1
|
Wang J, Zhang L, Cavallini M, Pahlevan A, Sun J, Morshedian A, Fain GL, Sampath AP, Peng YR. Molecular characterization of the sea lamprey retina illuminates the evolutionary origin of retinal cell types. Nat Commun 2024; 15:10761. [PMID: 39737973 DOI: 10.1038/s41467-024-55019-x] [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: 11/23/2023] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
The lamprey, a primitive jawless vertebrate whose ancestors diverged from all other vertebrates over 500 million years ago, offers a unique window into the ancient formation of the retina. Using single-cell RNA-sequencing, we characterize retinal cell types in the lamprey and compare them to those in mouse, chicken, and zebrafish. We find six cell classes and 74 distinct cell types, many shared with other vertebrate species. The conservation of cell types indicates their emergence early in vertebrate evolution, highlighting primordial designs of retinal circuits for the rod pathway, ON-OFF discrimination, and direction selectivity. The diversification of amacrine and some ganglion cell types appears, however, to be distinct in the lamprey. We further infer genetic regulators in specifying retinal cell classes and identify ancestral regulatory elements across species, noting decreased conservation in specifying amacrine cells. Altogether, our characterization of the lamprey retina illuminates the evolutionary origin of visual processing in the retina.
Collapse
Affiliation(s)
- Junqiang Wang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lin Zhang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martina Cavallini
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ali Pahlevan
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Junwei Sun
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ala Morshedian
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Gordon L Fain
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Almoril-Porras A, Calvo AC, Niu L, Beagan J, Díaz García M, Hawk JD, Aljobeh A, Wisdom EM, Ren I, Wang ZW, Colón-Ramos DA. Configuration of electrical synapses filters sensory information to drive behavioral choices. Cell 2024:S0092-8674(24)01378-3. [PMID: 39742807 DOI: 10.1016/j.cell.2024.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 07/26/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025]
Abstract
Synaptic configurations underpin how the nervous system processes sensory information to produce a behavioral response. This is best understood for chemical synapses, and we know far less about how electrical synaptic configurations modulate sensory information processing and context-specific behaviors. We discovered that innexin 1 (INX-1), a gap junction protein that forms electrical synapses, is required to deploy context-specific behavioral strategies underlying thermotaxis behavior in C. elegans. Within this well-defined circuit, INX-1 couples two bilaterally symmetric interneurons to integrate sensory information during migratory behavior across temperature gradients. In inx-1 mutants, uncoupled interneurons display increased excitability and responses to subthreshold sensory stimuli due to increased membrane resistance and reduced membrane capacitance, resulting in abnormal responses that extend run durations and trap the animals in context-irrelevant tracking of isotherms. Thus, a conserved configuration of electrical synapses enables differential processing of sensory information to deploy context-specific behavioral strategies.
Collapse
Affiliation(s)
- Agustin Almoril-Porras
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ana C Calvo
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jonathan Beagan
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Malcom Díaz García
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Josh D Hawk
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ahmad Aljobeh
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Elias M Wisdom
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ivy Ren
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Daniel A Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA; Wu Tsai Institute, Yale University, New Haven, CT 06510, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA; Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan 00901, Puerto Rico.
| |
Collapse
|
3
|
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
|
4
|
Fitzpatrick MJ, Krizan J, Hsiang JC, Shen N, Kerschensteiner D. A pupillary contrast response in mice and humans: Neural mechanisms and visual functions. Neuron 2024; 112:2404-2422.e9. [PMID: 38697114 PMCID: PMC11257825 DOI: 10.1016/j.neuron.2024.04.012] [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: 04/03/2023] [Revised: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
In the pupillary light response (PLR), increases in ambient light constrict the pupil to dampen increases in retinal illuminance. Here, we report that the pupillary reflex arc implements a second input-output transformation; it senses temporal contrast to enhance spatial contrast in the retinal image and increase visual acuity. The pupillary contrast response (PCoR) is driven by rod photoreceptors via type 6 bipolar cells and M1 ganglion cells. Temporal contrast is transformed into sustained pupil constriction by the M1's conversion of excitatory input into spike output. Computational modeling explains how the PCoR shapes retinal images. Pupil constriction improves acuity in gaze stabilization and predation in mice. Humans exhibit a PCoR with similar tuning properties to mice, which interacts with eye movements to optimize the statistics of the visual input for retinal encoding. Thus, we uncover a conserved component of active vision, its cell-type-specific pathway, computational mechanisms, and optical and behavioral significance.
Collapse
Affiliation(s)
- Michael J Fitzpatrick
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna Krizan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
5
|
Yadav SC, Ganzen L, Nawy S, Kramer RH. Retinal bipolar cells borrow excitability from electrically coupled inhibitory interneurons to amplify excitatory synaptic transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601922. [PMID: 39005421 PMCID: PMC11245017 DOI: 10.1101/2024.07.03.601922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Bipolar cells of the retina carry visual information from photoreceptors in the outer retina to retinal ganglion cells (RGCs) in the inner retina. Bipolar cells express L-type voltage-gated Ca2+ channels at the synaptic terminal, but generally lack other types of channels capable of regenerative activity. As a result, the flow of information from outer to inner retina along bipolar cell processes is generally passive in nature, with no opportunity for signal boost or amplification along the way. Here we report the surprising discovery that blocking voltage-gated Na+ channels profoundly reduces the synaptic output of one class of bipolar cell, the type 6 ON bipolar cell (CBC6), despite the fact that the CBC6 itself does not express voltage-gated Na+ channels. Instead, CBC6 borrows voltage-gated Na+ channels from its neighbor, the inhibitory AII amacrine cell, with whom it is connected via an electrical synapse. Thus, an inhibitory neuron aids in amplification of an excitatory signal as it moves through the retina, ensuring that small changes in the membrane potential of bipolar cells are reliably passed onto downstream RGCs.
Collapse
Affiliation(s)
- Shubhash Chandra Yadav
- University of California Berkeley, Department of Molecular and Cell Biology. Berkeley, CA, USA
| | - Logan Ganzen
- University of California Berkeley, Department of Molecular and Cell Biology. Berkeley, CA, USA
| | - Scott Nawy
- University of California Berkeley, Department of Molecular and Cell Biology. Berkeley, CA, USA
| | - Richard H Kramer
- University of California Berkeley, Department of Molecular and Cell Biology. Berkeley, CA, USA
| |
Collapse
|
6
|
Kilpeläinen M, Westö J, Tiihonen J, Laihi A, Takeshita D, Rieke F, Ala-Laurila P. Primate retina trades single-photon detection for high-fidelity contrast encoding. Nat Commun 2024; 15:4501. [PMID: 38802354 PMCID: PMC11130139 DOI: 10.1038/s41467-024-48750-y] [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: 02/01/2023] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
How the spike output of the retina enables human visual perception is not fully understood. Here, we address this at the sensitivity limit of vision by correlating human visual perception with the spike outputs of primate ON and OFF parasol (magnocellular) retinal ganglion cells in tightly matching stimulus conditions. We show that human vision at its ultimate sensitivity limit depends on the spike output of the ON but not the OFF retinal pathway. Consequently, nonlinear signal processing in the retinal ON pathway precludes perceptual detection of single photons in darkness but enables quantal-resolution discrimination of differences in light intensity.
Collapse
Affiliation(s)
- Markku Kilpeläinen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Johan Westö
- Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Jussi Tiihonen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
- Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Anton Laihi
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Daisuke Takeshita
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, US
| | - Petri Ala-Laurila
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland.
- Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland.
| |
Collapse
|
7
|
Hamadmad S, Heisler-Taylor T, Goswami S, Hawthorn E, Chaurasia S, Martini D, Summitt D, Zaatari A, Urbanski EG, Bernstein K, Racine J, Satoskar A, El-Hodiri HM, Fischer AJ, Cebulla CM. Ibudilast Protects Retinal Bipolar Cells from Excitotoxic Retinal Damage and Activates the mTOR Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585556. [PMID: 38562805 PMCID: PMC10983953 DOI: 10.1101/2024.03.18.585556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Ibudilast, an inhibitor of macrophage migration inhibitory factor (MIF) and phosphodiesterase (PDE), has been recently shown to have neuroprotective effects in a variety of neurologic diseases. We utilize a chick excitotoxic retinal damage model to investigate ibudilast's potential to protect retinal neurons. Using single cell RNA-sequencing (scRNA-seq), we find that MIF, putative MIF receptors CD74 and CD44, and several PDEs are upregulated in different retinal cells during damage. Intravitreal ibudilast is well tolerated in the eye and causes no evidence of toxicity. Ibudilast effectively protects neurons in the inner nuclear layer from NMDA-induced cell death, restores retinal layer thickness on spectral domain optical coherence tomography, and preserves retinal neuron function, particularly for the ON bipolar cells, as assessed by electroretinography. PDE inhibition seems essential for ibudilast's neuroprotection, as AV1013, the analogue that lacks PDE inhibitor activity, is ineffective. scRNA-seq analysis reveals upregulation of multiple signaling pathways, including mTOR, in damaged Müller glia (MG) with ibudilast treatment compared to AV1013. Components of mTORC1 and mTORC2 are upregulated in both bipolar cells and MG with ibudilast. The mTOR inhibitor rapamycin blocked accumulation of pS6 but did not reduce TUNEL positive dying cells. Additionally, through ligand-receptor interaction analysis, crosstalk between bipolar cells and MG may be important for neuroprotection. We have identified several paracrine signaling pathways that are known to contribute to cell survival and neuroprotection and might play essential roles in ibudilast function. These findings highlight ibudilast's potential to protect inner retinal neurons during damage and show promise for future clinical translation.
Collapse
|
8
|
Roy S, Yao X, Rathinavelu J, Field GD. GABAergic Inhibition Controls Receptive Field Size, Sensitivity, and Contrast Preference of Direction Selective Retinal Ganglion Cells Near the Threshold of Vision. J Neurosci 2024; 44:e1979232023. [PMID: 38182419 PMCID: PMC10941243 DOI: 10.1523/jneurosci.1979-23.2023] [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: 10/19/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Information about motion is encoded by direction-selective retinal ganglion cells (DSGCs). These cells reliably transmit this information across a broad range of light levels, spanning moonlight to sunlight. Previous work indicates that adaptation to low light levels causes heterogeneous changes to the direction tuning of ON-OFF (oo)DSGCs and suggests that superior-preferring ON-OFF DSGCs (s-DSGCs) are biased toward detecting stimuli rather than precisely signaling direction. Using a large-scale multielectrode array, we measured the absolute sensitivity of ooDSGCs and found that s-DSGCs are 10-fold more sensitive to dim flashes of light than other ooDSGCs. We measured their receptive field (RF) sizes and found that s-DSGCs also have larger receptive fields than other ooDSGCs; however, the size difference does not fully explain the sensitivity difference. Using a conditional knock-out of gap junctions and pharmacological manipulations, we demonstrate that GABA-mediated inhibition contributes to the difference in absolute sensitivity and receptive field size at low light levels, while the connexin36-mediated gap junction coupling plays a minor role. We further show that under scotopic conditions, ooDSGCs exhibit only an ON response, but pharmacologically removing GABA-mediated inhibition unmasks an OFF response. These results reveal that GABAergic inhibition controls and differentially modulates the responses of ooDSGCs under scotopic conditions.
Collapse
Affiliation(s)
- Suva Roy
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| | - Xiaoyang Yao
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Jay Rathinavelu
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Greg D Field
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| |
Collapse
|
9
|
Baden T. Ancestral photoreceptor diversity as the basis of visual behaviour. Nat Ecol Evol 2024; 8:374-386. [PMID: 38253752 DOI: 10.1038/s41559-023-02291-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/10/2023] [Indexed: 01/24/2024]
Abstract
Animal colour vision is based on comparing signals from different photoreceptors. It is generally assumed that processing different spectral types of photoreceptor mainly serves colour vision. Here I propose instead that photoreceptors are parallel feature channels that differentially support visual-motor programmes like motion vision behaviours, prey capture and predator evasion. Colour vision may have emerged as a secondary benefit of these circuits, which originally helped aquatic vertebrates to visually navigate and segment their underwater world. Specifically, I suggest that ancestral vertebrate vision was built around three main systems, including a high-resolution general purpose greyscale system based on ancestral red cones and rods to mediate visual body stabilization and navigation, a high-sensitivity specialized foreground system based on ancestral ultraviolet cones to mediate threat detection and prey capture, and a net-suppressive system based on ancestral green and blue cones for regulating red/rod and ultraviolet circuits. This ancestral strategy probably still underpins vision today, and different vertebrate lineages have since adapted their original photoreceptor circuits to suit their diverse visual ecologies.
Collapse
Affiliation(s)
- Tom Baden
- University of Sussex, Sussex Neuroscience, Sussex Center for Sensory Neuroscience and Computation, Brighton, UK.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Ribelayga CP, O’Brien J. When microscopy and electrophysiology meet connectomics-Steve Massey's contribution to unraveling the structure and function of the rod/cone gap junction. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1305131. [PMID: 38983007 PMCID: PMC11182179 DOI: 10.3389/fopht.2023.1305131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/31/2023] [Indexed: 07/11/2024]
Abstract
Electrical synapses, formed of gap junctions, are ubiquitous components of the central nervous system (CNS) that shape neuronal circuit connectivity and dynamics. In the retina, electrical synapses can create a circuit, control the signal-to-noise ratio in individual neurons, and support the coordinated neuronal firing of ganglion cells, hence, regulating signal processing at the network, single-cell, and dendritic level. We, the authors, and Steve Massey have had a long interest in gap junctions in retinal circuits, in general, and in the network of photoreceptors, in particular. Our combined efforts, based on a wide array of techniques of molecular biology, microscopy, and electrophysiology, have provided fundamental insights into the molecular structure and properties of the rod/cone gap junction. Yet, a full understanding of how rod/cone coupling controls circuit dynamics necessitates knowing its operating range. It is well established that rod/cone coupling can be greatly reduced or eliminated by bright-light adaptation or pharmacological treatment; however, the upper end of its dynamic range has long remained elusive. This held true until Steve Massey's recent interest for connectomics led to the development of a new strategy to assess this issue. The effort proved effective in establishing, with precision, the connectivity rules between rods and cones and estimating the theoretical upper limit of rod/cone electrical coupling. Comparing electrophysiological measurements and morphological data indicates that under pharmacological manipulation, rod/cone coupling can reach the theoretical maximum of its operating range, implying that, under these conditions, all the gap junction channels present at the junctions are open. As such, channel open probability is likely the main determinant of rod/cone coupling that can change momentarily in a time-of-day- and light-dependent manner. In this article we briefly review our current knowledge of the molecular structure of the rod/cone gap junction and of the mechanisms behind its modulation, and we highlight the recent work led by Steve Massey. Steve's contribution has been critical toward asserting the modulation depth of rod/cone coupling as well as elevating the rod/cone gap junction as one of the most suitable models to examine the role of electrical synapses and their plasticity in neural processing.
Collapse
Affiliation(s)
- Christophe P. Ribelayga
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX, United States
| | | |
Collapse
|
13
|
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
|
14
|
Almoril-Porras A, Calvo AC, Niu L, Beagan J, Hawk JD, Aljobeh A, Wisdom EM, Ren I, Díaz-García M, Wang ZW, Colón-Ramos DA. Specific configurations of electrical synapses filter sensory information to drive choices in behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551556. [PMID: 37577611 PMCID: PMC10418224 DOI: 10.1101/2023.08.01.551556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Synaptic configurations in precisely wired circuits underpin how sensory information is processed by the nervous system, and the emerging animal behavior. This is best understood for chemical synapses, but far less is known about how electrical synaptic configurations modulate, in vivo and in specific neurons, sensory information processing and context-specific behaviors. We discovered that INX-1, a gap junction protein that forms electrical synapses, is required to deploy context-specific behavioral strategies during C. elegans thermotaxis behavior. INX-1 couples two bilaterally symmetric interneurons, and this configuration is required for the integration of sensory information during migration of animals across temperature gradients. In inx-1 mutants, uncoupled interneurons display increased excitability and responses to subthreshold temperature stimuli, resulting in abnormally longer run durations and context-irrelevant tracking of isotherms. Our study uncovers a conserved configuration of electrical synapses that, by increasing neuronal capacitance, enables differential processing of sensory information and the deployment of context-specific behavioral strategies.
Collapse
Affiliation(s)
- Agustin Almoril-Porras
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Ana C. Calvo
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut Health Center; Farmington, CT 06030, USA
| | - Jonathan Beagan
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Josh D. Hawk
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Ahmad Aljobeh
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Elias M. Wisdom
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Ivy Ren
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Malcom Díaz-García
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut Health Center; Farmington, CT 06030, USA
| | - Daniel A. Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
- Wu Tsai Institute, Yale University; New Haven, CT 06510, USA
- Marine Biological Laboratory; Woods Hole, MA, USA
- Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico; San Juan 00901, Puerto Rico
| |
Collapse
|
15
|
Bhoi JD, Goel M, Ribelayga CP, Mangel SC. Circadian clock organization in the retina: From clock components to rod and cone pathways and visual function. Prog Retin Eye Res 2023; 94:101119. [PMID: 36503722 PMCID: PMC10164718 DOI: 10.1016/j.preteyeres.2022.101119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022]
Abstract
Circadian (24-h) clocks are cell-autonomous biological oscillators that orchestrate many aspects of our physiology on a daily basis. Numerous circadian rhythms in mammalian and non-mammalian retinas have been observed and the presence of an endogenous circadian clock has been demonstrated. However, how the clock and associated rhythms assemble into pathways that support and control retina function remains largely unknown. Our goal here is to review the current status of our knowledge and evaluate recent advances. We describe many previously-observed retinal rhythms, including circadian rhythms of morphology, biochemistry, physiology, and gene expression. We evaluate evidence concerning the location and molecular machinery of the retinal circadian clock, as well as consider findings that suggest the presence of multiple clocks. Our primary focus though is to describe in depth circadian rhythms in the light responses of retinal neurons with an emphasis on clock control of rod and cone pathways. We examine evidence that specific biochemical mechanisms produce these daily light response changes. We also discuss evidence for the presence of multiple circadian retinal pathways involving rhythms in neurotransmitter activity, transmitter receptors, metabolism, and pH. We focus on distinct actions of two dopamine receptor systems in the outer retina, a dopamine D4 receptor system that mediates circadian control of rod/cone gap junction coupling and a dopamine D1 receptor system that mediates non-circadian, light/dark adaptive regulation of gap junction coupling between horizontal cells. Finally, we evaluate the role of circadian rhythmicity in retinal degeneration and suggest future directions for the field of retinal circadian biology.
Collapse
Affiliation(s)
- Jacob D Bhoi
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA
| | - Manvi Goel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA.
| | - Stuart C Mangel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
16
|
Rodgers J, Hughes S, Lindner M, Allen AE, Ebrahimi AS, Storchi R, Peirson SN, Lucas RJ, Hankins MW. Functional integrity of visual coding following advanced photoreceptor degeneration. Curr Biol 2023; 33:474-486.e5. [PMID: 36630957 DOI: 10.1016/j.cub.2022.12.026] [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: 08/21/2022] [Revised: 11/01/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023]
Abstract
Photoreceptor degeneration sufficient to produce severe visual loss often spares the inner retina. This raises hope for vision restoration treatments using optogenetics or electrical stimulation, which generate a replacement light input signal in surviving neurons. The success of these approaches is dependent on the capacity of surviving circuits of the visual system to generate and propagate an appropriate visual code in the face of neuroanatomical remodeling. To determine whether retinally degenerate animals possess this capacity, we generated a transgenic mouse model expressing the optogenetic actuator ReaChR in ON bipolar cells (second-order neurons in the visual projection). After crossing this with the rd1 model of photoreceptor degeneration, we compared ReaChR-derived responses with photoreceptor-driven responses in wild-type (WT) mice at the level of retinal ganglion cells and the visual thalamus. The ReaChR-driven responses in rd1 animals showed low photosensitivity, but in other respects generated a visual code that was very similar to the WT. ReaChR rd1 responses had high trial-to-trial reproducibility and showed sensitivity normalization to code contrast across background intensities. At the single unit level, ReaChR-derived responses exhibited broadly similar variations in response polarity, contrast sensitivity, and temporal frequency tuning as the WT. Units from the WT and ReaChR rd1 mice clustered together when subjected to unsupervised community detection based on stimulus-response properties. Our data reveal an impressive ability for surviving circuitry to recreate a rich visual code following advanced retinal degeneration and are promising for regenerative medicine in the central nervous system.
Collapse
Affiliation(s)
- Jessica Rodgers
- Faculty of Biology, Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Steven Hughes
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Moritz Lindner
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps University, Deutschhausstr. 1-2, Marburg 35037, Germany
| | - Annette E Allen
- Faculty of Biology, Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Aghileh S Ebrahimi
- Faculty of Biology, Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Riccardo Storchi
- Faculty of Biology, Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Stuart N Peirson
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Robert J Lucas
- Faculty of Biology, Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK.
| | - Mark W Hankins
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
17
|
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
|
18
|
Pasmanter N, Petersen-Jones SM. Characterization of scotopic and mesopic rod signaling pathways in dogs using the On-Off electroretinogram. BMC Vet Res 2022; 18:422. [PMID: 36463174 PMCID: PMC9719241 DOI: 10.1186/s12917-022-03505-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND The On-Off, or long flash, full field electroretinogram (ERG) separates retinal responses to flash onset and offset. Depending on degree of dark-adaptation and stimulus strength the On and Off ERG can be shaped by rod and cone photoreceptors and postreceptoral cells, including ON and OFF bipolar cells. Interspecies differences have been shown, with predominantly positive Off-response in humans and other primates and a negative Off-response in rodents and dogs. However, the rod signaling pathways that contribute to these differential responses have not been characterized. In this study, we designed a long flash protocol in the dog that varied in background luminance and stimulus strength allowing for some rod components to be present to better characterize how rod pathways vary from scotopic to mesopic conditions. RESULTS With low background light the rod a-wave remains while the b-wave is significantly reduced resulting in a predominantly negative waveform in mesopic conditions. Through modeling and subtraction of the rod-driven response, we show that rod bipolar cells saturate with dimmer backgrounds than rod photoreceptors, resulting in rod hyperpolarization contributing to a large underlying negativity with mesopic backgrounds. CONCLUSIONS Reduction in rod bipolar cell responses in mesopic conditions prior to suppression of rod photoreceptor responses may reflect the changes in signaling pathway of rod-driven responses needed to extend the range of lighting conditions over which the retina functions.
Collapse
Affiliation(s)
- Nate Pasmanter
- grid.17088.360000 0001 2150 1785Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D208 East Lansing, MI USA
| | - Simon M. Petersen-Jones
- grid.17088.360000 0001 2150 1785Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D208 East Lansing, MI USA
| |
Collapse
|
19
|
Cao Y, Fajardo D, Guerrero-Given D, Samuel MA, Ohtsuka T, Boye SE, Kamasawa N, Martemyanov KA. Post-developmental plasticity of the primary rod pathway allows restoration of visually guided behaviors. Curr Biol 2022; 32:4783-4796.e3. [PMID: 36179691 PMCID: PMC9691582 DOI: 10.1016/j.cub.2022.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 01/24/2023]
Abstract
The formation of neural circuits occurs in a programmed fashion, but proper activity in the circuit is essential for refining the organization necessary for driving complex behavioral tasks. In the retina, sensory deprivation during the critical period of development is well known to perturb the organization of the visual circuit making the animals unable to use vision for behavior. However, the extent of plasticity, molecular factors involved, and malleability of individual channels in the circuit to manipulations outside of the critical period are not well understood. In this study, we selectively disconnected and reconnected rod photoreceptors in mature animals after completion of the retina circuit development. We found that introducing synaptic rod photoreceptor input post-developmentally allowed their integration into the circuit both anatomically and functionally. Remarkably, adult mice with newly integrated rod photoreceptors gained high-sensitivity vision, even when it was absent from birth. These observations reveal plasticity of the retina circuit organization after closure of the critical period and encourage the development of vision restoration strategies for congenital blinding disorders.
Collapse
Affiliation(s)
- Yan Cao
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Diego Fajardo
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Debbie Guerrero-Given
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Naomi Kamasawa
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA.
| |
Collapse
|
20
|
Percival KA, Gayet J, Khanjian R, Taylor WR, Puthussery T. Calcium-permeable AMPA receptors on AII amacrine cells mediate sustained signaling in the On-pathway of the primate retina. Cell Rep 2022; 41:111484. [PMID: 36223749 PMCID: PMC10518213 DOI: 10.1016/j.celrep.2022.111484] [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: 12/24/2021] [Revised: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022] Open
Abstract
Midget and parasol ganglion cells (GCs) represent the major output channels from the primate eye to the brain. On-type midget and parasol GCs exhibit a higher background spike rate and thus can respond more linearly to contrast changes than their Off-type counterparts. Here, we show that a calcium-permeable AMPA receptor (CP-AMPAR) antagonist blocks background spiking and sustained light-evoked firing in On-type GCs while preserving transient light responses. These effects are selective for On-GCs and are occluded by a gap-junction blocker suggesting involvement of AII amacrine cells (AII-ACs). Direct recordings from AII-ACs, cobalt uptake experiments, and analyses of transcriptomic data confirm that CP-AMPARs are expressed by primate AII-ACs. Overall, our data demonstrate that under some background light levels, CP-AMPARs at the rod bipolar to AII-AC synapse drive sustained signaling in On-type GCs and thus contribute to the more linear contrast signaling of the primate On- versus Off-pathway.
Collapse
Affiliation(s)
- Kumiko A Percival
- Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacqueline Gayet
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, Berkeley, CA 94720-2020, USA
| | - Roupen Khanjian
- Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - W Rowland Taylor
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, Berkeley, CA 94720-2020, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-2020, USA
| | - Teresa Puthussery
- Herbert Wertheim School of Optometry & Vision Science, University of California, Berkeley, Berkeley, CA 94720-2020, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-2020, USA.
| |
Collapse
|
21
|
Lindner M, Gilhooley MJ, Hughes S, Hankins MW. Optogenetics for visual restoration: From proof of principle to translational challenges. Prog Retin Eye Res 2022; 91:101089. [PMID: 35691861 DOI: 10.1016/j.preteyeres.2022.101089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/04/2023]
Abstract
Degenerative retinal disorders are a diverse family of diseases commonly leading to irreversible photoreceptor death, while leaving the inner retina relatively intact. Over recent years, innovative gene replacement therapies aiming to halt the progression of certain inherited retinal disorders have made their way into clinics. By rendering surviving retinal neurons light sensitive optogenetic gene therapy now offers a feasible treatment option that can restore lost vision, even in late disease stages and widely independent of the underlying cause of degeneration. Since proof-of-concept almost fifteen years ago, this field has rapidly evolved and a detailed first report on a treated patient has recently been published. In this article, we provide a review of optogenetic approaches for vision restoration. We discuss the currently available optogenetic tools and their relative advantages and disadvantages. Possible cellular targets will be discussed and we will address the question how retinal remodelling may affect the choice of the target and to what extent it may limit the outcomes of optogenetic vision restoration. Finally, we will analyse the evidence for and against optogenetic tool mediated toxicity and will discuss the challenges associated with clinical translation of this promising therapeutic concept.
Collapse
Affiliation(s)
- Moritz Lindner
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom; Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps University, 35037, Marburg, Germany
| | - Michael J Gilhooley
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom; The Institute of Ophthalmology, University College London, EC1V 9EL, United Kingdom; Moorfields Eye Hospital, London, EC1V 2PD, United Kingdom
| | - Steven Hughes
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Mark W Hankins
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom.
| |
Collapse
|
22
|
Westö J, Martyniuk N, Koskela S, Turunen T, Pentikäinen S, Ala-Laurila P. Retinal OFF ganglion cells allow detection of quantal shadows at starlight. Curr Biol 2022; 32:2848-2857.e6. [PMID: 35609606 DOI: 10.1016/j.cub.2022.04.092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023]
Abstract
Perception of light in darkness requires no more than a handful of photons, and this remarkable behavioral performance can be directly linked to a particular retinal circuit-the retinal ON pathway. However, the neural limits of shadow detection in very dim light have remained unresolved. Here, we unravel the neural mechanisms that determine the sensitivity of mice (CBA/CaJ) to light decrements at the lowest light levels by measuring signals from the most sensitive ON and OFF retinal ganglion cell types and by correlating their signals with visually guided behavior. We show that mice can detect shadows when only a few photon absorptions are missing among thousands of rods. Behavioral detection of such "quantal" shadows relies on the retinal OFF pathway and is limited by noise and loss of single-photon signals in retinal processing. Thus, in the dim-light regime, light increments and decrements are encoded separately via the ON and OFF retinal pathways, respectively.
Collapse
Affiliation(s)
- Johan Westö
- Department of Neuroscience and Biomedical Engineering, Aalto University, 02150 Espoo, Finland
| | - Nataliia Martyniuk
- Department of Neuroscience and Biomedical Engineering, Aalto University, 02150 Espoo, Finland
| | - Sanna Koskela
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00790 Helsinki, Finland
| | - Tuomas Turunen
- Department of Neuroscience and Biomedical Engineering, Aalto University, 02150 Espoo, Finland
| | - Santtu Pentikäinen
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00790 Helsinki, Finland
| | - Petri Ala-Laurila
- Department of Neuroscience and Biomedical Engineering, Aalto University, 02150 Espoo, Finland; Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00790 Helsinki, Finland.
| |
Collapse
|
23
|
Dmitriev AV, Dmitriev AA, Linsenmeier RA. Extracellular K+ reflects light-evoked changes in retinal energy metabolism. Exp Eye Res 2022; 221:109133. [PMID: 35636490 PMCID: PMC10392107 DOI: 10.1016/j.exer.2022.109133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Retinal neurons spend most of their energy to support the transmembrane movement of ions. Light-induced electrical activity is associated with a redistribution of ions, which affects the energy demand and results in a change in metabolism. Light-induced metabolic changes are expected to be different in distal and proximal retina due to differences in the light responses of different retinal cells. Extracellular K+ concentration ([K+]o) is a reliable indicator of local electrophysiological activity, and the purpose of this work was to compare [K+]o changes evoked by steady and flickering light in distal and proximal retina. Data were obtained from isolated mouse (C57Bl/6J) retinae. Double-barreled K+-selective microelectrodes were used to simultaneously record [K+]o and local ERGs. In the distal retina, photoreceptor hyperpolarization led to suppression of ion transfer, a decrease in [K+]o by 0.3-0.5 mM, reduced energy demand, and, as previously shown in vivo, decreased metabolism. Flickering light had the same effect on [K+]o in the distal retina as steady light of equivalent illumination. The conductance and voltage changes in postreceptor neurons are cell-specific, but the overall effect of steady light in the proximal retina is excitation, which is reflected in a [K+]o increase there (by a maximum of 0.2 mM). In steady light the [K+]o increase lasts only 1-2 s, but a sustained [K+]o increase is evoked by flickering light. A squarewave low frequency (1 Hz) flicker of photopic intensity produced the largest increases in [K+]o. Judging by measurements of [K+]o, steady illumination decreases energy metabolism in the distal retina, but not in the proximal retina (except for the first few seconds). Flickering light evokes the same decrease in the distal retina, but also evokes a sustained [K+]o increase in the proximal retina, suggesting an increase of metabolic demand there, especially at 1 Hz, when neurons of both on- and off-pathways appear to contribute maximally. This proximal retinal metabolic response to flicker correlates to the increase in blood flow during flicker that constitutes neurovascular coupling.
Collapse
|
24
|
Ishibashi M, Keung J, Morgans CW, Aicher SA, Carroll JR, Singer JH, Jia L, Li W, Fahrenfort I, Ribelayga CP, Massey SC. Analysis of rod/cone gap junctions from the reconstruction of mouse photoreceptor terminals. eLife 2022; 11:73039. [PMID: 35471186 PMCID: PMC9170248 DOI: 10.7554/elife.73039] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/25/2022] [Indexed: 12/14/2022] Open
Abstract
Electrical coupling, mediated by gap junctions, contributes to signal averaging, synchronization, and noise reduction in neuronal circuits. In addition, gap junctions may also provide alternative neuronal pathways. However, because they are small and especially difficult to image, gap junctions are often ignored in large-scale 3D reconstructions. Here, we reconstruct gap junctions between photoreceptors in the mouse retina using serial blockface-scanning electron microscopy, focused ion beam-scanning electron microscopy, and confocal microscopy for the gap junction protein Cx36. An exuberant spray of fine telodendria extends from each cone pedicle (including blue cones) to contact 40-50 nearby rod spherules at sites of Cx36 labeling, with approximately 50 Cx36 clusters per cone pedicle and 2-3 per rod spherule. We were unable to detect rod/rod or cone/cone coupling. Thus, rod/cone coupling accounts for nearly all gap junctions between photoreceptors. We estimate a mean of 86 Cx36 channels per rod/cone pair, which may provide a maximum conductance of ~1200 pS, if all gap junction channels were open. This is comparable to the maximum conductance previously measured between rod/cone pairs in the presence of a dopamine antagonist to activate Cx36, suggesting that the open probability of gap junction channels can approach 100% under certain conditions.
Collapse
Affiliation(s)
- Munenori Ishibashi
- Richard Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas at Houston, Houston, United States
| | - Joyce Keung
- Richard Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas at Houston, Houston, United States
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, United States
| | - Sue A Aicher
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, United States
| | - James R Carroll
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, United States
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, College Park, United States
| | - Li Jia
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, United States
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, United States
| | - Iris Fahrenfort
- Richard Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas at Houston, Houston, United States
| | - Christophe P Ribelayga
- Richard Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas at Houston, Houston, United States
| | - Stephen C Massey
- Richard Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas at Houston, Houston, United States
| |
Collapse
|
25
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Jin N, Tian LM, Fahrenfort I, Zhang Z, Postma F, Paul DL, Massey SC, Ribelayga CP. Genetic elimination of rod/cone coupling reveals the contribution of the secondary rod pathway to the retinal output. SCIENCE ADVANCES 2022; 8:eabm4491. [PMID: 35363529 PMCID: PMC10938630 DOI: 10.1126/sciadv.abm4491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/10/2022] [Indexed: 06/14/2023]
Abstract
In the retina, signals originating from rod and cone photoreceptors can reach retinal ganglion cells (RGCs)-the output neurons-through different pathways. However, little is known about the exact sensitivities and operating ranges of these pathways. Previously, we created rod- or cone-specific Cx36 knockout (KO) mouse lines. Both lines are deficient in rod/cone electrical coupling and therefore provide a way to selectively remove the secondary rod pathway. We measured the threshold of the primary rod pathway in RGCs of wild-type mice. Under pharmacological blockade of the primary rod pathway, the threshold was elevated. This secondary component was removed in the Cx36 KOs to unmask the threshold of the third rod pathway, still below cone threshold. In turn, the cone threshold was estimated by several independent methods. Our work defines the functionality of the secondary rod pathway and describes an additive contribution of the different pathways to the retinal output.
Collapse
Affiliation(s)
- Nange Jin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - Lian-Ming Tian
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - Iris Fahrenfort
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - Friso Postma
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - David L. Paul
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - Stephen C. Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Elizabeth Morford Distinguished Chair in Ophthalmology and Research Director, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - Christophe P. Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Bernice Weingarten Chair in Ophthalmology, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| |
Collapse
|
27
|
Pasmanter N, Occelli LM, Komáromy AM, Petersen-Jones SM. Use of extended protocols with nonstandard stimuli to characterize rod and cone contributions to the canine electroretinogram. Doc Ophthalmol 2022; 144:81-97. [PMID: 35247111 PMCID: PMC10426558 DOI: 10.1007/s10633-022-09866-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 02/08/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE In this study, we assessed several extended electroretinographic protocols using nonstandard stimuli. Our aim was to separate and quantify the contributions of different populations of retinal cells to the overall response, both to assess normal function and characterize dogs with inherited retinal disease. METHODS We investigated three different protocols for measuring the full-field flash electroretinogram-(1) chromatic dark-adapted red and blue flashes, (2) increasing luminance blue-background, (3) flicker with fixed frequency and increasing luminance, and flicker with increasing frequency at a fixed luminance-to assess rod and cone contributions to electroretinograms recorded in phenotypically normal control dogs and dogs lacking rod function. RESULTS Temporal separation of the rod- and cone-driven responses is possible in the fully dark-adapted eye using dim red flashes. A- and b-wave amplitudes decrease at different rates with increasing background luminance in control dogs. Flicker responses elicited with extended flicker protocols are well fit with mathematical models in control dogs. Dogs lacking rod function demonstrated larger amplitude dark-adapted compared to light-adapted flicker responses. CONCLUSIONS Using extended protocols of the full-field electroretinogram provides additional characterization of the health and function of different populations of cells in the normal retina and enables quantifiable comparison between phenotypically normal dogs and those with retinal disease.
Collapse
Affiliation(s)
- Nate Pasmanter
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D-208, East Lansing, MI, 48824, USA
| | - Laurence M Occelli
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D-208, East Lansing, MI, 48824, USA
| | - András M Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D-208, East Lansing, MI, 48824, USA
| | - Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 736 Wilson Road, D-208, East Lansing, MI, 48824, USA.
| |
Collapse
|
28
|
Adámek P, Langová V, Horáček J. Early-stage visual perception impairment in schizophrenia, bottom-up and back again. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:27. [PMID: 35314712 PMCID: PMC8938488 DOI: 10.1038/s41537-022-00237-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/17/2022] [Indexed: 01/01/2023]
Abstract
Visual perception is one of the basic tools for exploring the world. However, in schizophrenia, this modality is disrupted. So far, there has been no clear answer as to whether the disruption occurs primarily within the brain or in the precortical areas of visual perception (the retina, visual pathways, and lateral geniculate nucleus [LGN]). A web-based comprehensive search of peer-reviewed journals was conducted based on various keyword combinations including schizophrenia, saliency, visual cognition, visual pathways, retina, and LGN. Articles were chosen with respect to topic relevance. Searched databases included Google Scholar, PubMed, and Web of Science. This review describes the precortical circuit and the key changes in biochemistry and pathophysiology that affect the creation and characteristics of the retinal signal as well as its subsequent modulation and processing in other parts of this circuit. Changes in the characteristics of the signal and the misinterpretation of visual stimuli associated with them may, as a result, contribute to the development of schizophrenic disease.
Collapse
Affiliation(s)
- Petr Adámek
- Third Faculty of Medicine, Charles University, Prague, Czech Republic. .,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic.
| | - Veronika Langová
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic
| | - Jiří Horáček
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic
| |
Collapse
|
29
|
Hunt H, Dittmer KE, Garrick DJ, Fairley RA, Heap SJ, Jolly RD. An inherited night blindness in Wiltshire sheep. Vet Pathol 2022; 59:310-318. [PMID: 34974772 DOI: 10.1177/03009858211067461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Twelve cases of adult-onset blindness were identified in a flock of 130 polled Wiltshire sheep in New Zealand over a 3-year period. Affected sheep developed night blindness between 2 and 3 years of age, which progressed to complete blindness by 4 to 5 years of age. Fundic examination findings included progressive tapetal hyperreflectivity and attenuation of retinal blood vessels. Histologically, the retinas had a selective loss of rod photoreceptors with initial preservation of cone photoreceptors. Retinal degeneration was not accompanied by any other ocular or central nervous system abnormalities, and pedigree analysis suggested an inherited basis for the disease. Mating an affected Wiltshire ram to 2 affected Wiltshire ewes resulted in 6 progeny that all developed retinal degeneration by 2 years of age, while mating of the same affected ram to 6 unaffected ewes resulted in 8 unaffected progeny, consistent with autosomal recessive inheritance. Homozygosity mapping of 5 affected Wiltshire sheep and 1 unaffected Wiltshire sheep using an OvineSNP50 Genotyping BeadChip revealed an identical-by-descent region on chromosome 5, but none of the genes within this region were considered plausible candidate genes. Whole-genome sequencing of 2 affected sheep did not reveal any significant mutations in any of the genes associated with retinitis pigmentosa in humans or progressive retinal atrophy in dogs. Inherited progressive retinal degeneration affecting rod photoreceptors has not been previously reported in sheep, but this disease has several similarities to inherited retinal dystrophies in other species.
Collapse
Affiliation(s)
- Hayley Hunt
- Massey University, Palmerston North, New Zealand
| | | | | | | | - Stephen J Heap
- McMaster and Heap Veterinary Practice, Christchurch, New Zealand
| | | |
Collapse
|
30
|
Pang JJ, Gao F, Wu SM. Dual-Cell Patch-Clamp Recording Revealed a Mechanism for a Ribbon Synapse to Process Both Digital and Analog Inputs and Outputs. Front Cell Neurosci 2021; 15:722533. [PMID: 34720878 PMCID: PMC8552968 DOI: 10.3389/fncel.2021.722533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/13/2021] [Indexed: 12/02/2022] Open
Abstract
A chemical synapse is either an action potential (AP) synapse or a graded potential (GP) synapse but not both. This study investigated how signals passed the glutamatergic synapse between the rod photoreceptor and its postsynaptic hyperpolarizing bipolar cells (HBCs) and light responses of retinal neurons with dual-cell and single-cell patch-clamp recording techniques. The results showed that scotopic lights evoked GPs in rods, whose depolarizing Phase 3 associated with the light offset also evoked APs of a duration of 241.8 ms and a slope of 4.5 mV/ms. The depolarization speed of Phase 3 (Speed) was 0.0001–0.0111 mV/ms and 0.103–0.469 mV/ms for rods and cones, respectively. On pairs of recorded rods and HBCs, only the depolarizing limbs of square waves applied to rods evoked clear currents in HBCs which reversed at −6.1 mV, indicating cation currents. We further used stimuli that simulated the rod light response to stimulate rods and recorded the rod-evoked excitatory current (rdEPSC) in HBCs. The normalized amplitude (R/Rmax), delay, and rising slope of rdEPSCs were differentially exponentially correlated with the Speed (all p < 0.001). For the Speed < 0.1 mV/ms, R/Rmax grew while the delay and duration reduced slowly; for the Speed between 0.1 and 0.4 mV/ms, R/Rmax grew fast while the delay and duration dramatically decreased; for the Speed > 0.4 mV/ms, R/Rmax reached the plateau, while the delay and duration approached the minimum, resembling digital signals. The rdEPSC peak was left-shifted and much faster than currents in rods. The scotopic-light-offset-associated major and minor cation currents in retinal ganglion cells (RGCs), the gigantic excitatory transient currents (GTECs) in HBCs, and APs and Phase 3 in rods showed comparable light-intensity-related locations. The data demonstrate that the rod-HBC synapse is a perfect synapse that can differentially decode and code analog and digital signals to process enormously varied rod and coupled-cone inputs.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Fan Gao
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Samuel M Wu
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
31
|
Young BK, Ramakrishnan C, Ganjawala T, Wang P, Deisseroth K, Tian N. An uncommon neuronal class conveys visual signals from rods and cones to retinal ganglion cells. Proc Natl Acad Sci U S A 2021; 118:e2104884118. [PMID: 34702737 PMCID: PMC8612366 DOI: 10.1073/pnas.2104884118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/01/2023] Open
Abstract
Neurons in the central nervous system (CNS) are distinguished by the neurotransmitter types they release, their synaptic connections, morphology, and genetic profiles. To fully understand how the CNS works, it is critical to identify all neuronal classes and reveal their synaptic connections. The retina has been extensively used to study neuronal development and circuit formation. Here, we describe a previously unidentified interneuron in mammalian retina. This interneuron shares some morphological, physiological, and molecular features with retinal bipolar cells, such as receiving input from photoreceptors and relaying visual signals to retinal ganglion cells. It also shares some features with amacrine cells (ACs), particularly Aii-ACs, such as their neurite morphology in the inner plexiform layer, the expression of some AC-specific markers, and possibly the release of the inhibitory neurotransmitter glycine. Thus, we unveil an uncommon interneuron, which may play an atypical role in vision.
Collapse
Affiliation(s)
- Brent K Young
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
| | | | - Tushar Ganjawala
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202
| | - Ping Wang
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Ning Tian
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132;
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, UT 84148
| |
Collapse
|
32
|
Fusz K, Kovács-Öller T, Kóbor P, Szabó-Meleg E, Völgyi B, Buzás P, Telkes I. Regional Variation of Gap Junctional Connections in the Mammalian Inner Retina. Cells 2021; 10:2396. [PMID: 34572046 PMCID: PMC8466939 DOI: 10.3390/cells10092396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/24/2022] Open
Abstract
The retinas of many species show regional specialisations that are evident in the differences in the processing of visual input from different parts of the visual field. Regional specialisation is thought to reflect an adaptation to the natural visual environment, optical constraints, and lifestyle of the species. Yet, little is known about regional differences in synaptic circuitry. Here, we were interested in the topographical distribution of connexin-36 (Cx36), the major constituent of electrical synapses in the retina. We compared the retinas of mice, rats, and cats to include species with different patterns of regional specialisations in the analysis. First, we used the density of Prox1-immunoreactive amacrine cells as a marker of any regional specialisation, with higher cell density signifying more central regions. Double-labelling experiments showed that Prox1 is expressed in AII amacrine cells in all three species. Interestingly, large Cx36 plaques were attached to about 8-10% of Prox1-positive amacrine cell somata, suggesting the strong electrical coupling of pairs or small clusters of cell bodies. When analysing the regional changes in the volumetric density of Cx36-immunoreactive plaques, we found a tight correlation with the density of Prox1-expressing amacrine cells in the ON, but not in the OFF sublamina in all three species. The results suggest that the relative contribution of electrical synapses to the ON- and OFF-pathways of the retina changes with retinal location, which may contribute to functional ON/OFF asymmetries across the visual field.
Collapse
Affiliation(s)
- Katalin Fusz
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Institute of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
33
|
Temporal Contrast Sensitivity Increases despite Photoreceptor Degeneration in a Mouse Model of Retinitis Pigmentosa. eNeuro 2021; 8:ENEURO.0020-21.2021. [PMID: 33509952 PMCID: PMC8059883 DOI: 10.1523/eneuro.0020-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 11/21/2022] Open
Abstract
The detection of temporal variations in amplitude of light intensity, or temporal contrast sensitivity (TCS), depends on the kinetics of rod photoresponse recovery. Uncharacteristically fast rod recovery kinetics are facets of both human patients and transgenic animal models with a P23H rhodopsin mutation, a prevalent cause of retinitis pigmentosa (RP). Here, we show that mice with this mutation (RhoP23H/+) exhibit an age-dependent and illumination-dependent enhancement in TCS compared with controls. At retinal illumination levels producing ≥1000 R*/rod/s or more, postnatal day 30 (P30) RhoP23H/+ mice exhibit a 1.2-fold to 2-fold increase in retinal and optomotor TCS relative to controls in response to flicker frequencies of 3, 6, and 12 Hz despite significant photoreceptor degeneration and loss of flash electroretinogram (ERG) b-wave amplitude. Surprisingly, the TCS of RhoP23H/+ mice further increases as degeneration advances. Enhanced TCS is also observed in a second model (rhodopsin heterozygous mice, Rho+/-) with fast rod recovery kinetics and no apparent retinal degeneration. In both mouse models, enhanced TCS is explained quantitatively by a comprehensive model that includes photoresponse recovery kinetics, density and collecting area of degenerating rods. Measurement of TCS may be a non-invasive early diagnostic tool indicative of rod dysfunction in some forms of retinal degenerative disease.
Collapse
|
34
|
Kosta P, Iseri E, Loizos K, Paknahad J, Pfeiffer RL, Sigulinsky CL, Anderson JR, Jones BW, Lazzi G. Model-based comparison of current flow in rod bipolar cells of healthy and early-stage degenerated retina. Exp Eye Res 2021; 207:108554. [PMID: 33794197 DOI: 10.1016/j.exer.2021.108554] [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: 10/01/2020] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 12/27/2022]
Abstract
Retinal degenerative diseases, such as retinitis pigmentosa, are generally thought to initiate with the loss of photoreceptors, though recent work suggests that plasticity and remodeling occurs prior to photoreceptor cell loss. This degeneration subsequently leads to death of other retinal neurons, creating functional alterations and extensive remodeling of retinal networks. Retinal prosthetic devices stimulate the surviving retinal cells by applying external current using implanted electrodes. Although these devices restore partial vision, the quality of restored vision is limited. Further knowledge about the precise changes in degenerated retina as the disease progresses is essential to understand how current flows in retinas undergoing degenerative disease and to improve the performance of retinal prostheses. We developed computational models that describe current flow from rod photoreceptors to rod bipolar cells (RodBCs) in the healthy and early-stage degenerated retina. Morphologically accurate models of retinal cells with their synapses are constructed based on retinal connectome datasets, created using serial section transmission electron microscopy (TEM) images of 70 nm-thick slices of either healthy (RC1) or early-stage degenerated (RPC1) rabbit retina. The passive membrane and active ion currents of each cell are implemented using conductance-based models in the Neuron simulation environment. In response to photocurrent input at rod photoreceptors, the simulated membrane potential at RodBCs in early degenerate tissue is approximately 10-20 mV lower than that of RodBCs of that observed in wild type retina. Results presented here suggest that although RodBCs in RPC1 show early, altered morphology compared to RC1, the lower membrane potential is primarily a consequence of reduced rod photoreceptor input to RodBCs in the degenerated retina. Frequency response and step input analyses suggest that individual cell responses of RodBCs in either healthy or early-degenerated retina, prior to substantial photoreceptor cell loss, do not differ significantly.
Collapse
Affiliation(s)
- Pragya Kosta
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, USA.
| | - Ege Iseri
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Kyle Loizos
- Institute for Technology and Medical Systems Innovation (ITEMS), Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Javad Paknahad
- Department of Electrical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Rebecca L Pfeiffer
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | | | - James R Anderson
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA.
| | - Gianluca Lazzi
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Institute for Technology and Medical Systems Innovation (ITEMS), Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Electrical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Ophthalmology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Fournel R, Hartveit E, Veruki ML. Differential Contribution of Gap Junctions to the Membrane Properties of ON- and OFF-Bipolar Cells of the Rat Retina. Cell Mol Neurobiol 2021; 41:229-245. [PMID: 32323153 PMCID: PMC7870642 DOI: 10.1007/s10571-020-00845-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/08/2020] [Indexed: 01/31/2023]
Abstract
Gap junctions are ubiquitous within the retina, but in general, it remains to be determined whether gap junction coupling between specific cell types is sufficiently strong to mediate functionally relevant coupling via electrical synapses. From ultrastructural, tracer coupling and immunolabeling studies, there is clear evidence for gap junctions between cone bipolar cells, but it is not known if these gap junctions function as electrical synapses. Here, using whole-cell voltage-clamp recording in rat (male and female) retinal slices, we investigated whether the gap junctions of bipolar cells make a measurable contribution to the membrane properties of these cells. We measured the input resistance (RN) of bipolar cells before and after applying meclofenamic acid (MFA) to block gap junctions. In the presence of MFA, RN of ON-cone bipolar cells displayed a clear increase, paralleled by block of the electrical coupling between these cells and AII amacrine cells in recordings of coupled cell pairs. For OFF-cone and rod bipolar cells, RN did not increase in the presence of MFA. The results for rod bipolar cells are consistent with the lack of gap junctions in these cells. However, for OFF-cone bipolar cells, our results suggest that the morphologically identified gap junctions between these cells do not support a junctional conductance that is sufficient to mediate effective electrical coupling. Instead, these junctions might play a role in chemical and/or metabolic coupling between subcellular compartments.
Collapse
Affiliation(s)
- Rémi Fournel
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| | - Margaret Lin Veruki
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
36
|
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
|
37
|
Threshold vision under full-field stimulation: Revisiting the minimum number of quanta necessary to evoke a visual sensation. Vision Res 2020; 180:1-10. [PMID: 33359896 DOI: 10.1016/j.visres.2020.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 11/21/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022]
Abstract
At the absolute threshold of vision, Hecht, Shlaer and Pirenne estimate that 5-14 photons are absorbed within a retinal area containing ~500 rods. Other estimates of scotopic threshold vision based on stimuli with different durations and focal areas range up to ~100,000 photons. Given that rod density varies with retinal eccentricity and the magnitude of the intrinsic noise increases with increasing stimulus area and duration, here we determine whether the scotopic threshold estimates with focal stimuli can be extended to full-field stimulation and whether summation explains inter-study differences. We show that full-field threshold vision (~1018 mm2, 10 ms duration) is more sensitive than at absolute threshold, requiring the absorption of ~1000 photons across ~91.96 million rods. A summation model is presented integrating our and published data and using a nominal exposure duration, criterion frequency of seeing, rod density, and retinal area that largely explains the inter-study differences and allows estimation of rods per photon ratio for any stimulus size and duration. The highest signal to noise ratio is defined by a peak rod convergence estimated at 53:4:1:2 (rods:rod bipolar cells:AII amacrine cells:retinal ganglion cells), in line with macaque anatomical estimates that show AII amacrine cells form the bottleneck in the rod pathway to set the scotopic visual limit. Our model estimations that the rods per photon ratio under full-field stimulation is ~3000X higher than at absolute threshold are in accordance with visual summation effects and provide an alternative approach for understanding the limits of scotopic vision.
Collapse
|
38
|
Ortuño-Lizarán I, Sánchez-Sáez X, Lax P, Serrano GE, Beach TG, Adler CH, Cuenca N. Dopaminergic Retinal Cell Loss and Visual Dysfunction in Parkinson Disease. Ann Neurol 2020; 88:893-906. [PMID: 32881029 PMCID: PMC10005860 DOI: 10.1002/ana.25897] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Considering the demonstrated implication of the retina in Parkinson disease (PD) pathology and the importance of dopaminergic cells in this tissue, we aimed to analyze the state of the dopaminergic amacrine cells and some of their main postsynaptic neurons in the retina of PD. METHODS Using immunohistochemistry and confocal microscopy, we evaluated morphology, number, and synaptic connections of dopaminergic cells and their postsynaptic cells, AII amacrine and melanopsin-containing retinal ganglion cells, in control and PD eyes from human donors. RESULTS In PD, dopaminergic amacrine cell number was reduced between 58% and 26% in different retinal regions, involving a decline in the number of synaptic contacts with AII amacrine cells (by 60%) and melanopsin cells (by 35%). Despite losing their main synaptic input, AII cells were not reduced in number, but they showed cellular alterations compromising their adequate function: (1) a loss of mitochondria inside their lobular appendages, which may indicate an energetic failure; and (2) a loss of connexin 36, suggesting alterations in the AII coupling and in visual signal transmission from the rod pathway. INTERPRETATION The dopaminergic system impairment and the affection of the rod pathway through the AII cells may explain and be partially responsible for the reduced contrast sensitivity or electroretinographic response described in PD. Also, dopamine reduction and the loss of synaptic contacts with melanopsin cells may contribute to the melanopsin retinal ganglion cell loss previously described and to the disturbances in circadian rhythm and sleep reported in PD patients. These data support the idea that the retina reproduces brain neurodegeneration and is highly involved in PD pathology. ANN NEUROL 2020;88:893-906.
Collapse
Affiliation(s)
- Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | | | | | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
| |
Collapse
|
39
|
Aboelnour A, Noreldin AE, Massoud D, Abumandour MMA. Retinal characterization in the eyes of two bats endemic in the Egyptian fauna, the Egyptian fruit bat (Rousettus aegyptiacus) and insectivorous bat (Pipistrellus kuhlii), using the light microscope and transmission electron microscope. Microsc Res Tech 2020; 83:1391-1400. [PMID: 33405350 DOI: 10.1002/jemt.23530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/02/2020] [Accepted: 05/26/2020] [Indexed: 11/06/2022]
Abstract
Bats are the only mammals that can fly in the dark without eye usage. This study was conducted to describe the structural and functional adaptations of the retina of two bats very common in the Egyptian fauna having a different lifestyle: the Egyptian fruit bat (Rousettus aegyptiacus) and insectivorous bat (Pipistrellus kuhlii). Seven eyes were collected from adult individuals of each species. Examination of the retina using a light microscope and a transmission electron microscope was carried out. The retina of P. kuhlii was thicker than that of R. aegyptiacus, which had many projections extended from the choroid layer into retina forming papillae. Despite rods being dominant in retinae of both species, cone photoreceptors were encountered in both retinae. The outer plexiform layer of R. aegyptiacus was arranged into islets between the outer nuclear layer produced differences in its thickness. However, the retina of P. kuhlii showed a normal arrangement of retinal structure. The retinal pigment epithelium of both bat species consists of a single layer of the cuboidal cells with a round to oval vesicular nuclei, which showed a lack of pigmentation in R. aegyptiacus and poor pigmentation in the P. kuhlii. In conclusion, our investigation detected many structural and ultrastructural differences between the two bat species. The presence of many projections protruded from the choroid layer of R. aegyptiacus retina is considered the most characteristic difference between the retinae of R. aegyptiacus and P. kuhlii.
Collapse
Affiliation(s)
- Asmaa Aboelnour
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt.,Institute of Ophthalmology, University College London, London, United Kingdom
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Diaa Massoud
- Department of Biology, College of Science, Jouf University, Sakaka, Saudi Arabia.,Department of Zoology, Faculty of Science, Fayoum University, Fayoum, Egypt
| | - Mohamed M A Abumandour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
40
|
Cameron MA, Morley JW, Pérez-Fernández V. Seeing the light: different photoreceptor classes work together to drive adaptation in the mammalian retina. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Jin N, Zhang Z, Keung J, Youn SB, Ishibashi M, Tian LM, Marshak DW, Solessio E, Umino Y, Fahrenfort I, Kiyama T, Mao CA, You Y, Wei H, Wu J, Postma F, Paul DL, Massey SC, Ribelayga CP. Molecular and functional architecture of the mouse photoreceptor network. SCIENCE ADVANCES 2020; 6:eaba7232. [PMID: 32832605 PMCID: PMC7439306 DOI: 10.1126/sciadv.aba7232] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/21/2020] [Indexed: 06/11/2023]
Abstract
Mouse photoreceptors are electrically coupled via gap junctions, but the relative importance of rod/rod, cone/cone, or rod/cone coupling is unknown. Furthermore, while connexin36 (Cx36) is expressed by cones, the identity of the rod connexin has been controversial. We report that FACS-sorted rods and cones both express Cx36 but no other connexins. We created rod- and cone-specific Cx36 knockout mice to dissect the photoreceptor network. In the wild type, Cx36 plaques at rod/cone contacts accounted for more than 95% of photoreceptor labeling and paired recordings showed the transjunctional conductance between rods and cones was ~300 pS. When Cx36 was eliminated on one side of the gap junction, in either conditional knockout, Cx36 labeling and rod/cone coupling were almost abolished. We could not detect direct rod/rod coupling, and cone/cone coupling was minor. Rod/cone coupling is so prevalent that indirect rod/cone/rod coupling via the network may account for previous reports of rod coupling.
Collapse
Affiliation(s)
- Nange Jin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joyce Keung
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sean B. Youn
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Undergraduate Program, William Marsh Rice University, Houston, TX, USA
| | - Munenori Ishibashi
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lian-Ming Tian
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David W. Marshak
- Department of Neurobiology and Anatomy, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eduardo Solessio
- Center for Vision Research and SUNY Eye Institute, Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yumiko Umino
- Center for Vision Research and SUNY Eye Institute, Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Iris Fahrenfort
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanan You
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Jiaqian Wu
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Friso Postma
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - David L. Paul
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - Stephen C. Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Elizabeth Morford Distinguished Chair in Ophthalmology and Research Director, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christophe P. Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cellular Biology, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Bernice Weingarten Chair in Ophthalmology, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
42
|
Zhang S, Lyuboslavsky P, Dixon JA, Chrenek MA, Sellers JT, Hamm JM, Ribelayga CP, Zhang Z, Le YZ, Iuvone PM. Effects of Cone Connexin-36 Disruption on Light Adaptation and Circadian Regulation of the Photopic ERG. Invest Ophthalmol Vis Sci 2020; 61:24. [PMID: 32531058 PMCID: PMC7415284 DOI: 10.1167/iovs.61.6.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/07/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose The present study tested the hypothesis that connexin-36 (Cx36) and gap junctions between photoreceptor cells contribute to the circadian rhythm of the photopic electroretinogram (ERG) b-wave amplitude. Methods Cone-specific disruption of Cx36 was obtained in mice with a floxed Gjd2 gene and human red/green pigment promoter (HRGP)-driven Cre recombinase. Standard ERG, spectral-domain optical coherence tomography (SD-OCT) and histochemical methods were used. Results HRGPcreGjd2fl/fl mice had a selective reduction in Cx36 protein in the outer plexiform layer; no reduction in Cx36 was observed in the inner plexiform layer. Cx36 disruption had no effect on the number of cones, the thickness of the photoreceptor layer, or the scotopic ERG responses. However, there was a reduction of the photopic ERG circadian rhythm, with b-wave amplitudes in the day and the night locked in the daytime, light-adapted state. In HRGPcreGjd2+/+and Gjd2fl/fl controls, the circadian rhythm of light-adapted ERG persisted, similar to that in wild type mice. Conclusions Cx36 regulation contributes to the circadian rhythm of light-adapted ERG; in the absence of photoreceptor gap junctions, mice appear to be in a fully light-adapted state regardless of the time of day. The higher amplitudes and reduced circadian regulation of the b-wave of HRGPcreGjd2fl/fl mice may be due to increased synaptic strength at the cone to ON bipolar cell synapse due to electrotonic isolation of the terminals lacking gap junctions.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Capital Medical University, Beijing, China
| | - Polina Lyuboslavsky
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
| | - Jendayi Azeezah Dixon
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
| | - Jana T. Sellers
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
| | - Jessica M. Hamm
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
| | - Christophe P. Ribelayga
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Yun Z. Le
- Departments of Medicine, Cell Biology, and Ophthalmology and Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - P. Michael Iuvone
- Department of Ophthalmology, Emory University, School of Medicine, Atlanta, Georgia, United States
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
43
|
Comparison of AAV-Mediated Optogenetic Vision Restoration between Retinal Ganglion Cell Expression and ON Bipolar Cell Targeting. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:15-23. [PMID: 32548211 PMCID: PMC7287188 DOI: 10.1016/j.omtm.2020.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 01/06/2023]
Abstract
The loss of photoreceptors in individuals with retinal degenerative diseases leads to partial or complete blindness. Optogenetic therapy is a promising approach for restoring vision to the blind. Multiple strategies have been employed by targeting genetically encoded light sensors, particularly channelrhodopsins, to surviving retinal neurons in animal models. In particular, the strategy of targeting retinal bipolar cells has commonly been expected to result in better vision than ubiquitous expression in retinal ganglion cells. However, a direct comparison of the channelrhodopsin-restored vision between these two strategies has not been performed. Here, we compared the restored visual functions achieved by adeno-associated virus (AAV)-mediated expression of a channelrhodopsin in ON-type bipolar cells and retinal ganglion cells driven by an improved mGluR6 promoter and a CAG promoter, respectively, in a blind mouse model by performing electrophysiological recordings and behavioral assessments. Unexpectedly, the efficacy of the restored vision based on light sensitivity and visual acuity was much higher following ubiquitous retinal ganglion cell expression than that of the strategy targeting ON-type bipolar cells. Our study suggests that, at least based on currently available gene delivery techniques, the expression of genetically encoded light sensors in retinal ganglion cells is likely a practical and advantageous strategy for optogenetic vision restoration.
Collapse
|
44
|
Gorbatyuk MS, Starr CR, Gorbatyuk OS. Endoplasmic reticulum stress: New insights into the pathogenesis and treatment of retinal degenerative diseases. Prog Retin Eye Res 2020; 79:100860. [PMID: 32272207 DOI: 10.1016/j.preteyeres.2020.100860] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.
Collapse
Affiliation(s)
- Marina S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA.
| | - Christopher R Starr
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| | - Oleg S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| |
Collapse
|
45
|
Bligard GW, DeBrecht J, Smith RG, Lukasiewicz PD. Light-evoked glutamate transporter EAAT5 activation coordinates with conventional feedback inhibition to control rod bipolar cell output. J Neurophysiol 2020; 123:1828-1837. [PMID: 32233906 DOI: 10.1152/jn.00527.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the retina, modulation of the amplitude of dim visual signals primarily occurs at axon terminals of rod bipolar cells (RBCs). GABA and glycine inhibitory neurotransmitter receptors and the excitatory amino acid transporter 5 (EAAT5) modulate the RBC output. EAATs clear glutamate from the synapse, but they also have a glutamate-gated chloride conductance. EAAT5 acts primarily as an inhibitory glutamate-gated chloride channel. The relative role of visually evoked EAAT5 inhibition compared with GABA and glycine inhibition has not been addressed. In this study, we determine the contribution of EAAT5-mediated inhibition onto RBCs in response to light stimuli in mouse retinal slices. We find differences and similarities in the two forms of inhibition. Our results show that GABA and glycine mediate nearly all lateral inhibition onto RBCs, as EAAT5 is solely a mediator of RBC feedback inhibition. We also find that EAAT5 and conventional GABA inhibition both contribute to feedback inhibition at all stimulus intensities. Finally, our in silico modeling compares and contrasts EAAT5-mediated to GABA- and glycine-mediated feedback inhibition. Both forms of inhibition have a substantial impact on synaptic transmission to the postsynaptic AII amacrine cell. Our results suggest that the late phase EAAT5 inhibition acts with the early phase conventional, reciprocal GABA inhibition to modulate the rod signaling pathway between rod bipolar cells and their downstream synaptic targets.NEW & NOTEWORTHY Excitatory amino acid transporter 5 (EAAT5) glutamate transporters have a chloride channel that is strongly activated by glutamate, which modulates excitatory signaling. We found that EAAT5 is a major contributor to feedback inhibition on rod bipolar cells. Inhibition to rod bipolar cells is also mediated by GABA and glycine. GABA and glycine mediate the early phase of feedback inhibition, and EAAT5 mediates a more delayed inhibition. Together, inhibitory transmitters and EAAT5 coordinate to mediate feedback inhibition, controlling neuronal output.
Collapse
Affiliation(s)
- Gregory W Bligard
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri
| | - James DeBrecht
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri
| | - Robert G Smith
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter D Lukasiewicz
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri.,Department of Neuroscience, Washington University, St. Louis, Missouri
| |
Collapse
|
46
|
Grünert U, Martin PR. Cell types and cell circuits in human and non-human primate retina. Prog Retin Eye Res 2020; 78:100844. [PMID: 32032773 DOI: 10.1016/j.preteyeres.2020.100844] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
This review summarizes our current knowledge of primate including human retina focusing on bipolar, amacrine and ganglion cells and their connectivity. We have two main motivations in writing. Firstly, recent progress in non-invasive imaging methods to study retinal diseases mean that better understanding of the primate retina is becoming an important goal both for basic and for clinical sciences. Secondly, genetically modified mice are increasingly used as animal models for human retinal diseases. Thus, it is important to understand to which extent the retinas of primates and rodents are comparable. We first compare cell populations in primate and rodent retinas, with emphasis on how the fovea (despite its small size) dominates the neural landscape of primate retina. We next summarise what is known, and what is not known, about the postreceptoral neurone populations in primate retina. The inventories of bipolar and ganglion cells in primates are now nearing completion, comprising ~12 types of bipolar cell and at least 17 types of ganglion cell. Primate ganglion cells show clear differences in dendritic field size across the retina, and their morphology differs clearly from that of mouse retinal ganglion cells. Compared to bipolar and ganglion cells, amacrine cells show even higher morphological diversity: they could comprise over 40 types. Many amacrine types appear conserved between primates and mice, but functions of only a few types are understood in any primate or non-primate retina. Amacrine cells appear as the final frontier for retinal research in monkeys and mice alike.
Collapse
Affiliation(s)
- Ulrike Grünert
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia.
| | - Paul R Martin
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
47
|
Seilheimer RL, Sabharwal J, Wu SM. Genetic dissection of rod and cone pathways mediating light responses and receptive fields of ganglion cells in the mouse retina. Vision Res 2020; 167:15-23. [PMID: 31887538 PMCID: PMC7264069 DOI: 10.1016/j.visres.2019.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/14/2019] [Accepted: 12/14/2019] [Indexed: 10/25/2022]
Abstract
Retinal ganglion cells (GCs) are important visual neurons which carry complex spatiotemporal information from the retina to higher visual centers in the brain. By taking advantage of pathway-specific knockout/mutant mice and multi-electrode array (MEA) recording techniques, we analyze contributions of rod and cone pathways to responsiveness, kinetics and receptive field profiles of GCs under scotopic and photopic conditions. Our data suggest: (1) Scotopic responses of some GCs require all three rod pathways, some require only the secondary and tertiary rod pathways, and others require only the tertiary rod pathway. (2) There are more responsive GCs in photopic conditions than responsive GCs in scotopic conditions. (3) Gap junctions slow down GCs' scotopic light responses and increase GCs' ratio of antagonistic to center inputs. (4) Cone pathways do not affect the kinetics but alter the ratio of antagonistic to center inputs of scotopic GC responses, and they speed up GCs photopic responses and alter the ratio of GCs' antagonistic to center synaptic inputs and receptive field profiles. (5) Rod bipolar cells shorten response latency of ON GCs and increase the ratio of GCs' antagonistic to center synaptic inputs. (6) Light adaptation speeds up GCs' temporal processing and tunes GC photopic responses to higher frequencies, and the tertiary rod pathway plays a significant role in adaptation-induced TTP changes in some GCs. (7) GC RF center sizes are partially mediated by AIIACs and GC-GC coupling. (8) Connexin36 gap junctions and cone pathways alter synaptic circuits underlying antagonistic surround inputs to GCs in photopic conditions.
Collapse
Affiliation(s)
- R L Seilheimer
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - J Sabharwal
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - S M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
48
|
Goaillard JM, Moubarak E, Tapia M, Tell F. Diversity of Axonal and Dendritic Contributions to Neuronal Output. Front Cell Neurosci 2020; 13:570. [PMID: 32038171 PMCID: PMC6987044 DOI: 10.3389/fncel.2019.00570] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/09/2019] [Indexed: 11/13/2022] Open
Abstract
Our general understanding of neuronal function is that dendrites receive information that is transmitted to the axon, where action potentials (APs) are initiated and propagated to eventually trigger neurotransmitter release at synaptic terminals. Even though this canonical division of labor is true for a number of neuronal types in the mammalian brain (including neocortical and hippocampal pyramidal neurons or cerebellar Purkinje neurons), many neuronal types do not comply with this classical polarity scheme. In fact, dendrites can be the site of AP initiation and propagation, and even neurotransmitter release. In several interneuron types, all functions are carried out by dendrites as these neurons are devoid of a canonical axon. In this article, we present a few examples of "misbehaving" neurons (with a non-canonical polarity scheme) to highlight the diversity of solutions that are used by mammalian neurons to transmit information. Moreover, we discuss how the contribution of dendrites and axons to neuronal excitability may impose constraints on the morphology of these compartments in specific functional contexts.
Collapse
Affiliation(s)
- Jean-Marc Goaillard
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Estelle Moubarak
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Mónica Tapia
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Fabien Tell
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
49
|
Ko GYP. Circadian regulation in the retina: From molecules to network. Eur J Neurosci 2020; 51:194-216. [PMID: 30270466 PMCID: PMC6441387 DOI: 10.1111/ejn.14185] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/16/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
The mammalian retina is the most unique tissue among those that display robust circadian/diurnal oscillations. The retina is not only a light sensing tissue that relays light information to the brain, it has its own circadian "system" independent from any influence from other circadian oscillators. While all retinal cells and retinal pigment epithelium (RPE) possess circadian oscillators, these oscillators integrate by means of neural synapses, electrical coupling (gap junctions), and released neurochemicals (such as dopamine, melatonin, adenosine, and ATP), so the whole retina functions as an integrated circadian system. Dysregulation of retinal clocks not only causes retinal or ocular diseases, it also impacts the circadian rhythm of the whole body, as the light information transmitted from the retina entrains the brain clock that governs the body circadian rhythms. In this review, how circadian oscillations in various retinal cells are integrated, and how retinal diseases affect daily rhythms.
Collapse
Affiliation(s)
- Gladys Y-P Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas
| |
Collapse
|
50
|
Lee SCS, Martin PR, Grünert U. Topography of Neurons in the Rod Pathway of Human Retina. Invest Ophthalmol Vis Sci 2019; 60:2848-2859. [PMID: 31260035 DOI: 10.1167/iovs.19-27217] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The objective of this study was to map the distribution and density of the three major components of the classical scotopic "night vision" pathway (rods, rod bipolar, and AII amacrine cells) in postmortem human retinas. Methods Four postmortem donor eyes (male and female, aged 44-56 years) were used to cut vertical sections through the temporal horizontal meridian. The sections were processed for immunohistochemistry and imaged using high-resolution multichannel confocal microscopy. Rods, rod bipolar, and AII amacrine cells were counted along the temporal horizontal meridian. Two additional retinas were used for intracellular injections. Results Rod peak density is close to 150,000 cells/mm2 at 4 to 5 mm (15° to 20°) eccentricity, declining to below 70,000 cells/mm2 in peripheral retina. Rod bipolar density is lower but follows a similar distribution with peak density near 10,000 cells/mm2 between 2 and 4 mm (7° to 15°) eccentricity declining to below 4000 cells/mm2 in peripheral retina. The peak density of AII amacrine cells (near 4000 cells/mm2) is located close to the fovea, at 0.5- to 2 mm-eccentricity (2° to 7°) and declines to below 1000 cells/mm2 in the periphery. Thus, convergence between rods and AII cells increases from central to peripheral retina. Conclusions Comparison with human psychophysics and ganglion cell density indicates that the spatial resolution of scotopic vision is limited by the AII mosaic at eccentricities below 15° and by the midget ganglion cell mosaic at eccentricities above 15°.
Collapse
Affiliation(s)
- Sammy C S Lee
- The University of Sydney, Faculty of Medicine and Health, Save Sight Institute and Discipline of Clinical Ophthalmology, Sydney, New South Wales, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul R Martin
- The University of Sydney, Faculty of Medicine and Health, Save Sight Institute and Discipline of Clinical Ophthalmology, Sydney, New South Wales, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, Australia
| | - Ulrike Grünert
- The University of Sydney, Faculty of Medicine and Health, Save Sight Institute and Discipline of Clinical Ophthalmology, Sydney, New South Wales, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|