in gene expression, ultimately contributing to the final number of AII amacrine cells.
Collapse
Affiliation(s)
- Bridget Kulesh
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Rachel Bozadjian
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Ryan J. Parisi
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Stephanie A. Leong
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Amanda G. Kautzman
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Benjamin E. Reese
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Patrick W. Keeley
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
13
|
Whitney IE, Butrus S, Dyer MA, Rieke F, Sanes JR, Shekhar K. Vision-Dependent and -Independent Molecular Maturation of Mouse Retinal Ganglion Cells. Neuroscience 2023; 508:153-173. [PMID: 35870562 PMCID: PMC10809145 DOI: 10.1016/j.neuroscience.2022.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
The development and connectivity of retinal ganglion cells (RGCs), the retina's sole output neurons, are patterned by activity-independent transcriptional programs and activity-dependent remodeling. To inventory the molecular correlates of these influences, we applied high-throughput single-cell RNA sequencing (scRNA-seq) to mouse RGCs at six embryonic and postnatal ages. We identified temporally regulated modules of genes that correlate with, and likely regulate, multiple phases of RGC development, ranging from differentiation and axon guidance to synaptic recognition and refinement. Some of these genes are expressed broadly while others, including key transcription factors and recognition molecules, are selectively expressed by one or a few of the 45 transcriptomically distinct types defined previously in adult mice. Next, we used these results as a foundation to analyze the transcriptomes of RGCs in mice lacking visual experience due to dark rearing from birth or to mutations that ablate either bipolar or photoreceptor cells. 98.5% of visually deprived (VD) RGCs could be unequivocally assigned to a single RGC type based on their transcriptional profiles, demonstrating that visual activity is dispensable for acquisition and maintenance of RGC type identity. However, visual deprivation significantly reduced the transcriptomic distinctions among RGC types, implying that activity is required for complete RGC maturation or maintenance. Consistent with this notion, transcriptomic alternations in VD RGCs significantly overlapped with gene modules found in developing RGCs. Our results provide a resource for mechanistic analyses of RGC differentiation and maturation, and for investigating the role of activity in these processes.
Collapse
Affiliation(s)
- Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Karthik Shekhar
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Helen Wills Neuroscience Institute, California Institute for Quantitative Biosciences, QB3, Center for Computational Biology, University of California, Berkeley, CA 94720, USA; Biological Systems Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
14
|
Zhang C, Hellevik A, Takeuchi S, Wong RO. Hierarchical partner selection shapes rod-cone pathway specificity in the inner retina. iScience 2022; 25:105032. [PMID: 36117987 PMCID: PMC9474917 DOI: 10.1016/j.isci.2022.105032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Neurons form stereotyped microcircuits that underlie specific functions. In the vertebrate retina, the primary rod and cone pathways that convey dim and bright light signals, respectively, exhibit distinct wiring patterns. Rod and cone pathways are thought to be assembled separately during development. However, using correlative fluorescence imaging and serial electron microscopy, we show here that cross-pathway interactions are involved to achieve pathway-specific connectivity within the inner retina. We found that A17 amacrine cells, a rod pathway-specific cellular component, heavily bias their synaptogenesis with rod bipolar cells (RBCs) but increase their connectivity with cone bipolar cells (CBCs) when RBCs are largely ablated. This cross-pathway synaptic plasticity occurs during synaptogenesis and is triggered even on partial loss of RBCs. Thus, A17 cells adopt a hierarchical approach in selecting postsynaptic partners from functionally distinct pathways (RBC>CBC), in which contact and/or synaptogenesis with preferred partners (RBCs) influences connectivity with less-preferred partners (CBCs).
Collapse
Affiliation(s)
- Chi Zhang
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ayana Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Shunsuke Takeuchi
- Department of Biological Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Rachel O. Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
15
|
Sinha R, Grimes WN, Wallin J, Ebbinghaus BN, Luu K, Cherry T, Rieke F, Rudolph U, Wong RO, Hoon M. Transient expression of a GABA receptor subunit during early development is critical for inhibitory synapse maturation and function. Curr Biol 2021; 31:4314-4326.e5. [PMID: 34433078 DOI: 10.1016/j.cub.2021.07.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/29/2021] [Accepted: 07/23/2021] [Indexed: 10/20/2022]
Abstract
Developing neural circuits, including GABAergic circuits, switch receptor types. But the role of early GABA receptor expression for establishment of functional inhibitory circuits remains unclear. Tracking the development of GABAergic synapses across axon terminals of retinal bipolar cells (BCs), we uncovered a crucial role of early GABAA receptor expression for the formation and function of presynaptic inhibitory synapses. Specifically, early α3-subunit-containing GABAA (GABAAα3) receptors are a key developmental organizer. Before eye opening, GABAAα3 gives way to GABAAα1 at individual BC presynaptic inhibitory synapses. The developmental downregulation of GABAAα3 is independent of GABAAα1 expression. Importantly, lack of early GABAAα3 impairs clustering of GABAAα1 and formation of functional GABAA synapses across mature BC terminals. This impacts the sensitivity of visual responses transmitted through the circuit. Lack of early GABAAα3 also perturbs aggregation of LRRTM4, the organizing protein at GABAergic synapses of rod BC terminals, and their arrangement of output ribbon synapses.
Collapse
Affiliation(s)
- Raunak Sinha
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - William N Grimes
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA; National Institute of Neurological Disease and Stroke, NIH, Bethesda, MD, USA
| | - Julie Wallin
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Briana N Ebbinghaus
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelsey Luu
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Timothy Cherry
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington-Seattle and the Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Mrinalini Hoon
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Sawant A, Ebbinghaus BN, Bleckert A, Gamlin C, Yu WQ, Berson D, Rudolph U, Sinha R, Hoon M. Organization and emergence of a mixed GABA-glycine retinal circuit that provides inhibition to mouse ON-sustained alpha retinal ganglion cells. Cell Rep 2021; 34:108858. [PMID: 33730586 PMCID: PMC8030271 DOI: 10.1016/j.celrep.2021.108858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 12/07/2020] [Accepted: 02/19/2021] [Indexed: 12/04/2022] Open
Abstract
In the retina, amacrine interneurons inhibit retinal ganglion cell (RGC) dendrites to shape retinal output. Amacrine cells typically use either GABA or glycine to exert synaptic inhibition. Here, we combined transgenic tools with immunohistochemistry, electrophysiology, and 3D electron microscopy to determine the composition and organization of inhibitory synapses across the dendritic arbor of a well-characterized RGC type in the mouse retina: the ON-sustained alpha RGC. We find mixed GABA-glycine receptor synapses across this RGC type, unveiling the existence of "mixed" inhibitory synapses in the retinal circuit. Presynaptic amacrine boutons with dual release sites are apposed to ON-sustained alpha RGC postsynapses. We further reveal the sequence of postsynaptic assembly for these mixed synapses: GABA receptors precede glycine receptors, and a lack of early GABA receptor expression impedes the recruitment of glycine receptors. Together our findings uncover the organization and developmental profile of an additional motif of inhibition in the mammalian retina.
Collapse
Affiliation(s)
- Abhilash Sawant
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Briana N Ebbinghaus
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam Bleckert
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Clare Gamlin
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - David Berson
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Raunak Sinha
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
17
|
Glycinergic Inhibition Targets Specific Off Cone Bipolar Cells in Primate Retina. eNeuro 2021; 8:ENEURO.0432-20.2020. [PMID: 33188005 PMCID: PMC7920536 DOI: 10.1523/eneuro.0432-20.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/06/2020] [Indexed: 11/21/2022] Open
Abstract
Adapting between scotopic and photopic illumination involves switching the routing of retinal signals between rod and cone-dominated circuits. In the daytime, cone signals pass through parallel On and Off cone bipolar cells (CBCs), that are sensitive to increments and decrements in luminance, respectively. At night, rod signals are routed into these cone-pathways via a key glycinergic interneuron, the AII amacrine cell (AII-AC). AII-ACs also provide On-pathway-driven crossover inhibition to Off-CBCs under photopic conditions. In primates, it is not known whether all Off-bipolar cell types receive functional inputs from AII-ACs. Here, we show that select Off-CBC types receive significantly higher levels of On-pathway-driven glycinergic input than others. The rise and decay kinetics of the glycinergic events are consistent with involvement of the α1 glycine receptor (GlyR) subunit, a result supported by a higher level of GLRA1 transcript in these cells. The Off-bipolar types that receive glycinergic input have sustained physiological properties and include the flat midget bipolar (FMB) cells, which provide excitatory input to the Off-midget ganglion cells (GCs; parvocellular pathway). Our results suggest that only a subset of Off-bipolar cells have the requisite receptors to respond to AII-AC input. Taken together with results in mouse retina, our findings suggest a conserved motif whereby signal output from AII-ACs is preferentially routed into sustained Off-bipolar signaling pathways.
Collapse
|