1
|
Shen J, Xue T. Neural-circuit architecture underlying non-image-forming visual functions. Curr Opin Neurobiol 2025; 93:103052. [PMID: 40414167 DOI: 10.1016/j.conb.2025.103052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025]
Abstract
Perceiving and responding to environmental cues underpins survival and cognition. Light, emerging as one of the most ancient and powerful signals, has shaped life on Earth for billions of years. In mammals, light information is primarily detected by retinal photoreceptors: rods, cones, and intrinsically photosensitive retinal ganglion cells. While rods and cones enable image-forming vision, evolution has preserved and extended evolutionarily ancient yet critical non-image-forming visual functions, including circadian photoentrainment, pupillary light reflexes, and light-mediated modulation of metabolism, mood, and neurodevelopment. Although non-image-forming visual functions have been partially characterized in humans and model organisms, our understanding of the neural circuit mechanisms by which light orchestrates diverse behavior remains fragmented. The discovery of ipRGCs, combined with recent advances in systems neuroscience tools, has yielded critical breakthroughs in three domains: (1) light information encoding within photoreceptors, (2) systematic mapping of retinofugal pathways, and (3) central mechanisms of light-regulated physiological functions. These advances have progressively unraveled causal relationships between non-image-forming visual functions and their underlying eye-brain circuitry. This review summarizes groundbreaking progress in the three domains discussed above, highlighting key unresolved questions in the field.
Collapse
Affiliation(s)
- Jiawei Shen
- Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; State Key Laboratory of Eye Health, Institute of Advanced Technology, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
2
|
Duda S, Block CT, Pradhan DR, Arzhangnia Y, Klaiber A, Greschner M, Puller C. Spatial distribution and functional integration of displaced retinal ganglion cells. Sci Rep 2025; 15:7123. [PMID: 40016499 PMCID: PMC11868576 DOI: 10.1038/s41598-025-91045-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
The retina contains distinct types of ganglion cells, which form mosaics with cells of each type at each position of the visual field. Displaced retinal ganglion cells (dRGCs) occur with cell bodies in the inner nuclear layer (INL), and regularly placed RGCs with cell bodies in the ganglion cell layer. An example of mammalian dRGCs are M1-type intrinsically photosensitive ganglion cells (ipRGCs). Little is known, however, about their relationship with regularly placed ipRGCs. We identified mouse ipRGC types M1, M2, and M4/sONɑ by immunohistochemistry and light microscopy. Reconstruction of immunolabeled mosaics from M1 and sONɑ RGCs indicated that dRGCs tiled the retina with their regular RGC partners. Multi-electrode array recordings revealed conventional receptive fields of displaced sONɑ RGCs which fit into the mosaic of their regular counterparts. An RGC distribution analysis showed type-specific dRGC patterns which followed neither the global density distribution of all RGCs nor the local densities of corresponding cell types. The displacement of RGC bodies into the INL occurs in a type-dependent manner, where dRGCs are positioned to form complete mosaics with their regular partners. Our data suggest that dRGCs and regular RGCs serve the same functional role within their corresponding population of RGCs.
Collapse
Affiliation(s)
- Sabrina Duda
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Christoph T Block
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Dipti R Pradhan
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Yousef Arzhangnia
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Alina Klaiber
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Martin Greschner
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany
| | - Christian Puller
- Visual Neuroscience, Department of Neuroscience, Carl von Ossietzky University, 26111, Oldenburg, Germany.
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany.
| |
Collapse
|
3
|
Budoff SA, Poleg-Polsky A. A Complete Spatial Map of Mouse Retinal Ganglion Cells Reveals Density and Gene Expression Specializations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637538. [PMID: 39990332 PMCID: PMC11844403 DOI: 10.1101/2025.02.10.637538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information from the eye to the brain. In mice, several RGC subtypes show nonuniform spatial distributions, potentially mediating specific visual functions. However, the full extent of RGC specialization remains unknown. Here, we used en-face cryosectioning, spatial transcriptomics, and machine learning to map the spatial distribution of all RGC subtypes identified in previous single-cell studies. While two-thirds of RGC subtypes were evenly distributed, others showed strong biases toward ventral or dorso-temporal regions associated with sky vision and the area retinae temporalis (ART), the predicted homolog of the area centralis. Additionally, we observed unexpected spatial variation in gene expression within several subtypes along the dorso-ventral axis or within vs. outside the ART, independent of RGC density profiles. Finally, we found limited correlations between the gene profiles of the ART and the primate macula, suggesting divergent specialization between the mouse and primate central vision.
Collapse
Affiliation(s)
- Samuel A. Budoff
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| | - Alon Poleg-Polsky
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| |
Collapse
|
4
|
Kinder L, Lindner M. Expression of Osteopontin in M2 and M4 Intrinsically Photosensitive Retinal Ganglion Cells in the Mouse Retina. Invest Ophthalmol Vis Sci 2025; 66:14. [PMID: 39908128 PMCID: PMC11804889 DOI: 10.1167/iovs.66.2.14] [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] [Received: 10/31/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025] Open
Abstract
Purpose Melanopsin-expressing intrinsically photosensitive (ip) retinal ganglion cells (RGCs) can be divided into six different subtypes (M1 - M6). Yet, specific markers exist for only some of these subtypes that could be employed to study the function of individual subtypes. Osteopontin (Spp1) marks αRGC, suggesting that, across ipRGCs, it would only mark the M4-ipRGC subtype (synonymous to ON-sustained αRGCs). Recent evidence suggests that osteopontin expression could spread to other ipRGC subtypes. Therefore, this study aims to characterize the expression pattern of osteopontin across ipRGC subtypes in mice. Methods Single-cell RNA (scRNA-seq) sequencing data from murine RGCs were analyzed to identify expression patterns of Spp1 across ipRGCs. Immunohistochemistry (IHC) was performed on retinal cryosections and flatmounts from C57BL/6J mice to characterize the localization of osteopontin across ipRGCs. Neurite tracing was employed to study dendritic morphology and identify individual ipRGC subtypes. Results scRNA-seq analysis revealed Spp1 expression in two distinct clusters of ipRGCs. IHC confirmed osteopontin colocalization with neurofilament heavy chain, an established marker for αRGCs, including M4-ipRGCs. Spp1 immunoreactivity was moreover identified in one additional group of ipRGCs. By dendritic morphology and stratification, those cells were clearly identified as M2-ipRGCs. Conclusions Our findings demonstrate that osteopontin is expressed in both M2- and M4-ipRGCs, challenging the notion of osteopontin as a marker exclusively for αRGCs. IHC double-labeling for osteopontin and melanopsin provides a novel method to identify and differentiate M2 ipRGCs from other subtypes. This will support the study of ipRGC physiology in a subtype -specific manner and may, for instance, foster research in the field of optic nerve injury.
Collapse
Affiliation(s)
- Leonie Kinder
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps-University, Marburg, Germany
| | - Moritz Lindner
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps-University, Marburg, Germany
- The Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Department of Ophthalmology, Philipps-University, University Hospital of Giessen and Marburg GmbH, Marburg Campus, Marburg, Germany
| |
Collapse
|
5
|
Grimes WN, Berson DM, Sabnis A, Hoon M, Sinha R, Tian H, Diamond JS. Layer-specific anatomical and physiological features of the retina's neurovascular unit. Curr Biol 2025; 35:109-120.e4. [PMID: 39689705 PMCID: PMC11867051 DOI: 10.1016/j.cub.2024.11.023] [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: 04/08/2024] [Revised: 09/22/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
The neurovascular unit (NVU), comprising vascular, glial, and neural elements, supports the energetic demands of neural computation, but this aspect of the retina's trilaminar vessel network is poorly understood. Only the innermost vessel layer-the superficial vascular plexus (SVP)-is associated with astrocytes, like brain capillaries, whereas radial Müller glia interact with vessels in the other layers. Using serial electron microscopic reconstructions from mouse and primate retina, we find that Müller processes cover capillaries in a tessellating pattern, mirroring the wrapping of brain capillaries by tiled astrocytic endfeet. Gaps in the Müller sheath, found mainly in the intermediate vascular plexus (IVP), permit diverse neuron types to contact pericytes and the endothelial cells directly. Pericyte somata are a favored target, often at spine-like structures with reduced or absent vascular basement lamina. Focal application of ATP to the vitreal surface evoked Ca2+ signals in Müller sheaths in all three vascular layers. Pharmacological experiments confirmed that Müller sheaths express purinergic receptors that, when activated, trigger intracellular Ca2+ signals that are amplified by inositol triphosphate (IP3)-controlled intracellular Ca2+ stores. When rod photoreceptors die in a mouse model of retinitis pigmentosa (rd10), Müller sheaths dissociate from the deep vascular plexus (DVP) but are largely unchanged within the IVP or SVP. Thus, Müller glia interact with retinal vessels in a laminar, compartmentalized manner: glial sheaths are virtually complete in the SVP but fenestrated in the IVP, permitting direct neurovascular contacts. In the DVP, the glial sheath is only modestly fenestrated and is vulnerable to photoreceptor degeneration.
Collapse
Affiliation(s)
- William N Grimes
- Synaptic Physiology Section, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD 20814, USA.
| | - David M Berson
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Adit Sabnis
- Synaptic Physiology Section, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Raunak Sinha
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hua Tian
- Synaptic Physiology Section, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
| | - Jeffrey S Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD 20814, USA.
| |
Collapse
|
6
|
Zeng Y, Rong R, You M, Zhu P, Zhang J, Xia X. Light-eye-body axis: exploring the network from retinal illumination to systemic regulation. Theranostics 2025; 15:1496-1523. [PMID: 39816683 PMCID: PMC11729557 DOI: 10.7150/thno.106589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025] Open
Abstract
The human body is an intricate system, where diverse and complex signaling among different organs sustains physiological activities. The eye, as a primary organ for information acquisition, not only plays a crucial role in visual perception but also, as increasing evidence suggests, exerts a broad influence on the entire body through complex circuits upon receiving light signals which is called non-image-forming vision. However, the extent and mechanisms of light's impact on the body through the eyes remain insufficiently explored. There is also a dearth of comprehensive reviews elucidating the intricate interplay between light, the eye, and the systemic connections to the entire body. Herein, we propose the concept of the light-eye-body axis to systematically encapsulate the extensive non-image-forming effects of light signals received by the retina on the entire body. We reviewed the visual-neural structure basis of the light-eye-body axis, summarized the mechanism by which the eyes regulate the whole body and the current research status and challenges within the physiological and pathological processes involved in the light-eye-body axis. Future research should aim to expand the influence of the light-eye-body axis and explore its deeper mechanisms. Understanding and investigating the light-eye-body axis will contribute to improving lighting conditions to optimize health and guide the establishment of phototherapy standards in clinical practice.
Collapse
Affiliation(s)
- Yi Zeng
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Rong Rong
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Mengling You
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Peng Zhu
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jinglin Zhang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, 410008, P.R. China
- National clinical key specialty of ophthalmology, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Central South University, Changsha, Hunan, 410008, P.R. China
| |
Collapse
|
7
|
Höfling L, Szatko KP, Behrens C, Deng Y, Qiu Y, Klindt DA, Jessen Z, Schwartz GW, Bethge M, Berens P, Franke K, Ecker AS, Euler T. A chromatic feature detector in the retina signals visual context changes. eLife 2024; 13:e86860. [PMID: 39365730 PMCID: PMC11452179 DOI: 10.7554/elife.86860] [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: 02/09/2023] [Accepted: 08/25/2024] [Indexed: 10/06/2024] Open
Abstract
The retina transforms patterns of light into visual feature representations supporting behaviour. These representations are distributed across various types of retinal ganglion cells (RGCs), whose spatial and temporal tuning properties have been studied extensively in many model organisms, including the mouse. However, it has been difficult to link the potentially nonlinear retinal transformations of natural visual inputs to specific ethological purposes. Here, we discover a nonlinear selectivity to chromatic contrast in an RGC type that allows the detection of changes in visual context. We trained a convolutional neural network (CNN) model on large-scale functional recordings of RGC responses to natural mouse movies, and then used this model to search in silico for stimuli that maximally excite distinct types of RGCs. This procedure predicted centre colour opponency in transient suppressed-by-contrast (tSbC) RGCs, a cell type whose function is being debated. We confirmed experimentally that these cells indeed responded very selectively to Green-OFF, UV-ON contrasts. This type of chromatic contrast was characteristic of transitions from ground to sky in the visual scene, as might be elicited by head or eye movements across the horizon. Because tSbC cells performed best among all RGC types at reliably detecting these transitions, we suggest a role for this RGC type in providing contextual information (i.e. sky or ground) necessary for the selection of appropriate behavioural responses to other stimuli, such as looming objects. Our work showcases how a combination of experiments with natural stimuli and computational modelling allows discovering novel types of stimulus selectivity and identifying their potential ethological relevance.
Collapse
Affiliation(s)
- Larissa Höfling
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
| | - Klaudia P Szatko
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
| | - Christian Behrens
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Yuyao Deng
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
| | - Yongrong Qiu
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
| | | | - Zachary Jessen
- Feinberg School of Medicine, Department of Ophthalmology, Northwestern UniversityChicagoUnited States
| | - Gregory W Schwartz
- Feinberg School of Medicine, Department of Ophthalmology, Northwestern UniversityChicagoUnited States
| | - Matthias Bethge
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Tübingen AI Center, University of TübingenTübingenGermany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Tübingen AI Center, University of TübingenTübingenGermany
- Hertie Institute for AI in Brain HealthTübingenGermany
| | - Katrin Franke
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Alexander S Ecker
- Institute of Computer Science and Campus Institute Data Science, University of GöttingenGöttingenGermany
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Centre for Integrative Neuroscience, University of TübingenTübingenGermany
| |
Collapse
|
8
|
Franke K, Cai C, Ponder K, Fu J, Sokoloski S, Berens P, Tolias AS. Asymmetric distribution of color-opponent response types across mouse visual cortex supports superior color vision in the sky. eLife 2024; 12:RP89996. [PMID: 39234821 PMCID: PMC11377037 DOI: 10.7554/elife.89996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Color is an important visual feature that informs behavior, and the retinal basis for color vision has been studied across various vertebrate species. While many studies have investigated how color information is processed in visual brain areas of primate species, we have limited understanding of how it is organized beyond the retina in other species, including most dichromatic mammals. In this study, we systematically characterized how color is represented in the primary visual cortex (V1) of mice. Using large-scale neuronal recordings and a luminance and color noise stimulus, we found that more than a third of neurons in mouse V1 are color-opponent in their receptive field center, while the receptive field surround predominantly captures luminance contrast. Furthermore, we found that color-opponency is especially pronounced in posterior V1 that encodes the sky, matching the statistics of natural scenes experienced by mice. Using unsupervised clustering, we demonstrate that the asymmetry in color representations across cortex can be explained by an uneven distribution of green-On/UV-Off color-opponent response types that are represented in the upper visual field. Finally, a simple model with natural scene-inspired parametric stimuli shows that green-On/UV-Off color-opponent response types may enhance the detection of 'predatory'-like dark UV-objects in noisy daylight scenes. The results from this study highlight the relevance of color processing in the mouse visual system and contribute to our understanding of how color information is organized in the visual hierarchy across species.
Collapse
Affiliation(s)
- Katrin Franke
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, United States
- Stanford Bio-X, Stanford University, Stanford, United States
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, United States
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, United States
| | - Chenchen Cai
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Graduate Training Center of Neuroscience, International Max Planck Research School, University of Tübingen, Tübingen, Germany
| | - Kayla Ponder
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, United States
| | - Jiakun Fu
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, United States
| | - Sacha Sokoloski
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Andreas Savas Tolias
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, United States
- Stanford Bio-X, Stanford University, Stanford, United States
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, United States
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, United States
- Department of Electrical Engineering, Stanford University, Stanford, United States
| |
Collapse
|
9
|
Ling Y, Wang Y, Ye J, Luan C, Bi A, Gu Y, Shi X. Changes in Intrinsically Photosensitive Retinal Ganglion Cells, Dopaminergic Amacrine Cells, and Their Connectivity in the Retinas of Lid Suture Myopia. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39230992 PMCID: PMC11379095 DOI: 10.1167/iovs.65.11.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Purpose This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.
Collapse
Affiliation(s)
- Ying Ling
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Ye
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changlin Luan
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ailing Bi
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuefeng Shi
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
10
|
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
|
11
|
Dyer B, Yu SO, Brown RL, Lang RA, D'Souza SP. Defining spatial nonuniformities of all ipRGC types using an improved Opn4 cre recombinase mouse line. CELL REPORTS METHODS 2024; 4:100837. [PMID: 39127043 PMCID: PMC11384080 DOI: 10.1016/j.crmeth.2024.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/18/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study, we generate an improved Opn4cre knockin allele (Opn4cre(DSO)), which faithfully reproduces endogenous Opn4 expression and improves compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6) and defined their unique topographical distribution across the retina. In the brain, the Opn4cre(DSO) line labels ipRGC projections with minimal labeling of cell bodies. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.
Collapse
Affiliation(s)
- Brannen Dyer
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sue O Yu
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - R Lane Brown
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Richard A Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
12
|
Franke K, Cai C, Ponder K, Fu J, Sokoloski S, Berens P, Tolias AS. Asymmetric distribution of color-opponent response types across mouse visual cortex supports superior color vision in the sky. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.01.543054. [PMID: 37333280 PMCID: PMC10274736 DOI: 10.1101/2023.06.01.543054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Color is an important visual feature that informs behavior, and the retinal basis for color vision has been studied across various vertebrate species. While many studies have investigated how color information is processed in visual brain areas of primate species, we have limited understanding of how it is organized beyond the retina in other species, including most dichromatic mammals. In this study, we systematically characterized how color is represented in the primary visual cortex (V1) of mice. Using large-scale neuronal recordings and a luminance and color noise stimulus, we found that more than a third of neurons in mouse V1 are color-opponent in their receptive field center, while the receptive field surround predominantly captures luminance contrast. Furthermore, we found that color-opponency is especially pronounced in posterior V1 that encodes the sky, matching the statistics of natural scenes experienced by mice. Using unsupervised clustering, we demonstrate that the asymmetry in color representations across cortex can be explained by an uneven distribution of green-On/UV-Off color-opponent response types that are represented in the upper visual field. Finally, a simple model with natural scene-inspired parametric stimuli shows that green-On/UV-Off color-opponent response types may enhance the detection of "predatory"-like dark UV-objects in noisy daylight scenes. The results from this study highlight the relevance of color processing in the mouse visual system and contribute to our understanding of how color information is organized in the visual hierarchy across species.
Collapse
Affiliation(s)
- Katrin Franke
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA, US
- Stanford Bio-X, Stanford University, Stanford, CA, US
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, US
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
| | - Chenchen Cai
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Graduate Training Center of Neuroscience, International Max Planck Research School, University of Tübingen, Tübingen, Germany
| | - Kayla Ponder
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
| | - Jiakun Fu
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
| | - Sacha Sokoloski
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Andreas S Tolias
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA, US
- Stanford Bio-X, Stanford University, Stanford, CA, US
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, US
- Department of Neuroscience & Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, US
| |
Collapse
|
13
|
Contreras E, Liang C, Mahoney HL, Javier JL, Luce ML, Labastida Medina K, Bozza T, Schmidt TM. Flp-recombinase mouse line for genetic manipulation of ipRGCs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592761. [PMID: 38766000 PMCID: PMC11100754 DOI: 10.1101/2024.05.06.592761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Light has myriad impacts on behavior, health, and physiology. These signals originate in the retina and are relayed to the brain by more than 40 types of retinal ganglion cells (RGCs). Despite a growing appreciation for the diversity of RGCs, how these diverse channels of light information are ultimately integrated by the ~50 retinorecipient brain targets to drive these light-evoked effects is a major open question. This gap in understanding primarily stems from a lack of genetic tools that specifically label, manipulate, or ablate specific RGC types. Here, we report the generation and characterization of a new mouse line (Opn4FlpO), in which FlpO is expressed from the Opn4 locus, to manipulate the melanopsin-expressing, intrinsically photosensitive retinal ganglion cells. We find that the Opn4FlpO line, when crossed to multiple reporters, drives expression that is confined to ipRGCs and primarily labels the M1-M3 subtypes. Labeled cells in this mouse line show the expected intrinsic, melanopsin-based light response and morphological features consistent with the M1-M3 subtypes. In alignment with the morphological and physiological findings, we see strong innervation of non-image forming brain targets by ipRGC axons, and weaker innervation of image forming targets in Opn4FlpO mice labeled using AAV-based and FlpO-reporter lines. Consistent with the FlpO insertion disrupting the endogenous Opn4 transcript, we find that Opn4FlpO/FlpO mice show deficits in the pupillary light reflex, demonstrating their utility for behavioral research in future experiments. Overall, the Opn4FlpO mouse line drives Flp-recombinase expression that is confined to ipRGCs and most effectively drives recombination in M1-M3 ipRGCs. This mouse line will be of broad use to those interested in manipulating ipRGCs through a Flp-based recombinase for intersectional studies or in combination with other, non-Opn4 Cre driver lines.
Collapse
Affiliation(s)
- E Contreras
- Department of Neurobiology, Northwestern University, Evanston, IL
- Northwestern University Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, United States
| | - C Liang
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - H L Mahoney
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - J L Javier
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - M L Luce
- Department of Neurobiology, Northwestern University, Evanston, IL
| | | | - T Bozza
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - T M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL
- Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
14
|
Dyer B, Yu SO, Lane Brown R, Lang RA, D’Souza SP. A new Opn4cre recombinase mouse line to target intrinsically photosensitive retinal ganglion cells (ipRGCs). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589750. [PMID: 38659888 PMCID: PMC11042346 DOI: 10.1101/2024.04.16.589750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study we generate a new Opn4cre knock-in allele (Opn4cre(DSO)), in which cre is placed immediately downstream of the Opn4 start codon. This approach aims to faithfully reproduce endogenous Opn4 expression and improve compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain, and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs, with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6). Using this new tool, we describe the topographical distributions of ipRGC types across the retinal landscape, uncovering distinct ventronasal biases for M5 and M6 types, setting them apart from their M1-M4 counterparts. In the brain, we find vastly different labeling patterns between lines, with Opn4cre(DSO) only labeling ipRGC axonal projections to their targets. The combination of off-target effects of Opn4cre(Saha) across the retina and brain, coupled with diminished efficiencies of both Cre lines when coupled to less sensitive reporters, underscores the need for careful consideration in experimental design and validation with any Opn4cre driver. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.
Collapse
Affiliation(s)
- Brannen Dyer
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| | - Sue O. Yu
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - R. Lane Brown
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
- Department of Ophthalmology, University of Cincinnati, OH
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, OH
- Science of Light Center, Cincinnati Children’s Hospital Medical Center, OH
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, OH
| |
Collapse
|
15
|
Renna JM, Sondereker KB, Cors CL, Chaszeyka SN, Keenan KN, Corigliano MR, Milgrom LA, Onyak JR, Hamad EJ, Stabio ME. From 2D slices to a 3D model: Training students in digital microanatomy analysis techniques through a 3D printed neuron project. ANATOMICAL SCIENCES EDUCATION 2024; 17:499-505. [PMID: 38379173 DOI: 10.1002/ase.2396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024]
Abstract
The reconstruction of two-dimensional (2D) slices to three-dimensional (3D) digital anatomical models requires technical skills and software that are becoming increasingly important to the modern anatomist, but these skills are rarely taught in undergraduate science classrooms. Furthermore, learning opportunities that allow students to simultaneously explore anatomy in both 2D and 3D space are increasingly valuable. This report describes a novel learning activity that trains students to digitally trace a serially imaged neuron from a confocal stack and to model that neuron in 3D space for 3D printing. By engaging students in the production of a 3D digital model, this learning activity is designed to provide students a novel way to enhance their understanding of the content, including didactic knowledge of neuron morphology, technical research skills in image analysis, and career exploration of neuroanatomy research. Moreover, students engage with microanatomy in a way that starts in 2D but results in a 3D object they can see, touch, and keep. This discursive article presents the learning activity, including videos, instructional guides, and learning objectives designed to engage students on all six levels of Bloom's Taxonomy. Furthermore, this work is a proof of principle modeling workflow that is approachable, inexpensive, achievable, and adaptable to cell types in other organ systems. This work is designed to motivate the expansion of 3D printing technology into microanatomy and neuroanatomy education.
Collapse
Affiliation(s)
- Jordan M Renna
- Department of Biology, University of Akron, Akron, Ohio, USA
| | | | | | | | - Kristin N Keenan
- Department of Biology, University of Akron, Akron, Ohio, USA
- Lake Erie College of Osteopathic Medicine, Erie, Pennsylvania, USA
| | - Michael R Corigliano
- Modern Human Anatomy Program, Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Lindsey A Milgrom
- Modern Human Anatomy Program, Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jessica R Onyak
- Department of Biology, University of Akron, Akron, Ohio, USA
| | - Edward J Hamad
- Department of Biology, University of Akron, Akron, Ohio, USA
| | - Maureen E Stabio
- Modern Human Anatomy Program, Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
16
|
Barrionuevo PA, Sandoval Salinas ML, Fanchini JM. Are ipRGCs involved in human color vision? Hints from physiology, psychophysics, and natural image statistics. Vision Res 2024; 217:108378. [PMID: 38458004 DOI: 10.1016/j.visres.2024.108378] [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] [Received: 11/30/2023] [Revised: 02/09/2024] [Accepted: 02/25/2024] [Indexed: 03/10/2024]
Abstract
Human photoreceptors consist of cones, rods, and melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). First studied in circadian regulation and pupillary control, ipRGCs project to a variety of brain centers suggesting a broader involvement beyond non-visual functions. IpRGC responses are stable, long-lasting, and with a particular codification of photoreceptor signals. In comparison with the transient and adaptive nature of cone and rod signals, ipRGCs' signaling might provide an ecological advantage to different attributes of color vision. Previous studies have indicated melanopsin's influence on visual responses yet its contribution to color perception in humans remains debated. We summarized evidence and hypotheses (from physiology, psychophysics, and natural image statistics) about direct and indirect involvement of ipRGCs in human color vision, by first briefly assessing the current knowledge about the role of melanopsin and ipRGCs in vision and codification of spectral signals. We then approached the question about melanopsin activation eliciting a color percept, discussing studies using the silent substitution method. Finally, we explore various avenues through which ipRGCs might impact color perception indirectly, such as through involvement in peripheral color matching, post-receptoral pathways, color constancy, long-term chromatic adaptation, and chromatic induction. While there is consensus about the role of ipRGCs in brightness perception, confirming its direct contribution to human color perception requires further investigation. We proposed potential approaches for future research, emphasizing the need for empirical validation and methodological thoroughness to elucidate the exact role of ipRGCs in human color vision.
Collapse
Affiliation(s)
- Pablo A Barrionuevo
- Allgemeine Psychologie, Justus-Liebig-Universität Gießen, Germany; Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina.
| | - María L Sandoval Salinas
- Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina; Instituto de Investigaciones de Biodiversidad Argentina (PIDBA), Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán, Argentina
| | - José M Fanchini
- Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina; Departamento de Luminotecnia, Luz y Visión, Facultad de Ciencias Exactas y Tecnología, Universidad Nacional de Tucumán, Argentina
| |
Collapse
|
17
|
Su J, Byer L, Liang Y, Fox MA. Distribution, development, and identity of retinal ganglion cells labeled in the Sert-Cre reporter mouse. J Comp Neurol 2024; 532:e25606. [PMID: 38544361 DOI: 10.1002/cne.25606] [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: 05/25/2023] [Revised: 02/19/2024] [Accepted: 03/07/2024] [Indexed: 05/01/2024]
Abstract
The mouse retina contains over 40 types of retinal ganglion cells (RGCs) that differ in morphology, function, or gene expression. RGCs also differ by whether their axons target the brain.s ipsilateral or contralateral hemisphere. Contralaterally projecting RGCs (contraRGCs) are widespread in mouse retina, whereas ipsilateral projecting RGCs (ipsiRGCs) are confined to the ventro-temporal (VT) crescent of retina. In this study, we employed the Sert-Cre transgenic line, which had been reported to selectively label ipsiRGCs, to study ipsiRGCs during development. Although the number of Cre-expressing ipsiRGCs did not significantly increase with postnatal age, the region of retina that they occupied did, and by adulthood represented ~30% of the retinal surface. Unexpectedly, genetic ablation of Sert-Cre cells failed to fully disrupt ipsilateral projecting retinal axons, suggesting that not all ipsiRGCs generated Cre in Sert-Cre mice. To test this hypothesis, we retrogradely labeled ipsiRGCs in Sert-Cre mice which revealed that not all ipsiRGCs are labeled in Sert-Cre mice and a small population of contraRGCs flanking the VT crescent generates Cre in this line. These results do not negate the usefulness of the Sert-Cre mouse but do raise important caveats to the interpretation of such studies.
Collapse
Affiliation(s)
- Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, USA
| | - Lillian Byer
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- Department of Neuroscience, Davidson College, Davidson, North Carolina, USA
- NeuroSURF Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Yanping Liang
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Michael A Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, USA
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia, USA
- Department of Biology, College of Natural Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
18
|
Maloney R, Quattrochi L, Yoon J, Souza R, Berson D. Efficacy and specificity of melanopsin reporters for retinal ganglion cells. J Comp Neurol 2024; 532:e25591. [PMID: 38375612 PMCID: PMC11000424 DOI: 10.1002/cne.25591] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/03/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are specialized retinal output neurons that mediate behavioral, neuroendocrine, and developmental responses to environmental light. There are diverse molecular strategies for marking ipRGCs, especially in mice, making them among the best characterized retinal ganglion cells (RGCs). With the development of more sensitive reporters, new subtypes of ipRGCs have emerged. We therefore tested high-sensitivity reporter systems to see whether we could reveal yet more. Substantial confusion remains about which of the available methods, if any, label all and only ipRGCs. Here, we compared many different methods for labeling of ipRGCs, including anti-melanopsin immunofluorescence, Opn4-GFP BAC transgenic mice, and Opn4cre mice crossed with three different Cre-specific reporters (Z/EG, Ai9, and Ai14) or injected with Cre-dependent (DIO) AAV2. We show that Opn4cre mice, when crossed with sensitive Cre-reporter mice, label numerous ganglion cell types that lack intrinsic photosensitivity. Though other methods label ipRGCs specifically, they do not label the entire population of ipRGCs. We conclude that no existing method labels all and only ipRGCs. We assess the appropriateness of each reporter for particular applications and integrate findings across reporters to estimate that the overall abundance of ipRGCs among mouse RGCs may approach 11%.
Collapse
Affiliation(s)
- Ryan Maloney
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Lauren Quattrochi
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - James Yoon
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - Rachel Souza
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - David Berson
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
19
|
Li G, Chen L, Jiang Z, Yau KW. Coexistence within one cell of microvillous and ciliary phototransductions across M1- through M6-IpRGCs. Proc Natl Acad Sci U S A 2023; 120:e2315282120. [PMID: 38109525 PMCID: PMC10756192 DOI: 10.1073/pnas.2315282120] [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: 09/02/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) serve as primary photoceptors by expressing the photopigment, melanopsin, and also as retinal relay neurons for rod and cone signals en route to the brain, in both cases for the purpose of non-image vision as well as aspects of image vision. So far, six subtypes of ipRGCs (M1 through M6) have been characterized. Regarding their phototransduction mechanisms, we have previously found that, unconventionally, rhabdomeric (microvillous) and ciliary signaling motifs co-exist within a given M1-, M2-, and M4-ipRGC, with the first mechanism involving PLCβ4 and TRPC6,7 channels and the second involving cAMP and HCN channels. We have now examined M3-, M5-, and M6-cells and found that each cell likewise uses both signaling pathways for phototransduction, despite differences in the percentage representation by each pathway in a given ipRGC subtype for bright-flash responses (and saturated except for M6-cells). Generally, M3- and M5-cells show responses quite similar in kinetics to M2-responses, and M6-cell responses resemble broadly those of M1-cells although much lower in absolute sensitivity and amplitude. Therefore, similar to rod and cone subtypes in image vision, ipRGC subtypes possess the same phototransduction mechanism(s) even though they do not show microvilli or cilia morphologically.
Collapse
Affiliation(s)
- Guang Li
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Lujing Chen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Neuroscience Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Zheng Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
20
|
Berry MH, Leffler J, Allen CN, Sivyer B. Functional subtypes of rodent melanopsin ganglion cells switch roles between night and day illumination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554902. [PMID: 38168436 PMCID: PMC10760181 DOI: 10.1101/2023.08.26.554902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), contain the photopigment melanopsin, and influence both image and non-image forming behaviors. Despite being categorized into multiple types (M1-M6), physiological variability within these types suggests our current understanding of ipRGCs is incomplete. We used multi-electrode array (MEA) recordings and unbiased cluster analysis under synaptic blockade to identify 8 functional clusters of ipRGCs, each with distinct photosensitivity and response timing. We used Cre mice to drive the expression of channelrhodopsin in SON-ipRGCs, enabling the localization of distinct ipRGCs in the dorsal retina. Additionally, we conducted a retrospective unbiased cluster analysis of ipRGC photoresponses to light stimuli across scotopic, mesopic, and photopic intensities, aimed at activating both rod and cone inputs to ipRGCs. Our results revealed shared and distinct synaptic inputs to the identified functional clusters, demonstrating that ipRGCs encode visual information with high fidelity at low light intensities, but poorly at photopic light intensities, when melanopsin activation is highest. Collectively, our findings support a framework with at least 8 functional subtypes of ipRGCs, each encoding luminance with distinct spike outputs, highlighting the inherent functional diversity and complexity of ipRGCs and suggesting a reevaluation of their contributions to retinal function and visual perception under varying light conditions.
Collapse
Affiliation(s)
- Michael H. Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
- Medical Scientist Training program, Oregon Health & Science University, Portland, OR, 97239
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| | - Charles N. Allen
- Oregon Institute of Occupational Health Sciences, Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| |
Collapse
|
21
|
Feord RC, Gomoliszewska A, Pienaar A, Mouland JW, Brown TM. Colour opponency is widespread across the mouse subcortical visual system and differentially targets GABAergic and non-GABAergic neurons. Sci Rep 2023; 13:9313. [PMID: 37291239 PMCID: PMC10250360 DOI: 10.1038/s41598-023-35885-z] [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: 12/16/2022] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Colour vision plays many important roles in animal behaviour but the brain pathways processing colour remain surprisingly poorly understood, including in the most commonly used laboratory mammal, mice. Indeed, particular features of mouse retinal organisation present challenges in defining the mechanisms underlying colour vision in mice and have led to suggestions that this may substantially rely on 'non-classical' rod-cone opponency. By contrast, studies using mice with altered cone spectral sensitivity, to facilitate application of photoreceptor-selective stimuli, have revealed widespread cone-opponency across the subcortical visual system. To determine the extent to which such findings are truly reflective of wildtype mouse colour vision, and facilitate neural circuit mapping of colour-processing pathways using intersectional genetic approaches, we here establish and validate stimuli for selectively manipulating excitation of the native mouse S- and M-cone opsin classes. We then use these to confirm the widespread appearance of cone-opponency (> 25% of neurons) across the mouse visual thalamus and pretectum. We further extend these approaches to map the occurrence of colour-opponency across optogenetically identified GABAergic (GAD2-expressing) cells in key non-image forming visual centres (pretectum and intergeniculate leaflet/ventral lateral geniculate; IGL/vLGN). Strikingly, throughout, we find S-ON/M-OFF opponency is specifically enriched in non-GABAergic cells, with identified GABAergic cells in the IGL/VLGN entirely lacking this property. Collectively, therefore, we establish an important new approach for studying cone function in mice, confirming a surprisingly extensive appearance of cone-opponent processing in the mouse visual system and providing new insight into functional specialisation of the pathways processing such signals.
Collapse
Affiliation(s)
- R C Feord
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Gomoliszewska
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Pienaar
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J W Mouland
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - T M Brown
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
22
|
Linne C, Mon KY, D’Souza S, Jeong H, Jiang X, Brown DM, Zhang K, Vemaraju S, Tsubota K, Kurihara T, Pardue MT, Lang RA. Encephalopsin (OPN3) is required for normal refractive development and the GO/GROW response to induced myopia. Mol Vis 2023; 29:39-57. [PMID: 37287644 PMCID: PMC10243678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Purpose Myopia, or nearsightedness, is the most common form of refractive error and is increasing in prevalence. While significant efforts have been made to identify genetic variants that predispose individuals to myopia, these variants are believed to account for only a small portion of the myopia prevalence, leading to a feedback theory of emmetropization, which depends on the active perception of environmental visual cues. Consequently, there has been renewed interest in studying myopia in the context of light perception, beginning with the opsin family of G-protein coupled receptors (GPCRs). Refractive phenotypes have been characterized in every opsin signaling pathway studied, leaving only Opsin 3 (OPN3), the most widely expressed and blue-light sensing noncanonical opsin, to be investigated for function in the eye and refraction. Methods Opn3 expression was assessed in various ocular tissues using an Opn3eGFP reporter. Weekly refractive development in Opn3 retinal and germline mutants from 3 to 9 weeks of age was measured using an infrared photorefractor and spectral domain optical coherence tomography (SD-OCT). Susceptibility to lens-induced myopia was then assessed using skull-mounted goggles with a -30 diopter experimental and a 0 diopter control lens. Mouse eye biometry was similarly tracked from 3 to 6 weeks. A myopia gene expression signature was assessed 24 h after lens induction for germline mutants to further assess myopia-induced changes. Results Opn3 was found to be expressed in a subset of retinal ganglion cells and a limited number of choroidal cells. Based on an assessment of Opn3 mutants, the OPN3 germline, but not retina conditional Opn3 knockout, exhibits a refractive myopia phenotype, which manifests in decreased lens thickness, shallower aqueous compartment depth, and shorter axial length, atypical of traditional axial myopias. Despite the short axial length, Opn3 null eyes demonstrate normal axial elongation in response to myopia induction and mild changes in choroidal thinning and myopic shift, suggesting that susceptibility to lens-induced myopia is largely unchanged. Additionally, the Opn3 null retinal gene expression signature in response to induced myopia after 24 h is distinct, with opposing Ctgf, Cx43, and Egr1 polarity compared to controls. Conclusions The data suggest that an OPN3 expression domain outside the retina can control lens shape and thus the refractive performance of the eye. Prior to this study, the role of Opn3 in the eye had not been investigated. This work adds OPN3 to the list of opsin family GPCRs that are implicated in emmetropization and myopia. Further, the work to exclude retinal OPN3 as the contributing domain in this refractive phenotype is unique and suggests a distinct mechanism when compared to other opsins.
Collapse
Affiliation(s)
- Courtney Linne
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Khine Yin Mon
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shane D’Souza
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Heonuk Jeong
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Dillon M. Brown
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Kevin Zhang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Shruti Vemaraju
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Machelle T. Pardue
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Richard A. Lang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
23
|
Sawant A, Saha A, Khoussine J, Sinha R, Hoon M. New insights into retinal circuits through EM connectomics: what we have learnt and what remains to be learned. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1168548. [PMID: 38983069 PMCID: PMC11182165 DOI: 10.3389/fopht.2023.1168548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/05/2023] [Indexed: 07/11/2024]
Abstract
The retinal neural circuit is intricately wired for efficient processing of visual signals. This is well-supported by the specialized connections between retinal neurons at both the functional and ultrastructural levels. Through 3D electron microscopic (EM) reconstructions of retinal neurons and circuits we have learnt much about the specificities of connections within the retinal layers including new insights into how retinal neurons establish connections and perform sophisticated visual computations. This mini-review will summarize the retinal circuitry and provide details about the novel insights EM connectomics has brought into our understanding of the retinal circuitry. We will also discuss unresolved questions about the retinal circuitry that can be addressed by EM connectomics in the future.
Collapse
Affiliation(s)
- Abhilash Sawant
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Aindrila Saha
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Jacob Khoussine
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Raunak Sinha
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
24
|
Neitz A, Rice A, Casiraghi L, Bussi IL, Buhr ED, Neitz M, Neitz J, de la Iglesia HO, Kuchenbecker JA. Toward an indoor lighting solution for social jet lag. RESEARCH SQUARE 2023:rs.3.rs-2649098. [PMID: 36993397 PMCID: PMC10055510 DOI: 10.21203/rs.3.rs-2649098/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
There is growing interest in developing artificial lighting that stimulates intrinsically photosensitive retinal ganglion cells (ipRGCs) to entrain circadian rhythms to improve mood, sleep, and health. Efforts have focused on stimulating the intrinsic photopigment, melanopsin; however, recently, specialized color vision circuits have been elucidated in the primate retina that transmit blue-yellow cone-opponent signals to ipRGCs. We designed a light that stimulates color-opponent inputs to ipRGCs by temporally alternating short and longer wavelength components that strongly modulate short-wavelength sensitive (S) cones. Two-hour exposure to this S-cone modulating light produced an average circadian phase advance of one hour and twenty minutes in 6 subjects (mean age = 30 years) compared to no phase advance for the subjects after exposure to a 500-lux white light equated for melanopsin effectiveness. These results are promising for developing artificial lighting that is highly effective in controlling circadian rhythms by invisibly modulating cone-opponent circuits.
Collapse
Affiliation(s)
- Alex Neitz
- Department of Biology and The Molecular and Cellular Biology
graduate program, University of Washington, Seattle, Washington, USA
| | - Alicia Rice
- Department of Biology, University of Washington, Seattle,
Washington, USA
| | - Leandro Casiraghi
- Department of Biology, University of Washington, Seattle,
Washington, USA
| | - Ivana L. Bussi
- Department of Biology, University of Washington, Seattle,
Washington, USA
| | - Ethan D. Buhr
- Department of Ophthalmology, University of Washington, Seattle,
Washington, USA
| | - Maureen Neitz
- Department of Ophthalmology, University of Washington, Seattle,
Washington, USA
| | - Jay Neitz
- Department of Ophthalmology, University of Washington, Seattle,
Washington, USA
| | | | | |
Collapse
|
25
|
Mouland JW, Watson AJ, Martial FP, Lucas RJ, Brown TM. Colour and melanopsin mediated responses in the murine retina. Front Cell Neurosci 2023; 17:1114634. [PMID: 36993934 PMCID: PMC10040579 DOI: 10.3389/fncel.2023.1114634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023] Open
Abstract
Introduction: Intrinsically photosensitive retinal ganglion cells (ipRGCs) integrate melanopsin and rod/cone-mediated inputs to signal to the brain. Whilst originally identified as a cell type specialised for encoding ambient illumination, several lines of evidence indicate a strong association between colour discrimination and ipRGC-driven responses. Thus, cone-mediated colour opponent responses have been widely found across ipRGC target regions in the mouse brain and influence a key ipRGC-dependent function, circadian photoentrainment. Although ipRGCs exhibiting spectrally opponent responses have also been identified, the prevalence of such properties have not been systematically evaluated across the mouse retina or yet been found in ipRGC subtypes known to influence the circadian system. Indeed, there is still uncertainty around the overall prevalence of cone-dependent colour opponency across the mouse retina, given the strong retinal gradient in S and M-cone opsin (co)-expression and overlapping spectral sensitivities of most mouse opsins.Methods: To address this, we use photoreceptor isolating stimuli in multielectrode recordings from human red cone opsin knock-in mouse (Opn1mwR) retinas to systematically survey cone mediated responses and the occurrence of colour opponency across ganglion cell layer (GCL) neurons and identify ipRGCs based on spectral comparisons and/or the persistence of light responses under synaptic blockade.Results: Despite detecting robust cone-mediated responses across the retina, we find cone opponency is rare, especially outside of the central retina (overall ~3% of GCL neurons). In keeping with previous suggestions we also see some evidence of rod-cone opponency (albeit even more rare under our experimental conditions), but find no evidence for any enrichment of cone (or rod) opponent responses among functionally identified ipRGCs.Conclusion: In summary, these data suggest the widespread appearance of cone-opponency across the mouse early visual system and ipRGC-related responses may be an emergent feature of central visual processing mechanisms.
Collapse
Affiliation(s)
- Joshua W. Mouland
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- *Correspondence: Joshua W. Mouland
| | - Alex J. Watson
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Franck P. Martial
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J. Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Timothy M. Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
26
|
Raja S, Milosavljevic N, Allen AE, Cameron MA. Burning the candle at both ends: Intraretinal signaling of intrinsically photosensitive retinal ganglion cells. Front Cell Neurosci 2023; 16:1095787. [PMID: 36687522 PMCID: PMC9853061 DOI: 10.3389/fncel.2022.1095787] [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: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are photoreceptors located in the ganglion cell layer. They project to brain regions involved in predominately non-image-forming functions including entrainment of circadian rhythms, control of the pupil light reflex, and modulation of mood and behavior. In addition to possessing intrinsic photosensitivity via the photopigment melanopsin, these cells receive inputs originating in rods and cones. While most research in the last two decades has focused on the downstream influence of ipRGC signaling, recent studies have shown that ipRGCs also act retrogradely within the retina itself as intraretinal signaling neurons. In this article, we review studies examining intraretinal and, in addition, intraocular signaling pathways of ipRGCs. Through these pathways, ipRGCs regulate inner and outer retinal circuitry through both chemical and electrical synapses, modulate the outputs of ganglion cells (both ipRGCs and non-ipRGCs), and influence arrangement of the correct retinal circuitry and vasculature during development. These data suggest that ipRGC function plays a significant role in the processing of image-forming vision at its earliest stage, positioning these photoreceptors to exert a vital role in perceptual vision. This research will have important implications for lighting design to optimize the best chromatic lighting environments for humans, both in adults and potentially even during fetal and postnatal development. Further studies into these unique ipRGC signaling pathways could also lead to a better understanding of the development of ocular dysfunctions such as myopia.
Collapse
Affiliation(s)
- Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Nina Milosavljevic
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia,*Correspondence: Morven A. Cameron,
| |
Collapse
|
27
|
Unusual phototransduction via cross-motif signaling from G q to adenylyl cyclase in intrinsically photosensitive retinalganglion cells. Proc Natl Acad Sci U S A 2023; 120:e2216599120. [PMID: 36584299 PMCID: PMC9910442 DOI: 10.1073/pnas.2216599120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Nonimage-forming vision in mammals is mediated primarily by melanopsin (OPN4)-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs). In mouse M1-ipRGCs, melanopsin predominantly activates, via Gαq,11,14, phospholipase C-β4 to open transient receptor 6 (TRPC6) and TRPC7 channels. In M2- and M4-ipRGCs, however, a prominent phototransduction mechanism involves the opening of hyperpolarization- and cyclic nucleotide-gated channels via cyclic nucleotide, although the upstream steps remain uncertain. We report here experiments, primarily on M4-ipRGCs, with photo-uncaging of cyclic nucleotides and virally expressed CNGA2 channels to conclude that the second messenger is cyclic adenosine monophosphate (cAMP) - very surprising considering that cyclic guanosine monophosphate (cGMP) is used in almost all cyclic nucleotide-mediated phototransduction mechanisms across the animal kingdom. We further found that the upstream G protein is likewise Gq, which via its Gβγ subunits directly activates adenylyl cyclase (AC). Our findings are a demonstration in a native cell of a cross-motif GPCR signaling pathway from Gq directly to AC with a specific function.
Collapse
|
28
|
Procyk CA, Rodgers J, Zindy E, Lucas RJ, Milosavljevic N. Quantitative characterisation of ipRGCs in retinal degeneration using a computation platform for extracting and reconstructing single neurons in 3D from a multi-colour labeled population. Front Cell Neurosci 2022; 16:1009321. [PMID: 36385954 PMCID: PMC9664085 DOI: 10.3389/fncel.2022.1009321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Light has a profound impact on mammalian physiology and behavior. Intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin, rendering them sensitive to light, and are involved in both image-forming vision and non-image forming responses to light such as circadian photo-entrainment and the pupillary light reflex. Following outer photoreceptor degeneration, the death of rod and cone photoreceptors results in global re-modeling of the remnant neural retina. Although ipRGCs can continue signaling light information to the brain even in advanced stages of degeneration, it is unknown if all six morphologically distinct subtypes survive, or how their dendritic architecture may be affected. To answer these questions, we generated a computational platform-BRIAN (Brainbow Analysis of individual Neurons) to analyze Brainbow labeled tissues by allowing objective identification of voxels clusters in Principal Component Space, and their subsequent extraction to produce 3D images of single neurons suitable for analysis with existing tracing technology. We show that BRIAN can efficiently recreate single neurons or individual axonal projections from densely labeled tissue with sufficient anatomical resolution for subtype quantitative classification. We apply this tool to generate quantitative morphological information about ipRGCs in the degenerate retina including soma size, dendritic field size, dendritic complexity, and stratification. Using this information, we were able to identify cells whose characteristics match those reported for all six defined subtypes of ipRGC in the wildtype mouse retina (M1-M6), including the rare and complex M3 and M6 subtypes. This indicates that ipRGCs survive outer retinal degeneration with broadly normal morphology. We additionally describe one cell in the degenerate retina which matches the description of the Gigantic M1 cell in Humans which has not been previously identified in rodent.
Collapse
Affiliation(s)
- Christopher A. Procyk
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Jessica Rodgers
- Faculty of Biology Medicine and Health, Centre for Biological Timing and Division of Neuroscience, University of Manchester, Manchester, United Kingdom
| | - Egor Zindy
- Centre for Microscopy and Molecular Imaging, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert J. Lucas
- Faculty of Biology Medicine and Health, Centre for Biological Timing and Division of Neuroscience, University of Manchester, Manchester, United Kingdom
| | - Nina Milosavljevic
- Faculty of Biology Medicine and Health, Centre for Biological Timing and Division of Neuroscience, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Gao J, Provencio I, Liu X. Intrinsically photosensitive retinal ganglion cells in glaucoma. Front Cell Neurosci 2022; 16:992747. [PMID: 36212698 PMCID: PMC9537624 DOI: 10.3389/fncel.2022.992747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Glaucoma is a group of eye diseases afflicting more than 70 million people worldwide. It is characterized by damage to retinal ganglion cells (RGCs) that ultimately leads to the death of the cells and vision loss. The diversity of RGC types has been appreciated for decades, and studies, including ours, have shown that RGCs degenerate and die in a type-specific manner in rodent models of glaucoma. The type-specific loss of RGCs results in differential damage to visual and non-visual functions. One type of RGC, the intrinsically photosensitive retinal ganglion cell (ipRGC), expressing the photopigment melanopsin, serves a broad array of non-visual responses to light. Since its discovery, six subtypes of ipRGC have been described, each contributing to various image-forming and non-image-forming functions such as circadian photoentrainment, the pupillary light reflex, the photic control of mood and sleep, and visual contrast sensitivity. We recently demonstrated a link between type-specific ipRGC survival and behavioral deficits in a mouse model of chronic ocular hypertension. This review focuses on the type-specific ipRGC degeneration and associated behavioral changes in animal models and glaucoma patients. A better understanding of how glaucomatous insult impacts the ipRGC-based circuits will have broad impacts on improving the treatment of glaucoma-associated non-visual disorders.
Collapse
Affiliation(s)
- Jingyi Gao
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | - Ignacio Provencio
- Department of Biology, University of Virginia, Charlottesville, VA, United States
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, United States
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, VA, United States
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, United States
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, United States
- Department of Psychology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
30
|
Melanopsin retinal ganglion cells mediate light-promoted brain development. Cell 2022; 185:3124-3137.e15. [PMID: 35944541 DOI: 10.1016/j.cell.2022.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 05/10/2022] [Accepted: 07/15/2022] [Indexed: 02/05/2023]
Abstract
During development, melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) become light sensitive much earlier than rods and cones. IpRGCs project to many subcortical areas, whereas physiological functions of these projections are yet to be fully elucidated. Here, we found that ipRGC-mediated light sensation promotes synaptogenesis of pyramidal neurons in various cortices and the hippocampus. This phenomenon depends on activation of ipRGCs and is mediated by the release of oxytocin from the supraoptic nucleus (SON) and the paraventricular nucleus (PVN) into cerebral-spinal fluid. We further characterized a direct connection between ipRGCs and oxytocin neurons in the SON and mutual projections between oxytocin neurons in the SON and PVN. Moreover, we showed that the lack of ipRGC-mediated, light-promoted early cortical synaptogenesis compromised learning ability in adult mice. Our results highlight the importance of light sensation early in life on the development of learning ability and therefore call attention to suitable light environment for infant care.
Collapse
|
31
|
Gao J, Griner EM, Liu M, Moy J, Provencio I, Liu X. Differential effects of experimental glaucoma on intrinsically photosensitive retinal ganglion cells in mice. J Comp Neurol 2022; 530:1494-1506. [PMID: 34958682 PMCID: PMC9010357 DOI: 10.1002/cne.25293] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/08/2022]
Abstract
Glaucoma is a group of eye diseases characterized by retinal ganglion cell (RGC) loss and optic nerve damage. Studies, including this study, support that RGCs degenerate and die in a type-specific manner following the disease insult. Here we specifically examined one RGC type, the intrinsically photosensitive retinal ganglion cell (ipRGC), and its associated functional deficits in a mouse model of experimental glaucoma. We induced chronic ocular hypertension (OHT) by laser photocoagulation and then characterized the survival of ipRGC subtypes. We found that ipRGCs suffer significant loss, similar to the general RGC population, but ipRGC subtypes are differentially affected following chronic OHT. M4 ipRGCs, which are involved in pattern vision, are susceptible to chronic OHT. Correspondingly, mice with chronic OHT experience reduced contrast sensitivity and visual acuity. By comparison, M1 ipRGCs, which project to the suprachiasmatic nuclei to regulate circadian rhythmicity, exhibit almost no cell loss following chronic OHT. Accordingly, we observed that circadian re-entrainment and circadian rhythmicity are largely not disrupted in OHT mice. Our study demonstrates the link between subtype-specific ipRGC survival and behavioral deficits in glaucomatous mice. These findings provide insight into glaucoma-induced visual behavioral deficits and their underlying mechanisms.
Collapse
Affiliation(s)
- Jingyi Gao
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Erin M. Griner
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Mingna Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Joanna Moy
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Ignacio Provencio
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, USA
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
32
|
Tools and Biomarkers for the Study of Retinal Ganglion Cell Degeneration. Int J Mol Sci 2022; 23:ijms23084287. [PMID: 35457104 PMCID: PMC9025234 DOI: 10.3390/ijms23084287] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The retina is part of the central nervous system, its analysis may provide an idea of the health and functionality, not only of the retina, but also of the entire central nervous system, as has been shown in Alzheimer’s or Parkinson’s diseases. Within the retina, the ganglion cells (RGC) are the neurons in charge of processing and sending light information to higher brain centers. Diverse insults and pathological states cause degeneration of RGC, leading to irreversible blindness or impaired vision. RGCs are the measurable endpoints in current research into experimental therapies and diagnosis in multiple ocular pathologies, like glaucoma. RGC subtype classifications are based on morphological, functional, genetical, and immunohistochemical aspects. Although great efforts are being made, there is still no classification accepted by consensus. Moreover, it has been observed that each RGC subtype has a different susceptibility to injury. Characterizing these subtypes together with cell death pathway identification will help to understand the degenerative process in the different injury and pathological models, and therefore prevent it. Here we review the known RGC subtypes, as well as the diagnostic techniques, probes, and biomarkers for programmed and unprogrammed cell death in RGC.
Collapse
|
33
|
Abstract
Retinal circuits transform the pixel representation of photoreceptors into the feature representations of ganglion cells, whose axons transmit these representations to the brain. Functional, morphological, and transcriptomic surveys have identified more than 40 retinal ganglion cell (RGC) types in mice. RGCs extract features of varying complexity; some simply signal local differences in brightness (i.e., luminance contrast), whereas others detect specific motion trajectories. To understand the retina, we need to know how retinal circuits give rise to the diverse RGC feature representations. A catalog of the RGC feature set, in turn, is fundamental to understanding visual processing in the brain. Anterograde tracing indicates that RGCs innervate more than 50 areas in the mouse brain. Current maps connecting RGC types to brain areas are rudimentary, as is our understanding of how retinal signals are transformed downstream to guide behavior. In this article, I review the feature selectivities of mouse RGCs, how they arise, and how they are utilized downstream. Not only is knowledge of the behavioral purpose of RGC signals critical for understanding the retinal contributions to vision; it can also guide us to the most relevant areas of visual feature space. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences; Department of Neuroscience; Department of Biomedical Engineering; and Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA;
| |
Collapse
|
34
|
Abed S, Reilly A, Arnold SJ, Feldheim DA. Adult Expression of Tbr2 Is Required for the Maintenance but Not Survival of Intrinsically Photosensitive Retinal Ganglion Cells. Front Cell Neurosci 2022; 16:826590. [PMID: 35401124 PMCID: PMC8983909 DOI: 10.3389/fncel.2022.826590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells expressing the photopigment melanopsin are intrinsically photosensitive (ipRGCs). ipRGCs regulate subconscious non-image-forming behaviors such as circadian rhythms, pupil dilation, and light-mediated mood. Previously, we and others showed that the transcription factor Tbr2 (EOMES) is required during retinal development for the formation of ipRGCs. Tbr2 is also expressed in the adult retina leading to the hypothesis that it plays a role in adult ipRGC function. To test this, we removed Tbr2 in adult mice. We found that this results in the loss of melanopsin expression in ipRGCs but does not lead to cell death or morphological changes to their dendritic or axonal termination patterns. Additionally, we found ectopic expression of Tbr2 in conventional RGCs does not induce melanopsin expression but can increase melanopsin expression in existing ipRGCs. An interesting feature of ipRGCs is their superior survival relative to conventional RGCs after an optic nerve injury. We find that loss of Tbr2 decreases the survival rate of ipRGCs after optic nerve damage suggesting that Tbr2 plays a role in ipRGC survival after injury. Lastly, we show that the GABAergic amacrine cell marker Meis2, is expressed in the majority of Tbr2-expressing displaced amacrine cells as well as in a subset of Tbr2-expressing RGCs. These findings demonstrate that Tbr2 is necessary but not sufficient for melanopsin expression, that Tbr2 is involved in ipRGC survival after optic nerve injury, and identify a marker for Tbr2-expressing displaced amacrine cells.
Collapse
Affiliation(s)
- Sadaf Abed
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Andreea Reilly
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Sebastian J. Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - David A. Feldheim
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
- *Correspondence: David A. Feldheim,
| |
Collapse
|
35
|
Sheng Y, Chen L, Ren X, Jiang Z, Yau KW. Molecular determinants of response kinetics of mouse M1 intrinsically-photosensitive retinal ganglion cells. Sci Rep 2021; 11:23424. [PMID: 34873237 PMCID: PMC8648817 DOI: 10.1038/s41598-021-02832-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022] Open
Abstract
Intrinsically-photosensitive retinal ganglion cells (ipRGCs) are non-rod/non-cone retinal photoreceptors expressing the visual pigment, melanopsin, to detect ambient irradiance for various non-image-forming visual functions. The M1-subtype, amongst the best studied, mediates primarily circadian photoentrainment and pupillary light reflex. Their intrinsic light responses are more prolonged than those of rods and cones even at the single-photon level, in accordance with the typically slower time course of non-image-forming vision. The short (OPN4S) and long (OPN4L) alternatively-spliced forms of melanopsin proteins are both present in M1-ipRGCs, but their functional difference is unclear. We have examined this point by genetically removing the Opn4 gene (Opn4-/-) in mouse and re-expressing either OPN4S or OPN4L singly in Opn4-/- mice by using adeno-associated virus, but found no obvious difference in their intrinsic dim-flash responses. Previous studies have indicated that two dominant slow steps in M1-ipRGC phototransduction dictate these cells' intrinsic dim-flash-response kinetics, with time constants (τ1 and τ2) at room temperature of ~ 2 s and ~ 20 s, respectively. Here we found that melanopsin inactivation by phosphorylation or by β-arrestins may not be one of these two steps, because their genetic disruptions did not prolong the two time constants or affect the response waveform. Disruption of GAP (GTPase-Activating-Protein) activity on the effector enzyme, PLCβ4, in M1-ipRGC phototransduction to slow down G-protein deactivation also did not prolong the response decay, but caused its rising phase to become slightly sigmoidal by giving rise to a third time constant, τ3, of ~ 2 s (room temperature). This last observation suggests that GAP-mediated G-protein deactivation does partake in the flash-response termination, although normally with a time constant too short to be visible in the response waveform.
Collapse
Affiliation(s)
- Yanghui Sheng
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Graduate Neuroscience Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lujing Chen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Graduate Neuroscience Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Xiaozhi Ren
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Vedere Bio II, Inc., 700 Main St, Cambridge, MA, 02139, USA
| | - Zheng Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, Houston, TX, 77030, USA
| | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA.
| |
Collapse
|
36
|
Contreras E, Nobleman AP, Robinson PR, Schmidt TM. Melanopsin phototransduction: beyond canonical cascades. J Exp Biol 2021; 224:273562. [PMID: 34842918 PMCID: PMC8714064 DOI: 10.1242/jeb.226522] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Melanopsin is a visual pigment that is expressed in a small subset of intrinsically photosensitive retinal ganglion cells (ipRGCs). It is involved in regulating non-image forming visual behaviors, such as circadian photoentrainment and the pupillary light reflex, while also playing a role in many aspects of image-forming vision, such as contrast sensitivity. Melanopsin was initially discovered in the melanophores of the skin of the frog Xenopus, and subsequently found in a subset of ganglion cells in rat, mouse and primate retinas. ipRGCs were initially thought to be a single retinal ganglion cell population, and melanopsin was thought to activate a single, invertebrate-like Gq/transient receptor potential canonical (TRPC)-based phototransduction cascade within these cells. However, in the 20 years since the discovery of melanopsin, our knowledge of this visual pigment and ipRGCs has expanded dramatically. Six ipRGC subtypes have now been identified in the mouse, each with unique morphological, physiological and functional properties. Multiple subtypes have also been identified in other species, suggesting that this cell type diversity is a general feature of the ipRGC system. This diversity has led to a renewed interest in melanopsin phototransduction that may not follow the canonical Gq/TRPC cascade in the mouse or in the plethora of other organisms that express the melanopsin photopigment. In this Review, we discuss recent findings and discoveries that have challenged the prevailing view of melanopsin phototransduction as a single pathway that influences solely non-image forming functions.
Collapse
Affiliation(s)
- Ely Contreras
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
| | - Alexis P. Nobleman
- University of Maryland Baltimore County, Department of Biological Sciences, Baltimore, MD 21250, USA,Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Phyllis R. Robinson
- University of Maryland Baltimore County, Department of Biological Sciences, Baltimore, MD 21250, USA,Authors for correspondence (; )
| | - Tiffany M. Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA,Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL 60611, USA,Authors for correspondence (; )
| |
Collapse
|
37
|
Muzyka VV, Badea TC. Genetic interplay between transcription factor Pou4f1/Brn3a and neurotrophin receptor Ret in retinal ganglion cell type specification. Neural Dev 2021; 16:5. [PMID: 34548095 PMCID: PMC8454062 DOI: 10.1186/s13064-021-00155-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background While the transcriptional code governing retinal ganglion cell (RGC) type specification begins to be understood, its interplay with neurotrophic signaling is largely unexplored. In mice, the transcription factor Brn3a/Pou4f1 is expressed in most RGCs, and is required for the specification of RGCs with small dendritic arbors. The Glial Derived Neurotrophic Factor (GDNF) receptor Ret is expressed in a subset of RGCs, including some expressing Brn3a, but its role in RGC development is not defined. Methods Here we use combinatorial genetic experiments using conditional knock-in reporter alleles at the Brn3a and Ret loci, in combination with retina- or Ret specific Cre drivers, to generate complete or mosaic genetic ablations of either Brn3a or Ret in RGCs. We then use sparse labelling to investigate Brn3a and Ret gene dosage effects on RGC dendritic arbor morphology. In addition, we use immunostaining and/or gene expression profiling by RNASeq to identify transcriptional targets relevant for the potential Brn3a-Ret interaction in RGC development. Results We find that mosaic gene dosage manipulation of the transcription factor Brn3a/Pou4f1 in neurotrophic receptor Ret heterozygote RGCs results in altered cell fate decisions and/or morphological dendritic defects. Specific RGC types are lost if Brn3a is ablated during embryogenesis and only mildly affected by postnatal Brn3a ablation. Sparse but not complete Brn3a heterozygosity combined with complete Ret heterozygosity has striking effects on RGC type distribution. Brn3a only mildly modulates Ret transcription, while Ret knockouts exhibit slightly skewed Brn3a and Brn3b expression during development that is corrected by adult age. Brn3a loss of function modestly but significantly affects distribution of Ret co-receptors GFRα1-3, and neurotrophin receptors TrkA and TrkC in RGCs. Conclusions Based on these observations, we propose that Brn3a and Ret converge onto developmental pathways that control RGC type specification, potentially through a competitive mechanism requiring signaling from the surrounding tissue. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-021-00155-z.
Collapse
Affiliation(s)
- Vladimir Vladimirovich Muzyka
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, USA. .,Institute of Cytology and Genetics, Novosibirsk State University, Novosibirsk, Russia.
| | - Tudor Constantin Badea
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, USA. .,Research and Development Institute, School of Medicine, Transilvania University of Brasov, Brasov, Romania.
| |
Collapse
|
38
|
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only anterogradely to drive behavioral responses, but also retrogradely to some amacrine interneurons to modulate retinal physiology. We previously found that all displaced amacrine cells with spiking, tonic excitatory photoresponses receive gap-junction input from ipRGCs, but the connectivity patterns and functional roles of ipRGC-amacrine coupling remained largely unknown. Here, we injected PoPro1 fluorescent tracer into all six types of mouse ipRGCs to identify coupled amacrine cells, and analyzed the latter's morphological and electrophysiological properties. We also examined how genetically disrupting ipRGC-amacrine coupling affected ipRGC photoresponses. Results showed that ipRGCs couple with not just ON- and ON/OFF-stratified amacrine cells in the ganglion-cell layer as previously reported, but also OFF-stratified amacrine cells in both ganglion-cell and inner nuclear layers. M1- and M3-type ipRGCs couple mainly with ON/OFF-stratified amacrine cells, whereas the other ipRGC types couple almost exclusively with ON-stratified ones. ipRGCs transmit melanopsin-based light responses to at least 93% of the coupled amacrine cells. Some of the ON-stratifying ipRGC-coupled amacrine cells exhibit transient hyperpolarizing light responses. We detected bidirectional electrical transmission between an ipRGC and a coupled amacrine cell, although transmission was asymmetric for this particular cell pair, favoring the ipRGC-to-amacrine direction. We also observed electrical transmission between two amacrine cells coupled to the same ipRGC. In both scenarios of coupling, the coupled cells often spiked synchronously. While ipRGC-amacrine coupling somewhat reduces the peak firing rates of ipRGCs' intrinsic melanopsin-based photoresponses, it renders these responses more sustained and longer-lasting. In summary, ipRGCs' gap junctional network involves more amacrine cell types and plays more roles than previously appreciated.
Collapse
|
39
|
Mouland JW, Pienaar A, Williams C, Watson AJ, Lucas RJ, Brown TM. Extensive cone-dependent spectral opponency within a discrete zone of the lateral geniculate nucleus supporting mouse color vision. Curr Biol 2021; 31:3391-3400.e4. [PMID: 34111401 PMCID: PMC8360768 DOI: 10.1016/j.cub.2021.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/23/2021] [Accepted: 05/13/2021] [Indexed: 12/15/2022]
Abstract
Color vision, originating with opponent processing of spectrally distinct photoreceptor signals, plays important roles in animal behavior.1-4 Surprisingly, however, comparatively little is understood about color processing in the brain, including in widely used laboratory mammals such as mice. The retinal gradient in S- and M-cone opsin (co-)expression has traditionally been considered an impediment to mouse color vision.5-8 However, recent data indicate that mice exhibit robust chromatic discrimination within the central-upper visual field.9 Retinal color opponency has been reported to emerge from superimposing inhibitory surround receptive fields on the cone opsin expression gradient, and by introducing opponent rod signals in retinal regions with sparse M-cone opsin expression.10-13 The relative importance of these proposed mechanisms in determining the properties of neurons at higher visual processing stages remains unknown. We address these questions using multielectrode recordings from the lateral geniculate nucleus (LGN) in mice with altered M-cone spectral sensitivity (Opn1mwR) and multispectral stimuli that allow selective modulation of signaling by individual opsin classes. Remarkably, we find many (∼25%) LGN cells are color opponent, that such cells are localized to a distinct medial LGN zone and that their properties cannot simply be explained by the proposed retinal opponent mechanisms. Opponent responses in LGN can be driven solely by cones, independent of cone-opsin expression gradients and rod input, with many cells exhibiting spatially congruent antagonistic receptive fields. Our data therefore suggest previously unidentified mechanisms may support extensive and sophisticated color processing in the mouse LGN.
Collapse
Affiliation(s)
- Josh W Mouland
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Abigail Pienaar
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Christopher Williams
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Alex J Watson
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Robert J Lucas
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Timothy M Brown
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
40
|
Hannibal J. Comparative Neurology of Circadian Photoreception: The Retinohypothalamic Tract (RHT) in Sighted and Naturally Blind Mammals. Front Neurosci 2021; 15:640113. [PMID: 34054403 PMCID: PMC8160255 DOI: 10.3389/fnins.2021.640113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
The mammalian eye contains two systems for light perception: an image detecting system constituted primarily of the classical photoreceptors, rods and cones, and a non-image forming system (NIF) constituted of a small group of intrinsically photosensitive retinal ganglion cells driven by melanopsin (mRGCs). The mRGCs receive input from the outer retina and NIF mediates light entrainment of circadian rhythms, masking behavior, light induced inhibition of nocturnal melatonin secretion, pupillary reflex (PLR), and affect the sleep/wake cycle. This review focuses on the mammalian NIF and its anatomy in the eye as well as its neuronal projection to the brain. This pathway is known as the retinohypothalamic tract (RHT). The development and functions of the NIF as well as the knowledge gained from studying gene modified mice is highlighted. Furthermore, the similarities of the NIF between sighted (nocturnal and diurnal rodent species, monkeys, humans) and naturally blind mammals (blind mole rats Spalax ehrenbergi and the Iberian mole, Talpa occidentalis) are discussed in relation to a changing world where increasing exposure to artificial light at night (ALAN) is becoming a challenge for humans and animals in the modern society.
Collapse
Affiliation(s)
- Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Patterson SS, Neitz M, Neitz J. S-cone circuits in the primate retina for non-image-forming vision. Semin Cell Dev Biol 2021; 126:66-70. [PMID: 33994300 DOI: 10.1016/j.semcdb.2021.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 11/16/2022]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) respond directly to light by virtue of containing melanopsin which peaks at about 483 nm. However, in primates, ipRGCs also receive color opponent inputs from short-wavelength-sensitive (S) cone circuits that are well-suited to encode circadian changes in the color of the sky that accompany the rising and setting sun. Here, we review the retinal circuits that endow primate ipRGCs with the cone-opponency capable of encoding the color of the sky and contributing to the wide-ranging effects of short-wavelength light on ipRGC-mediated non-image-forming visual function in humans.
Collapse
Affiliation(s)
- Sara S Patterson
- Center for Visual Science, University of Rochester, Rochester, NY 14607, USA
| | - Maureen Neitz
- Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA
| | - Jay Neitz
- Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
42
|
Khani MH, Gollisch T. Linear and nonlinear chromatic integration in the mouse retina. Nat Commun 2021; 12:1900. [PMID: 33772000 PMCID: PMC7997992 DOI: 10.1038/s41467-021-22042-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/23/2021] [Indexed: 11/09/2022] Open
Abstract
The computations performed by a neural circuit depend on how it integrates its input signals into an output of its own. In the retina, ganglion cells integrate visual information over time, space, and chromatic channels. Unlike the former two, chromatic integration is largely unexplored. Analogous to classical studies of spatial integration, we here study chromatic integration in mouse retina by identifying chromatic stimuli for which activation from the green or UV color channel is maximally balanced by deactivation through the other color channel. This reveals nonlinear chromatic integration in subsets of On, Off, and On-Off ganglion cells. Unlike the latter two, nonlinear On cells display response suppression rather than activation under balanced chromatic stimulation. Furthermore, nonlinear chromatic integration occurs independently of nonlinear spatial integration, depends on contributions from the rod pathway and on surround inhibition, and may provide information about chromatic boundaries, such as the skyline in natural scenes.
Collapse
Affiliation(s)
- Mohammad Hossein Khani
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany.
- Bernstein Center for Computational Neuroscience, Göttingen, Germany.
- International Max Planck Research School for Neuroscience, Göttingen, Germany.
| | - Tim Gollisch
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany.
- Bernstein Center for Computational Neuroscience, Göttingen, Germany.
| |
Collapse
|
43
|
Pottackal J, Walsh HL, Rahmani P, Zhang K, Justice NJ, Demb JB. Photoreceptive Ganglion Cells Drive Circuits for Local Inhibition in the Mouse Retina. J Neurosci 2021; 41:1489-1504. [PMID: 33397711 PMCID: PMC7896016 DOI: 10.1523/jneurosci.0674-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 11/11/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) exhibit melanopsin-dependent light responses that persist in the absence of rod and cone photoreceptor-mediated input. In addition to signaling anterogradely to the brain, ipRGCs signal retrogradely to intraretinal circuitry via gap junction-mediated electrical synapses with amacrine cells (ACs). However, the targets and functions of these intraretinal signals remain largely unknown. Here, in mice of both sexes, we identify circuitry that enables M5 ipRGCs to locally inhibit retinal neurons via electrical synapses with a nonspiking GABAergic AC. During pharmacological blockade of rod- and cone-mediated input, whole-cell recordings of corticotropin-releasing hormone-expressing (CRH+) ACs reveal persistent visual responses that require both melanopsin expression and gap junctions. In the developing retina, ipRGC-mediated input to CRH+ ACs is weak or absent before eye opening, indicating a primary role for this input in the mature retina (i.e., in parallel with rod- and cone-mediated input). Among several ipRGC types, only M5 ipRGCs exhibit consistent anatomical and physiological coupling to CRH+ ACs. Optogenetic stimulation of local CRH+ ACs directly drives IPSCs in M4 and M5, but not M1-M3, ipRGCs. CRH+ ACs also inhibit M2 ipRGC-coupled spiking ACs, demonstrating direct interaction between discrete networks of ipRGC-coupled interneurons. Together, these results demonstrate a functional role for electrical synapses in translating ipRGC activity into feedforward and feedback inhibition of local retinal circuits.SIGNIFICANCE STATEMENT Melanopsin directly generates light responses in intrinsically photosensitive retinal ganglion cells (ipRGCs). Through gap junction-mediated electrical synapses with retinal interneurons, these uniquely photoreceptive RGCs may also influence the activity and output of neuronal circuits within the retina. Here, we identified and studied an electrical synaptic circuit that, in principle, could couple ipRGC activity to the chemical output of an identified retinal interneuron. Specifically, we found that M5 ipRGCs form electrical synapses with corticotropin-releasing hormone-expressing amacrine cells, which locally release GABA to inhibit specific RGC types. Thus, ipRGCs are poised to influence the output of diverse retinal circuits via electrical synapses with interneurons.
Collapse
Affiliation(s)
| | | | | | | | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030
| | - Jonathan B Demb
- Interdepartmental Neuroscience Program
- Department of Ophthalmology and Visual Science
- Department of Cellular and Molecular Physiology
- Department of Neuroscience, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
44
|
Orexin-A Intensifies Mouse Pupillary Light Response by Modulating Intrinsically Photosensitive Retinal Ganglion Cells. J Neurosci 2021; 41:2566-2580. [PMID: 33536197 DOI: 10.1523/jneurosci.0217-20.2021] [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: 01/26/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/24/2022] Open
Abstract
We show for the first time that the neuropeptide orexin modulates pupillary light response, a non-image-forming visual function, in mice of either sex. Intravitreal injection of the orexin receptor (OXR) antagonist TCS1102 and orexin-A reduced and enhanced pupillary constriction in response to light, respectively. Orexin-A activated OX1Rs on M2-type intrinsically photosensitive retinal ganglion cells (M2 cells), and caused membrane depolarization of these cells by modulating inward rectifier potassium channels and nonselective cation channels, thus resulting in an increase in intrinsic excitability. The increased intrinsic excitability could account for the orexin-A-evoked increase in spontaneous discharges and light-induced spiking rates of M2 cells, leading to an intensification of pupillary constriction. Orexin-A did not alter the light response of M1 cells, which could be because of no or weak expression of OX1Rs on them, as revealed by RNAscope in situ hybridization. In sum, orexin-A is likely to decrease the pupil size of mice by influencing M2 cells, thereby improving visual performance in awake mice via enhancing the focal depth of the eye's refractive system.SIGNIFICANCE STATEMENT This study reveals the role of the neuropeptide orexin in mouse pupillary light response, a non-image-forming visual function. Intravitreal orexin-A administration intensifies light-induced pupillary constriction via increasing the excitability of M2 intrinsically photosensitive retinal ganglion cells by activating the orexin receptor subtype OX1R. Modulation of inward rectifier potassium channels and nonselective cation channels were both involved in the ionic mechanisms underlying such intensification. Orexin could improve visual performance in awake mice by reducing the pupil size and thereby enhancing the focal depth of the eye's refractive system.
Collapse
|
45
|
Vit JP, Fuchs DT, Angel A, Levy A, Lamensdorf I, Black KL, Koronyo Y, Koronyo-Hamaoui M. Color and contrast vision in mouse models of aging and Alzheimer's disease using a novel visual-stimuli four-arm maze. Sci Rep 2021; 11:1255. [PMID: 33441984 PMCID: PMC7806734 DOI: 10.1038/s41598-021-80988-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
We introduce a novel visual-stimuli four-arm maze (ViS4M) equipped with spectrally- and intensity-controlled LED emitters and dynamic grayscale objects that relies on innate exploratory behavior to assess color and contrast vision in mice. Its application to detect visual impairments during normal aging and over the course of Alzheimer’s disease (AD) is evaluated in wild-type (WT) and transgenic APPSWE/PS1∆E9 murine models of AD (AD+) across an array of irradiance, chromaticity, and contrast conditions. Substantial color and contrast-mode alternation deficits appear in AD+ mice at an age when hippocampal-based memory and learning is still intact. Profiling of timespan, entries and transition patterns between the different arms uncovers variable AD-associated impairments in contrast sensitivity and color discrimination, reminiscent of tritanomalous defects documented in AD patients. Transition deficits are found in aged WT mice in the absence of alternation decline. Overall, ViS4M is a versatile, controlled device to measure color and contrast-related vision in aged and diseased mice.
Collapse
Affiliation(s)
- Jean-Philippe Vit
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.,Biobehavioral Research Core, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ariel Angel
- Pharmaseed Ltd., 9 Hamazmera St., 74047, Ness Ziona, Israel
| | - Aharon Levy
- Pharmaseed Ltd., 9 Hamazmera St., 74047, Ness Ziona, Israel
| | | | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA. .,Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Orlowska-Feuer P, Smyk MK, Alwani A, Lewandowski MH. Neuronal Responses to Short Wavelength Light Deficiency in the Rat Subcortical Visual System. Front Neurosci 2021; 14:615181. [PMID: 33488355 PMCID: PMC7815651 DOI: 10.3389/fnins.2020.615181] [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: 10/08/2020] [Accepted: 12/08/2020] [Indexed: 12/26/2022] Open
Abstract
The amount and spectral composition of light changes considerably during the day, with dawn and dusk being the most crucial moments when light is within the mesopic range and short wavelength enriched. It was recently shown that animals use both cues to adjust their internal circadian clock, thereby their behavior and physiology, with the solar cycle. The role of blue light in circadian processes and neuronal responses is well established, however, an unanswered question remains: how do changes in the spectral composition of light (short wavelengths blocking) influence neuronal activity? In this study we addressed this question by performing electrophysiological recordings in image (dorsal lateral geniculate nucleus; dLGN) and non-image (the olivary pretectal nucleus; OPN, the suprachiasmatic nucleus; SCN) visual structures to determine neuronal responses to spectrally varied light stimuli. We found that removing short-wavelength from the polychromatic light (cut off at 525 nm) attenuates the most transient ON and sustained cells in the dLGN and OPN, respectively. Moreover, we compared the ability of different types of sustained OPN neurons (either changing or not their response profile to filtered polychromatic light) to irradiance coding, and show that both groups achieve it with equal efficacy. On the other hand, even very dim monochromatic UV light (360 nm; log 9.95 photons/cm2/s) evokes neuronal responses in the dLGN and SCN. To our knowledge, this is the first electrophysiological experiment supporting previous behavioral findings showing visual and circadian functions disruptions under short wavelength blocking environment. The current results confirm that neuronal activity in response to polychromatic light in retinorecipient structures is affected by removing short wavelengths, however, with type and structure – specific action. Moreover, they show that rats are sensitive to even very dim UV light.
Collapse
Affiliation(s)
- Patrycja Orlowska-Feuer
- Malopolska Centre of Biotechnology (MCB), Jagiellonian University in Kraków, Kraków, Poland.,Department of Neurophysiology and Chronobiology, Jagiellonian University in Kraków, Kraków, Poland
| | - Magdalena Kinga Smyk
- Malopolska Centre of Biotechnology (MCB), Jagiellonian University in Kraków, Kraków, Poland.,Department of Neurophysiology and Chronobiology, Jagiellonian University in Kraków, Kraków, Poland
| | - Anna Alwani
- Department of Neurophysiology and Chronobiology, Jagiellonian University in Kraków, Kraków, Poland
| | | |
Collapse
|
47
|
Photosensitive ganglion cells: A diminutive, yet essential population. ARCHIVOS DE LA SOCIEDAD ESPAÑOLA DE OFTALMOLOGÍA 2020; 96:299-315. [PMID: 34092284 DOI: 10.1016/j.oftale.2020.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022]
Abstract
Our visual system has evolved to provide us with an image of the scene that surrounds us, informing us of its texture, colour, movement, and depth with an enormous spatial and temporal resolution, and for this purpose, the image formation (IF) dedicates the vast majority of our retinal ganglion cell (RGC) population and much of our cerebral cortex. On the other hand, a minuscule proportion of RGCs, in addition to receiving information from classic cone and rod photoreceptors, express melanopsin and are intrinsically photosensitive (ipRGC). These ipRGC are dedicated to non-image-forming (NIF) visual functions, of which we are unaware, but which are essential for aspects related to our daily physiology, such as the timing of our circadian rhythms and our pupillary light reflex, among many others. Before the discovery of ipRGCs, it was thought that the IF and NIF functions were distinct compartments regulated by different RGCs, but this concept has evolved in recent years with the discovery of new types of ipRGCs that innervate subcortical IF regions, and therefore have IF visual functions. Six different types of ipRGCs are currently known. These are termed M1-M6, and differ in their morphological, functional, molecular properties, central projections, and visual behaviour responsibilities. A review is presented on the melanopsin visual system, the most active field of research in vision, for which knowledge has grown exponentially during the last two decades, when RGCs giving rise to this pathway were first discovered.
Collapse
|
48
|
Beier C, Zhang Z, Yurgel M, Hattar S. Projections of ipRGCs and conventional RGCs to retinorecipient brain nuclei. J Comp Neurol 2020; 529:1863-1875. [PMID: 33104235 PMCID: PMC10081000 DOI: 10.1002/cne.25061] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022]
Abstract
Retinal ganglion cells (RGCs), the output neurons of the retina, allow us to perceive our visual environment. RGCs respond to rod/cone input through the retinal circuitry, however, a small population of RGCs are in addition intrinsically photosensitive (ipRGCs) and project to unique targets in the brain to modulate a broad range of subconscious visual behaviors such as pupil constriction and circadian photoentrainment. Despite the discovery of ipRGCs nearly two decades ago, there is still little information about how or if conventional RGCs (non-ipRGCs) target ipRGC-recipient nuclei to influence subconscious visual behavior. Using a dual recombinase fluorescent reporter strategy, we showed that conventional RGCs innervate many subconscious ipRGC-recipient nuclei, apart from the suprachiasmatic nucleus. We revealed previously unrecognized stratification patterns of retinal innervation from ipRGCs and conventional RGCs in the ventral portion of the lateral geniculate nucleus. Further, we found that the percent innervation of ipRGCs and conventional RGCs across ipsi- and contralateral nuclei differ. Our data provide a blueprint to understand how conventional RGCs and ipRGCs innervate different brain regions to influence subconscious visual behaviors.
Collapse
Affiliation(s)
- Corinne Beier
- Section on Light and Circadian Rhythms, NIMH, NIH, Bethesda, Maryland, USA
| | - Ze Zhang
- Section on Light and Circadian Rhythms, NIMH, NIH, Bethesda, Maryland, USA
| | - Maria Yurgel
- Section on Light and Circadian Rhythms, NIMH, NIH, Bethesda, Maryland, USA
| | - Samer Hattar
- Section on Light and Circadian Rhythms, NIMH, NIH, Bethesda, Maryland, USA
| |
Collapse
|
49
|
Perfect appearance match between self-luminous and surface colors can be performed with isomeric spectra. Sci Rep 2020; 10:18350. [PMID: 33110204 PMCID: PMC7591860 DOI: 10.1038/s41598-020-75510-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/16/2020] [Indexed: 11/08/2022] Open
Abstract
Surface color results from a reflected light bounced off a material, such as a paper. By contrast, self-luminous color results directly from an emitting light, such as a Liquid Crystal (LC) display. These are completely different mechanisms, and thus, surface color and self-luminous color cannot be matched even though both have identical tristimulus values. In fact, previous research has reported that metameric color matching fails among diverse media. However, the reason for this failure remains unclear. In the present study, we created isomeric color-matching pairs between self-luminous and surface colors by modulating the spectral distribution of the light for surface colors. Then, we experimentally verified whether such color matching can be performed. The results show that isomeric color matching between self-luminous and surface colors can be performed for all participants. However, metameric color matching fails for most participants, indicating that differences in the spectral distributions rather than the different color-generating mechanisms themselves are the reason for the color matching failure between different devices. We experimentally demonstrated that there is no essential problem in cross-media color matching by generating isomeric pairs. Our results can be considered to be of great significance not only for color science, but also for the color industry.
Collapse
|
50
|
Abstract
A retina completely devoid of topographic variations would be homogenous, encoding any given feature uniformly across the visual field. In a naive view, such homogeneity would appear advantageous. However, it is now clear that retinal topographic variations exist across mammalian species in a variety of forms and patterns. We briefly review some of the more established topographic variations in retinas of different mammalian species and focus on the recent discovery that cells belonging to a single neuronal subtype may exhibit distinct topographic variations in distribution, morphology, and even function. We concentrate on the mouse retina-originally viewed as homogenous-in which genetic labeling of distinct neuronal subtypes and other advanced techniques have revealed unexpected anatomical and physiological topographic variations. Notably, different subtypes reveal different patterns of nonuniformity, which may even be opposite or orthogonal to one another. These topographic variations in the encoding of visual space should be considered when studying visual processing in the retina and beyond.
Collapse
Affiliation(s)
- Alina Sophie Heukamp
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel; , ,
| | - Rebekah Anne Warwick
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel; , ,
| | - Michal Rivlin-Etzion
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel; , ,
| |
Collapse
|