1
|
Shi Y, Hu J, Xue T. Light, opsins, and life: Mammalian photophysiological functions beyond image perception. Neuron 2025:S0896-6273(25)00396-4. [PMID: 40527322 DOI: 10.1016/j.neuron.2025.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 05/12/2025] [Accepted: 05/23/2025] [Indexed: 06/19/2025]
Abstract
Light, a fundamental form of energy and sensory input, has significantly shaped life forms on Earth. In mammals, light perception through the eyes, which enables image formation, is crucial for survival. However, beyond image-forming (IF) vision, light also mediates non-image-forming (NIF) functions, such as circadian photoentrainment and the pupillary light reflex. Recent studies have further demonstrated that light influences a wide range of physiological and behavioral processes, including mood, metabolism, cognition, pain perception, sleep, and neuronal development. The diverse types of opsins, the major photosensitive proteins in mammals, are expressed not only in the rods, cones, and intrinsically photosensitive retinal ganglion cells (ipRGCs) of the retina but also in extraocular tissues, such as the brain, skin, and adipose tissue. Opsins in both ocular and extraocular tissues jointly contribute to light detection and mediate diverse NIF functions. In this review, we focus on the NIF effects of light on mammals, emphasizing its regulation of physiological functions as well as the corresponding roles of light receptors and associated neuronal circuits. It also highlights the implications of these findings for human health, underscoring the need for a comprehensive understanding of the interactions between light and life.
Collapse
Affiliation(s)
- Yiming Shi
- State Key Laboratory of Eye Health, CAS Key Laboratory of Brain Function and Disease, 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
| | - Jiaxi Hu
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Tian Xue
- State Key Laboratory of Eye Health, CAS Key Laboratory of Brain Function and Disease, 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.
| |
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
|
Riccitelli S, Yaakov H, Heukamp AS, Ankri L, Rivlin-Etzion M. Retinal ganglion cells encode the direction of motion outside their classical receptive field. Proc Natl Acad Sci U S A 2025; 122:e2415223122. [PMID: 39793063 PMCID: PMC11725840 DOI: 10.1073/pnas.2415223122] [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: 07/28/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Abstract
Retinal ganglion cells (RGCs) typically respond to light stimulation over their spatially restricted receptive field. Using large-scale recordings in the mouse retina, we show that a subset of non- direction-selective (DS) RGCs exhibit asymmetric activity, selective to motion direction, in response to a stimulus crossing an area far beyond the classic receptive field. The extraclassical response arises via inputs from an asymmetric distal zone and is enhanced by desensitization mechanisms and an inherent DS component, creating a network of neurons responding to motion toward the optic disc. Pharmacological manipulations revealed the necessity of glycinergic amacrine cells for this response. Using in vivo recordings, we identified similar extraclassical responses in lateral geniculate nucleus neurons, suggesting such non conventional DS information is transferred to downstream structures. Our results suggest a complex integration of motion direction processing across the visual field, which arises beyond the classical receptive field boundaries.
Collapse
Affiliation(s)
- Serena Riccitelli
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Hadar Yaakov
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Alina S. Heukamp
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Lea Ankri
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Michal Rivlin-Etzion
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zangen E, Hadar S, Lawrence C, Obeid M, Rasras H, Hanzin E, Aslan O, Zur E, Schulcz N, Cohen-Hatab D, Samama Y, Nir S, Li Y, Dobrotvorskia I, Sabbah S. Prefrontal cortex neurons encode ambient light intensity differentially across regions and layers. Nat Commun 2024; 15:5501. [PMID: 38951486 PMCID: PMC11217280 DOI: 10.1038/s41467-024-49794-w] [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: 08/09/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
While light can affect emotional and cognitive processes of the medial prefrontal cortex (mPFC), no light-encoding was hitherto identified in this region. Here, extracellular recordings in awake mice revealed that over half of studied mPFC neurons showed photosensitivity, that was diminished by inhibition of intrinsically photosensitive retinal ganglion cells (ipRGCs), or of the upstream thalamic perihabenular nucleus (PHb). In 15% of mPFC photosensitive neurons, firing rate changed monotonically along light-intensity steps and gradients. These light-intensity-encoding neurons comprised four types, two enhancing and two suppressing their firing rate with increased light intensity. Similar types were identified in the PHb, where they exhibited shorter latency and increased sensitivity. Light suppressed prelimbic activity but boosted infralimbic activity, mirroring the regions' contrasting roles in fear-conditioning, drug-seeking, and anxiety. We posit that prefrontal photosensitivity represents a substrate of light-susceptible, mPFC-mediated functions, which could be ultimately studied as a therapeutical target in psychiatric and addiction disorders.
Collapse
Affiliation(s)
- Elyashiv Zangen
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Shira Hadar
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Christopher Lawrence
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Mustafa Obeid
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Hala Rasras
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Ella Hanzin
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Ori Aslan
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Eyal Zur
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Nadav Schulcz
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Daniel Cohen-Hatab
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Yona Samama
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Sarah Nir
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Yi Li
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Irina Dobrotvorskia
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel
| | - Shai Sabbah
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112102, Israel.
| |
Collapse
|
7
|
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
|
8
|
Aranda ML, Bhoi JD, Parra OAP, Lee SK, Yamada T, Yang Y, Schmidt TM. Genetic tuning of intrinsically photosensitive retinal ganglion cell subtype identity to drive visual behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590656. [PMID: 38712084 PMCID: PMC11071530 DOI: 10.1101/2024.04.25.590656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) comprise a subset of the ∼40 retinal ganglion cell types in the mouse retina and drive a diverse array of light-evoked behaviors from circadian photoentrainment to pupil constriction to contrast sensitivity for visual perception. Central to the ability of ipRGCs to control this diverse array of behaviors is the distinct complement of morphophysiological features and gene expression patterns found in the M1-M6 ipRGC subtypes. However, the genetic regulatory programs that give rise to subtypes of ipRGCs are unknown. Here, we identify the transcription factor Brn3b (Pou4f2) as a key genetic regulator that shapes the unique functions of ipRGC subtypes and their diverse downstream visual behaviors.
Collapse
|
9
|
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
|
10
|
Mahoney HL, Schmidt TM. The cognitive impact of light: illuminating ipRGC circuit mechanisms. Nat Rev Neurosci 2024; 25:159-175. [PMID: 38279030 DOI: 10.1038/s41583-023-00788-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/28/2024]
Abstract
Ever-present in our environments, light entrains circadian rhythms over long timescales, influencing daily activity patterns, health and performance. Increasing evidence indicates that light also acts independently of the circadian system to directly impact physiology and behaviour, including cognition. Exposure to light stimulates brain areas involved in cognition and appears to improve a broad range of cognitive functions. However, the extent of these effects and their mechanisms are unknown. Intrinsically photosensitive retinal ganglion cells (ipRGCs) have emerged as the primary conduit through which light impacts non-image-forming behaviours and are a prime candidate for mediating the direct effects of light on cognition. Here, we review the current state of understanding of these effects in humans and mice, and the tools available to uncover circuit-level and photoreceptor-specific mechanisms. We also address current barriers to progress in this area. Current and future efforts to unravel the circuits through which light influences cognitive functions may inform the tailoring of lighting landscapes to optimize health and cognitive function.
Collapse
Affiliation(s)
- Heather L Mahoney
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
11
|
Contreras E, Bhoi JD, Sonoda T, Birnbaumer L, Schmidt TM. Melanopsin activates divergent phototransduction pathways in intrinsically photosensitive retinal ganglion cell subtypes. eLife 2023; 12:e80749. [PMID: 37937828 PMCID: PMC10712949 DOI: 10.7554/elife.80749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/06/2023] [Indexed: 11/09/2023] Open
Abstract
Melanopsin signaling within intrinsically photosensitive retinal ganglion cell (ipRGC) subtypes impacts a broad range of behaviors from circadian photoentrainment to conscious visual perception. Yet, how melanopsin phototransduction within M1-M6 ipRGC subtypes impacts cellular signaling to drive diverse behaviors is still largely unresolved. The identity of the phototransduction channels in each subtype is key to understanding this central question but has remained controversial. In this study, we resolve two opposing models of M4 phototransduction, demonstrating that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are dispensable for this process and providing support for a pathway involving melanopsin-dependent potassium channel closure and canonical transient receptor potential (TRPC) channel opening. Surprisingly, we find that HCN channels are likewise dispensable for M2 phototransduction, contradicting the current model. We instead show that M2 phototransduction requires TRPC channels in conjunction with T-type voltage-gated calcium channels, identifying a novel melanopsin phototransduction target. Collectively, this work resolves key discrepancies in our understanding of ipRGC phototransduction pathways in multiple subtypes and adds to mounting evidence that ipRGC subtypes employ diverse phototransduction cascades to fine-tune cellular responses for downstream behaviors.
Collapse
Affiliation(s)
- Ely Contreras
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- Northwestern University Interdisciplinary Biological Sciences Program, Northwestern UniversityEvanstonUnited States
| | - Jacob D Bhoi
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- Northwestern University Interdepartmental Neuroscience Program, Northwestern UniversityChicagoUnited States
| | - Takuma Sonoda
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- Northwestern University Interdepartmental Neuroscience Program, Northwestern UniversityChicagoUnited States
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute of Environmental Health SciencesDurhamUnited States
- Institute of Biomedical Research (BIOMED), Catholic University of ArgentinaBuenos AiresArgentina
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- Department of Ophthalmology, Feinberg School of MedicineChicagoUnited States
| |
Collapse
|
12
|
Pan D, Wang Z, Chen Y, Cao J. Melanopsin-mediated optical entrainment regulates circadian rhythms in vertebrates. Commun Biol 2023; 6:1054. [PMID: 37853054 PMCID: PMC10584931 DOI: 10.1038/s42003-023-05432-7] [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: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Melanopsin (OPN4) is a light-sensitive protein that plays a vital role in the regulation of circadian rhythms and other nonvisual functions. Current research on OPN4 has focused on mammals; more evidence is needed from non-mammalian vertebrates to fully assess the significance of the non-visual photosensitization of OPN4 for circadian rhythm regulation. There are species differences in the regulatory mechanisms of OPN4 for vertebrate circadian rhythms, which may be due to the differences in the cutting variants, tissue localization, and photosensitive activation pathway of OPN4. We here summarize the distribution of OPN4 in mammals, birds, and teleost fish, and the classical excitation mode for the non-visual photosensitive function of OPN4 in mammals is discussed. In addition, the role of OPN4-expressing cells in regulating circadian rhythm in different vertebrates is highlighted, and the potential rhythmic regulatory effects of various neuropeptides or neurotransmitters expressed in mammalian OPN4-expressing ganglion cells are summarized among them.
Collapse
Affiliation(s)
- Deng Pan
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China.
| |
Collapse
|
13
|
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
|
14
|
Berry MH, Moldavan M, Garrett T, Meadows M, Cravetchi O, White E, Leffler J, von Gersdorff H, Wright KM, Allen CN, Sivyer B. A melanopsin ganglion cell subtype forms a dorsal retinal mosaic projecting to the supraoptic nucleus. Nat Commun 2023; 14:1492. [PMID: 36932080 PMCID: PMC10023714 DOI: 10.1038/s41467-023-36955-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Visual input to the hypothalamus from intrinsically photosensitive retinal ganglion cells (ipRGCs) influences several functions including circadian entrainment, body temperature, and sleep. ipRGCs also project to nuclei such as the supraoptic nucleus (SON), which is involved in systemic fluid homeostasis, maternal behavior, social behaviors, and appetite. However, little is known about the SON-projecting ipRGCs or their relationship to well-characterized ipRGC subtypes. Using a GlyT2Cre mouse line, we show a subtype of ipRGCs restricted to the dorsal retina that selectively projects to the SON. These ipRGCs tile a dorsal region of the retina, forming a substrate for encoding ground luminance. Optogenetic activation of their axons demonstrates they release the neurotransmitter glutamate in multiple regions, including the suprachiasmatic nucleus (SCN) and SON. Our results challenge the idea that ipRGC dendrites overlap to optimize photon capture and suggests non-image forming vision operates to sample local regions of the visual field to influence diverse behaviors.
Collapse
Affiliation(s)
- Michael H Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Medical Scientist Training Program, Oregon Health & Science University, Portland, OR, USA
| | - Michael Moldavan
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Tavita Garrett
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
| | - Marc Meadows
- Neuroscience Graduate program, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Henrique von Gersdorff
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Charles N Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA.
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
15
|
Santana NNM, Silva EHA, dos Santos SF, Costa MSMO, Nascimento Junior ES, Engelberth RCJG, Cavalcante JS. Retinorecipient areas in the common marmoset ( Callithrix jacchus): An image-forming and non-image forming circuitry. Front Neural Circuits 2023; 17:1088686. [PMID: 36817647 PMCID: PMC9932520 DOI: 10.3389/fncir.2023.1088686] [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: 11/03/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
The mammalian retina captures a multitude of diverse features from the external environment and conveys them via the optic nerve to a myriad of retinorecipient nuclei. Understanding how retinal signals act in distinct brain functions is one of the most central and established goals of neuroscience. Using the common marmoset (Callithrix jacchus), a monkey from Northeastern Brazil, as an animal model for parsing how retinal innervation works in the brain, started decades ago due to their marmoset's small bodies, rapid reproduction rate, and brain features. In the course of that research, a large amount of new and sophisticated neuroanatomical techniques was developed and employed to explain retinal connectivity. As a consequence, image and non-image-forming regions, functions, and pathways, as well as retinal cell types were described. Image-forming circuits give rise directly to vision, while the non-image-forming territories support circadian physiological processes, although part of their functional significance is uncertain. Here, we reviewed the current state of knowledge concerning retinal circuitry in marmosets from neuroanatomical investigations. We have also highlighted the aspects of marmoset retinal circuitry that remain obscure, in addition, to identify what further research is needed to better understand the connections and functions of retinorecipient structures.
Collapse
Affiliation(s)
- Nelyane Nayara M. Santana
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eryck H. A. Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sâmarah F. dos Santos
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Miriam S. M. O. Costa
- Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Expedito S. Nascimento Junior
- Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Rovena Clara J. G. Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jeferson S. Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil,*Correspondence: Jeferson S. Cavalcante,
| |
Collapse
|
16
|
Fu Y, Liu S, Dong Y, Gan Y, Guo X, Liu H, Xu Q, Yuan R, Ning A, Hong W, Peng Y, Yu S. Chronic restraint stress-induced depression-like behavior is mediated by upregulation of melanopsin expression in C57BL/6 mice retina. Psychopharmacology (Berl) 2023; 240:283-293. [PMID: 36580134 DOI: 10.1007/s00213-022-06302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Depression is associated with circadian disturbances in which melanopsin was a key mechanism. Further studies have demonstrated that melanopsin gene variations are associated with some depressive disorders and aberrant light can impair mood through melanopsin-expressing retinal ganglion cells (mRGCs). The goal of this study was to explore the direct relationship between depression and melanopsin. METHODS Adult C57BL/6 male mice were physically restrained for 16 h in a 50-ml polypropylene centrifuge tube and all behavioral tests were performed after CRS treatment. Western blot analysis and immunofluorescence were used to detect melanopsin expression in the retina of C57BL/6 mice. And we observed the change of the electrophysiological function and release of glutamate of mRGCs. RESULTS The melanopsin expression upregulate in mRGCs of chronic restraint stress (CRS)-treating mice which exhibit depression-like behavior. The frequency of blue light-induced action potentials and light-induced glutamate release mediated by melanopsin also increase significantly. This change of melanopsin is mediated by the CRS-induced glucocorticoid. CONCLUSIONS CRS may induce the depression-like behavior in mice via glucocorticoid-melanopsin pathway. Our findings provide a novel mechanistic link between CRS-induced depression and melanopsin in mice.
Collapse
Affiliation(s)
- Yingmei Fu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shanshan Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing, China
| | - Yigang Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, 200241, China
| | - Yixia Gan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, 200241, China
| | - Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Liu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixue Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ailing Ning
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanmin Peng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shunying Yu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Tapia ML, Nascimento-Dos-Santos G, Park KK. Subtype-specific survival and regeneration of retinal ganglion cells in response to injury. Front Cell Dev Biol 2022; 10:956279. [PMID: 36035999 PMCID: PMC9411869 DOI: 10.3389/fcell.2022.956279] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons that function synchronously to convey visual information through the optic nerve to retinorecipient target areas in the brain. Injury or disease to the optic nerve results in RGC degeneration and loss of visual function, as few RGCs survive, and even fewer can be provoked to regenerate their axons. Despite causative insults being broadly shared, regeneration studies demonstrate that RGC types exhibit differential resilience to injury and undergo selective survival and regeneration of their axons. While most early studies have identified these RGC types based their morphological and physiological characteristics, recent advances in transgenic and gene sequencing technologies have further enabled type identification based on unique molecular features. In this review, we provide an overview of the well characterized RGC types and identify those shown to preferentially survive and regenerate in various regeneration models. Furthermore, we discuss cellular characteristics of both the resilient and susceptible RGC types including the combinatorial expression of different molecular markers that identify these specific populations. Lastly, we discuss potential molecular mechanisms and genes found to be selectively expressed by specific types that may contribute to their reparative capacity. Together, we describe the studies that lay the important groundwork for identifying factors that promote neural regeneration and help advance the development of targeted therapy for the treatment of RGC degeneration as well as neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Mary L Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gabriel Nascimento-Dos-Santos
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
20
|
Sabbah S, Worden MS, Laniado DD, Berson DM, Sanes JN. Luxotonic signals in human prefrontal cortex as a possible substrate for effects of light on mood and cognition. Proc Natl Acad Sci U S A 2022; 119:e2118192119. [PMID: 35867740 PMCID: PMC9282370 DOI: 10.1073/pnas.2118192119] [Citation(s) in RCA: 9] [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: 10/03/2021] [Accepted: 05/16/2022] [Indexed: 01/09/2023] Open
Abstract
Studies with experimental animals have revealed a mood-regulating neural pathway linking intrinsically photosensitive retinal ganglion cells (ipRGCs) and the prefrontal cortex (PFC), involved in the pathophysiology of mood disorders. Since humans also have light-intensity-encoding ipRGCs, we asked whether a similar pathway exists in humans. Here, functional MRI was used to identify PFC regions and other areas exhibiting light-intensity-dependent signals. We report 26 human brain regions having activation that either monotonically decreases or monotonically increases with light intensity. Luxotonic-related activation occurred across the cerebral cortex, in diverse subcortical structures, and in the cerebellum, encompassing regions with functions related to visual image formation, motor control, cognition, and emotion. Light suppressed PFC activation, which monotonically decreased with increasing light intensity. The sustained time course of light-evoked PFC responses and their susceptibility to prior light exposure resembled those of ipRGCs. These findings offer a functional link between light exposure and PFC-mediated cognitive and affective phenomena.
Collapse
Affiliation(s)
- Shai Sabbah
- Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Michael S. Worden
- Department of Neuroscience, Brown University, Providence, RI 02912
- Carney Institute for Brain Science, Brown University, Providence, RI 02912
| | - Dimitrios D. Laniado
- Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - David M. Berson
- Department of Neuroscience, Brown University, Providence, RI 02912
- Carney Institute for Brain Science, Brown University, Providence, RI 02912
| | - Jerome N. Sanes
- Department of Neuroscience, Brown University, Providence, RI 02912
- Carney Institute for Brain Science, Brown University, Providence, RI 02912
- Center for Neurorestoration and Neurotechnology, Veterans Affairs Providence Healthcare System, Providence, RI 02908
| |
Collapse
|
21
|
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
|
22
|
Guido ME, Marchese NA, Rios MN, Morera LP, Diaz NM, Garbarino-Pico E, Contin MA. Non-visual Opsins and Novel Photo-Detectors in the Vertebrate Inner Retina Mediate Light Responses Within the Blue Spectrum Region. Cell Mol Neurobiol 2022; 42:59-83. [PMID: 33231827 PMCID: PMC11441211 DOI: 10.1007/s10571-020-00997-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
In recent decades, a number of novel non-visual opsin photopigments belonging to the family of G protein- coupled receptors, likely involved in a number of non-image-forming processes, have been identified and characterized in cells of the inner retina of vertebrates. It is now known that the vertebrate retina is composed of visual photoreceptor cones and rods responsible for diurnal/color and nocturnal/black and white vision, and cells like the intrinsically photosensitive retinal ganglion cells (ipRGCs) and photosensitive horizontal cells in the inner retina, both detecting blue light and expressing the photopigment melanopsin (Opn4). Remarkably, these non-visual photopigments can continue to operate even in the absence of vision under retinal degeneration. Moreover, inner retinal neurons and Müller glial cells have been shown to express other photopigments such as the photoisomerase retinal G protein-coupled receptor (RGR), encephalopsin (Opn3), and neuropsin (Opn5), all able to detect blue/violet light and implicated in chromophore recycling, retinal clock synchronization, neuron-to-glia communication, and other activities. The discovery of these new photopigments in the inner retina of vertebrates is strong evidence of novel light-regulated activities. This review focuses on the features, localization, photocascade, and putative functions of these novel non-visual opsins in an attempt to shed light on their role in the inner retina of vertebrates and in the physiology of the whole organism.
Collapse
Affiliation(s)
- Mario E Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina.
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina.
| | - Natalia A Marchese
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Maximiliano N Rios
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Luis P Morera
- Instituto de Organizaciones Saludables, Universidad Siglo 21, Córdoba, Argentina
| | - Nicolás M Diaz
- Department of Ophthalmology, University of Washington School of Medicine, 750 Republican St., Campus, Box 358058, Seattle, WA, 98109, USA
| | - Eduardo Garbarino-Pico
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - María Ana Contin
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| |
Collapse
|
23
|
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
|
24
|
Lee S, Chen M, Shi Y, Zhou ZJ. Selective glycinergic input from vGluT3 amacrine cells confers a suppressed-by-contrast trigger feature in a subtype of M1 ipRGCs in the mouse retina. J Physiol 2021; 599:5047-5060. [PMID: 34292589 PMCID: PMC8741526 DOI: 10.1113/jp281717] [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: 03/31/2021] [Accepted: 07/21/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS M1 intrinsically photosensitive retinal ganglion cells (ipRGCs) are known to encode absolute light intensity (irradiance) for non-image-forming visual functions (subconscious vision), such as circadian photoentrainment and the pupillary light reflex. It remains unclear how M1 cells respond to relative light intensity (contrast) and patterned visual signals. The present study identified a special form of contrast sensitivity (suppressed-by-contrast) in M1 cells, suggesting a role of patterned visual signals in regulating non-image-forming vision and a potential role of M1 ipRGCs in encoding image-forming visual cues. The study also uncovered a synaptic mechanism and a retinal circuit mediated by vesicular glutamate transporter 3 (vGluT3) amacrine cells that underlie the suppressed-by-contrast response of M1 cells. M1 ipRGC subtypes (M1a and M1b) were revealed that are distinguishable based on synaptic connectivity with vGluT3 amacrine cells, receptive field properties, intrinsic photo sensitivity and membrane excitability, and morphological features, suggesting a division of visual tasks among discrete M1 subpopulations. ABSTRACT The M1 type ipRGC (intrinsically photosensitive retinal ganglion cell) is known to encode ambient light signals for non-image-forming visual functions such as circadian photo-entrainment and the pupillary light reflex. Here, we report that a subpopulation of M1 cells (M1a) in the mouse retina possess the suppressed-by-contrast (sbc) trigger feature that is a receptive field property previously found only in ganglion cells mediating image-forming vision. Using optogenetics and the dual patch clamp technique, we found that vesicular glutamate transporter 3 (vGluT3) (vGluT3) amacrine cells make glycinergic, but not glutamatergic, synapses specifically onto M1a cells. The spatiotemporal and pharmacological properties of visually evoked responses of M1a cells closely matched the receptive field characteristics of vGluT3 cells, suggesting a major role of the vGluT3 amacrine cell input in shaping the sbc trigger feature of M1a cells. We found that the other subpopulation of M1 cells (M1b), which did not receive a direct vGluT3 cell input, lacked the sbc trigger feature, being distinctively different from M1a cells in intrinsic photo responses, membrane excitability, receptive-field characteristics and morphological features. Together, the results reveal a retinal circuit that uses the sbc trigger feature to regulate irradiance coding and potentially send image-forming cues to non-image-forming visual centres in the brain.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Minggang Chen
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Yuelin Shi
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Z Jimmy Zhou
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
25
|
Arns M, Kooij JJS, Coogan AN. Review: Identification and Management of Circadian Rhythm Sleep Disorders as a Transdiagnostic Feature in Child and Adolescent Psychiatry. J Am Acad Child Adolesc Psychiatry 2021; 60:1085-1095. [PMID: 33556454 DOI: 10.1016/j.jaac.2020.12.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/24/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Sleep disturbances are highly frequent features in a range of child and adolescent psychiatric conditions. However, it is commonly not clear if such sleep problems represent symptomatic features of, comorbidities of, or risk factors for these conditions. It is believed that underlying dysfunction in the daily biological (circadian) clock may play important roles in the etiology of many sleep disorders, and circadian rhythm changes are reported in a number of neuropsychiatric conditions. The aim of this review was to explore the key identifying features of circadian rhythm disorders (CRDs) in child and adolescent psychiatry and address how such disorders may be managed in the clinic. METHOD A narrative review was conducted of the extant literature of CRDs in children and adolescents with psychiatric conditions. RESULTS Key biological and social factors that contribute to CRDs in children and adolescents, and the cognitive and neurobehavioral consequences resulting from insufficient sleep were outlined. The roles of melatonin and other chronotherapeutic and behavioral interventions for the management of CRDs were also outlined. Further, the importance of careful investigation of circadian rhythm abnormalities in shaping the most effective treatment plan according to chronobiological principles was highlighted. CONCLUSION CRDs are common in children and adolescents with psychiatric conditions and arise out of complex interactions between biological and social factors. Careful clinical attention to and management of CRDs in child and adolescent psychiatry have the potential for significant benefit not only in the domain of sleep but also in a range of cognitive, affective, and behavioral outcomes.
Collapse
Affiliation(s)
- Martijn Arns
- Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Research Institute Brainclinics, Brainclinics Foundation, Nijmegen, and Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - J J Sandra Kooij
- Amsterdam University Medical Center, Amsterdam, and PsyQ, Expertise Center Adult ADHD, the Hague, the Netherlands
| | | |
Collapse
|
26
|
Opsins outside the eye and the skin: a more complex scenario than originally thought for a classical light sensor. Cell Tissue Res 2021; 385:519-538. [PMID: 34236517 DOI: 10.1007/s00441-021-03500-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Since the discovery of melanopsin as a retinal non-visual photopigment, opsins have been described in several organs and cells. This distribution is strikingly different from the classical localization of photopigments in light-exposed tissues such as the eyes and the skin. More than 10 years ago, a new paradigm in the field was created as opsins were shown, to detect not only light, but also thermal energy in Drosophila. In agreement with these findings, thermal detection by opsins was also reported in mammalian cells. Considering the presence of opsins in tissues not reached by light, an intriguing question has emerged: What is the role of a classical light-sensor, and more recently appreciated thermo-sensor, in these tissues? To tackle this question, we address in this review the most recent studies in the field, with emphasis in mammals. We provide the present view about the role of opsins in peripheral tissues, aiming to integrate the current knowledge of the presence and function of opsins in organs that are not directly affected by light.
Collapse
|
27
|
Photobiological Neuromodulation of Resting-State EEG and Steady-State Visual-Evoked Potentials by 40 Hz Violet Light Optical Stimulation in Healthy Individuals. J Pers Med 2021; 11:jpm11060557. [PMID: 34203878 PMCID: PMC8232632 DOI: 10.3390/jpm11060557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
Photobiological neuromodulation and its clinical application has been investigated in recent years. The response of the gamma-oscillation to human visual stimuli is known to be both burst and resonant in nature, and the coupling between alpha and gamma oscillations may play a functional role in visual processing. To date, there is no study that examined the effects of gamma-frequency violet light (VL) stimulation on human electroencephalography (EEG). In this study, we investigated the neurophysiological changes induced by light stimulation using EEG. The purpose of this study was to evaluate the specific effects of 40 Hz gamma-frequency VL stimulation on EEG activity by comparing the effects of white light (WL) with the same condition. Twenty healthy participants (10 females: 37.5 ± 14.3 years; 10 males: 38.0 ± 13.3 years) participated in this study and the following results were observed. First, when compared with the power spectrum density (PSD) of baseline EEG, 40 Hz-WL induced significant increase of PSD in theta band. Second, compared the PSDs between EEG with 40 Hz-VL and EEG with 40 Hz-WL, 40 Hz-VL induced significantly lower enhancement in delta and theta bands than 40 Hz-WL. Third, when focused on the occipital area, negative peak of VEP with 40 Hz-VL was smaller than that of 40 Hz-WL. Fourth, 40 Hz-VL induced an increase of alpha-gamma coupling during the VEP at the F5 electrode site as well as post-EEG at the C4 electrode site, compared with baseline EEG. Thus, the present study suggested that 40 Hz-VL stimulation may induce unique photobiological neuromodulations on human EEG activity.
Collapse
|
28
|
Stinchcombe AR, Hu C, Walch OJ, Faught SD, Wong KY, Forger DB. M1-Type, but Not M4-Type, Melanopsin Ganglion Cells Are Physiologically Tuned to the Central Circadian Clock. Front Neurosci 2021; 15:652996. [PMID: 34025341 PMCID: PMC8134526 DOI: 10.3389/fnins.2021.652996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022] Open
Abstract
Proper circadian photoentrainment is crucial for the survival of many organisms. In mammals, intrinsically photosensitive retinal ganglion cells (ipRGCs) can use the photopigment melanopsin to sense light independently from rod and cone photoreceptors and send this information to many brain nuclei such as the suprachiasmatic nucleus (SCN), the site of the central circadian pacemaker. Here, we measure ionic currents and develop mathematical models of the electrical activity of two types of ipRGCs: M1, which projects to the SCN, and M4, which does not. We illustrate how their ionic properties differ, mainly how ionic currents generate lower spike rates and depolarization block in M1 ipRGCs. Both M1 and M4 cells have large geometries and project to higher visual centers of the brain via the optic nerve. Using a partial differential equation model, we show how axons of M1 and M4 cells faithfully convey information from the soma to the synapse even when the signal at the soma is attenuated due to depolarization block. Finally, we consider an ionic model of circadian photoentrainment from ipRGCs synapsing on SCN neurons and show how the properties of M1 ipRGCs are tuned to create accurate transmission of visual signals from the retina to the central pacemaker, whereas M4 ipRGCs would not evoke nearly as efficient a postsynaptic response. This work shows how ipRGCs and SCN neurons' electrical activities are tuned to allow for accurate circadian photoentrainment.
Collapse
Affiliation(s)
| | - Caiping Hu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Olivia J Walch
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Samuel D Faught
- Department of Mathematics, University of Michigan, Ann Arbor, MI, United States
| | - Kwoon Y Wong
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States.,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Daniel B Forger
- Department of Mathematics, University of Michigan, Ann Arbor, MI, United States.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States.,Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
29
|
Mouland JW, Martial FP, Lucas RJ, Brown TM. Modulations in irradiance directed at melanopsin, but not cone photoreceptors, reliably alter electrophysiological activity in the suprachiasmatic nucleus and circadian behaviour in mice. J Pineal Res 2021; 70:e12735. [PMID: 33793975 DOI: 10.1111/jpi.12735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 12/25/2022]
Abstract
Intrinsically photosensitive retinal ganglion cells convey intrinsic, melanopsin-based, photoreceptive signals alongside those produced by rods and cones to the suprachiasmatic nucleus (SCN) circadian clock. To date, experimental data suggest that melanopsin plays a more significant role in measuring ambient light intensity than cone photoreception. Such studies have overwhelmingly used diffuse light stimuli, whereas light intensity in the world around us varies across space and time. Here, we investigated the extent to which melanopsin or cone signals support circadian irradiance measurements in the presence of naturalistic spatiotemporal variations in light intensity. To address this, we first presented high- and low-contrast movies to anaesthetised mice whilst recording extracellular electrophysiological activity from the SCN. Using a mouse line with altered cone sensitivity (Opn1mwR mice) and multispectral light sources we then selectively varied irradiance of the movies for specific photoreceptor classes. We found that steps in melanopic irradiance largely account for the light induced-changes in SCN activity over a range of starting light intensities and in the presence of spatiotemporal modulation. By contrast, cone-directed changes in irradiance only influenced SCN activity when spatiotemporal contrast was low. Consistent with these findings, under housing conditions where we could independently adjust irradiance for melanopsin versus cones, the period lengthening effects of constant light on circadian rhythms in behaviour were reliably determined by melanopic irradiance, regardless of irradiance for cones. These data add to the growing evidence that modulating effective irradiance for melanopsin is an effective strategy for controlling the circadian impact of light.
Collapse
Affiliation(s)
- Josh W Mouland
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Franck P Martial
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Robert J Lucas
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- Division of Neuroscience and Experimental Psychology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Timothy M Brown
- Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
30
|
Tabuchi M, Coates KE, Bautista OB, Zukowski LH. Light/Clock Influences Membrane Potential Dynamics to Regulate Sleep States. Front Neurol 2021; 12:625369. [PMID: 33854471 PMCID: PMC8039321 DOI: 10.3389/fneur.2021.625369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
The circadian rhythm is a fundamental process that regulates the sleep-wake cycle. This rhythm is regulated by core clock genes that oscillate to create a physiological rhythm of circadian neuronal activity. However, we do not know much about the mechanism by which circadian inputs influence neurons involved in sleep-wake architecture. One possible mechanism involves the photoreceptor cryptochrome (CRY). In Drosophila, CRY is receptive to blue light and resets the circadian rhythm. CRY also influences membrane potential dynamics that regulate neural activity of circadian clock neurons in Drosophila, including the temporal structure in sequences of spikes, by interacting with subunits of the voltage-dependent potassium channel. Moreover, several core clock molecules interact with voltage-dependent/independent channels, channel-binding protein, and subunits of the electrogenic ion pump. These components cooperatively regulate mechanisms that translate circadian photoreception and the timing of clock genes into changes in membrane excitability, such as neural firing activity and polarization sensitivity. In clock neurons expressing CRY, these mechanisms also influence synaptic plasticity. In this review, we propose that membrane potential dynamics created by circadian photoreception and core clock molecules are critical for generating the set point of synaptic plasticity that depend on neural coding. In this way, membrane potential dynamics drive formation of baseline sleep architecture, light-driven arousal, and memory processing. We also discuss the machinery that coordinates membrane excitability in circadian networks found in Drosophila, and we compare this machinery to that found in mammalian systems. Based on this body of work, we propose future studies that can better delineate how neural codes impact molecular/cellular signaling and contribute to sleep, memory processing, and neurological disorders.
Collapse
Affiliation(s)
- Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | | | | |
Collapse
|
31
|
Mure LS. Intrinsically Photosensitive Retinal Ganglion Cells of the Human Retina. Front Neurol 2021; 12:636330. [PMID: 33841306 PMCID: PMC8027232 DOI: 10.3389/fneur.2021.636330] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Light profoundly affects our mental and physical health. In particular, light, when not delivered at the appropriate time, may have detrimental effects. In mammals, light is perceived not only by rods and cones but also by a subset of retinal ganglion cells that express the photopigment melanopsin that renders them intrinsically photosensitive (ipRGCs). ipRGCs participate in contrast detection and play critical roles in non-image-forming vision, a set of light responses that include circadian entrainment, pupillary light reflex (PLR), and the modulation of sleep/alertness, and mood. ipRGCs are also found in the human retina, and their response to light has been characterized indirectly through the suppression of nocturnal melatonin and PLR. However, until recently, human ipRGCs had rarely been investigated directly. This gap is progressively being filled as, over the last years, an increasing number of studies provided descriptions of their morphology, responses to light, and gene expression. Here, I review the progress in our knowledge of human ipRGCs, in particular, the different morphological and functional subtypes described so far and how they match the murine subtypes. I also highlight questions that remain to be addressed. Investigating ipRGCs is critical as these few cells play a major role in our well-being. Additionally, as ipRGCs display increased vulnerability or resilience to certain disorders compared to conventional RGCs, a deeper knowledge of their function could help identify therapeutic approaches or develop diagnostic tools. Overall, a better understanding of how light is perceived by the human eye will help deliver precise light usage recommendations and implement light-based therapeutic interventions to improve cognitive performance, mood, and life quality.
Collapse
Affiliation(s)
- Ludovic S Mure
- Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Neurology, Zentrum für Experimentelle Neurologie, Inselspital University Hospital Bern, Bern, Switzerland
| |
Collapse
|
32
|
Abstract
SIGNIFICANCE Pupillometry protocols evaluating rod/cone- and melanopsin-driven responses often use mydriatics to ensure maximal stimulus exposure; however, retinal effects of mydriatics are not fully understood. We demonstrate that dilation with either atropine or phenylephrine results in similar enhancements of rod/cone- and melanopsin-driven pupil responses. PURPOSE The purposes of this study were to compare the effects of atropine, a muscarinic antagonist, and phenylephrine, an adrenergic agonist, on consensual pupil responses and to assess the repeatability of pupil metrics without mydriasis. METHODS Right eye pupil responses of 20 adults aged 21 to 42 years were recorded before and 45 minutes after instillation of 0.5% atropine or 2.5% phenylephrine in the left eye. Stimuli were presented to the left eye and included six alternating 1-second 651-nm "red" and 456-nm "blue" flashes. Metrics included baseline pupil diameter, maximal constriction, 6- and 30-second post-illumination pupil responses, and early (0 to 10 seconds) and late (10 to 30 seconds) areas under the curve. RESULTS Dilation of the stimulated eye with either mydriatic significantly increased the 6-second post-illumination pupil response and early and late areas under the curve for blue stimuli, and early area under the curve for red stimuli (P < .05 for all). Melanopsin-driven post-illumination pupil responses, achieved with either phenylephrine or atropine, did not significantly differ from each other (P > .05 for all). Without mydriasis, intersession intraclass correlation coefficients for pupil metrics were 0.63 and 0.50 (6- and 30-second post-illumination pupil responses, respectively) and 0.78 and 0.44 (early and late areas under the curve, respectively) for blue stimuli, with no significant difference between sessions (P > .05 for all). CONCLUSIONS Dilation with phenylephrine or atropine resulted in similar enhancements of the rod/cone- and melanopsin-driven pupil responses, despite differing mechanisms. Early pupil metrics without mydriasis demonstrated moderate to good intersession repeatability.
Collapse
|
33
|
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
|
34
|
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
|
35
|
Aranda ML, Schmidt TM. Diversity of intrinsically photosensitive retinal ganglion cells: circuits and functions. Cell Mol Life Sci 2021; 78:889-907. [PMID: 32965515 PMCID: PMC8650628 DOI: 10.1007/s00018-020-03641-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022]
Abstract
The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) are a relatively recently discovered class of atypical ganglion cell photoreceptor. These ipRGCs are a morphologically and physiologically heterogeneous population that project widely throughout the brain and mediate a wide array of visual functions ranging from photoentrainment of our circadian rhythms, to driving the pupillary light reflex to improve visual function, to modulating our mood, alertness, learning, sleep/wakefulness, regulation of body temperature, and even our visual perception. The presence of melanopsin as a unique molecular signature of ipRGCs has allowed for the development of a vast array of molecular and genetic tools to study ipRGC circuits. Given the emerging complexity of this system, this review will provide an overview of the genetic tools and methods used to study ipRGCs, how these tools have been used to dissect their role in a variety of visual circuits and behaviors in mice, and identify important directions for future study.
Collapse
Affiliation(s)
- Marcos L Aranda
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
36
|
Melanopic Limits of Metamer Spectral Optimisation in Multi-Channel Smart Lighting Systems. ENERGIES 2021. [DOI: 10.3390/en14030527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Modern indoor lighting faces the challenge of finding an appropriate balance between energy consumption, legal requirements, visual performance, and the circadian effectiveness of a spectrum. Multi-channel LED luminaires have the option of keeping image-forming metrics steady while varying the melanopic radiance through metamer spectra for non-visual purposes. Here, we propose the theoretical concept of an automated smart lighting system that is designed to satisfy the user’s visual preference through neural networks while triggering the non-visual pathway via metamers. To quantify the melanopic limits of metamers at a steady chromaticity point, we have used 561 chromaticity coordinates along the Planckian locus (2700 K to 7443 K, ±Duv 0 to 0.048) as optimisation targets and generated the spectra by using a 6-channel, 8-channel, and 11-channel LED combination at three different luminance levels. We have found that in a best-case scenario, the melanopic radiance can be varied up to 65% while keeping the chromaticity coordinates constant (Δu′v′≤7.05×10−5) by using metamer spectra. The highest melanopic metamer contrast can be reached near the Planckian locus between 3292 and 4717 K within a Duv range of −0.009 to 0.006. Additionally, we publish over 1.2 million optimised spectra generated by multichannel LED luminaires as an open-source dataset along with this work.
Collapse
|
37
|
Harrison KR, Chervenak AP, Resnick SM, Reifler AN, Wong KY. Amacrine Cells Forming Gap Junctions With Intrinsically Photosensitive Retinal Ganglion Cells: ipRGC Types, Neuromodulator Contents, and Connexin Isoform. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 33410914 PMCID: PMC7804497 DOI: 10.1167/iovs.62.1.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only centrally to non-image-forming visual centers of the brain but also intraretinally to amacrine interneurons through gap junction electrical coupling, potentially modulating image-forming retinal processing. We aimed to determine (1) which ipRGC types couple with amacrine cells, (2) the neuromodulator contents of ipRGC-coupled amacrine cells, and (3) whether connexin36 (Cx36) contributes to ipRGC-amacrine coupling. Methods Gap junction-permeable Neurobiotin tracer was injected into green fluorescent protein (GFP)-labeled ipRGCs in Opn4Cre/+; Z/EG mice to stain coupled amacrine cells, and immunohistochemistry was performed to reveal the neuromodulator contents of the Neurobiotin-stained amacrine cells. We also created Opn4Cre/+; Cx36flox/flox; Z/EG mice to knock out Cx36 in GFP-labeled ipRGCs and looked for changes in the number of ipRGC-coupled amacrine cells. Results Seventy-three percent of ipRGCs, including all six types (M1-M6), were tracer-coupled with amacrine somas 5.7 to 16.5 µm in diameter but not with ganglion cells. Ninety-two percent of the ipRGC-coupled somas were in the ganglion cell layer and the rest in the inner nuclear layer. Some ipRGC-coupled amacrine cells were found to accumulate serotonin or to contain nitric oxide synthase or neuropeptide Y. Knocking out Cx36 in M2 and M4 dramatically reduced the number of coupled somas. Conclusions Heterologous gap junction coupling with amacrine cells is widespread across mouse ipRGC types. ipRGC-coupled amacrine cells probably comprise multiple morphologic types and use multiple neuromodulators, suggesting that gap junctional ipRGC-to-amacrine signaling likely exerts diverse modulatory effects on retinal physiology. ipRGC-amacrine coupling is mediated partly, but not solely, by Cx36.
Collapse
Affiliation(s)
- Krystal R. Harrison
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| | - Andrew P. Chervenak
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Sarah M. Resnick
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Aaron N. Reifler
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Kwoon Y. Wong
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
38
|
Abstract
Melanopsin retinal ganglion cells (mRGCs) are the third class of retinal photoreceptors with unique anatomical, electrophysiological, and biological features. There are different mRGC subtypes with differential projections to the brain. These cells contribute to many nonimage-forming functions of the eye, the most relevant being the photoentrainment of circadian rhythms through the projections to the suprachiasmatic nucleus of the hypothalamus. Other relevant biological functions include the regulation of the pupillary light reflex, mood, alertness, and sleep, as well as a possible role in formed vision. The relevance of the mRGC-related pathways in the brain is highlighted by the role that the dysfunction and/or loss of these cells may play in affecting circadian rhythms and sleep in many neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's disease and in aging. Moreover, the occurrence of circadian dysfunction is a known risk factor for dementia. In this chapter, the anatomy, physiology, and functions of these cells as well as their resistance to neurodegeneration in mitochondrial optic neuropathies or their predilection to be lost in other neurodegenerative disorders will be discussed.
Collapse
|
39
|
Foster RG, Hughes S, Peirson SN. Circadian Photoentrainment in Mice and Humans. BIOLOGY 2020; 9:biology9070180. [PMID: 32708259 PMCID: PMC7408241 DOI: 10.3390/biology9070180] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/26/2022]
Abstract
Light around twilight provides the primary entrainment signal for circadian rhythms. Here we review the mechanisms and responses of the mouse and human circadian systems to light. Both utilize a network of photosensitive retinal ganglion cells (pRGCs) expressing the photopigment melanopsin (OPN4). In both species action spectra and functional expression of OPN4 in vitro show that melanopsin has a λmax close to 480 nm. Anatomical findings demonstrate that there are multiple pRGC sub-types, with some evidence in mice, but little in humans, regarding their roles in regulating physiology and behavior. Studies in mice, non-human primates and humans, show that rods and cones project to and can modulate the light responses of pRGCs. Such an integration of signals enables the rods to detect dim light, the cones to detect higher light intensities and the integration of intermittent light exposure, whilst melanopsin measures bright light over extended periods of time. Although photoreceptor mechanisms are similar, sensitivity thresholds differ markedly between mice and humans. Mice can entrain to light at approximately 1 lux for a few minutes, whilst humans require light at high irradiance (>100’s lux) and of a long duration (>30 min). The basis for this difference remains unclear. As our retinal light exposure is highly dynamic, and because photoreceptor interactions are complex and difficult to model, attempts to develop evidence-based lighting to enhance human circadian entrainment are very challenging. A way forward will be to define human circadian responses to artificial and natural light in the “real world” where light intensity, duration, spectral quality, time of day, light history and age can each be assessed.
Collapse
|
40
|
Abstract
A small fraction of mammalian retinal ganglion cells are directly photoreceptive thanks to their expression of the photopigment melanopsin. These intrinsically photosensitive retinal ganglion cells (ipRGCs) have well-established roles in a variety of reflex responses to changes in ambient light intensity, including circadian photoentrainment. In this article, we review the growing evidence, obtained primarily from laboratory mice and humans, that the ability to sense light via melanopsin is also an important component of perceptual and form vision. Melanopsin photoreception has low temporal resolution, making it fundamentally biased toward detecting changes in ambient light and coarse patterns rather than fine details. Nevertheless, melanopsin can indirectly impact high-acuity vision by driving aspects of light adaptation ranging from pupil constriction to changes in visual circuit performance. Melanopsin also contributes directly to perceptions of brightness, and recent data suggest that this influences the appearance not only of overall scene brightness, but also of low-frequency patterns.
Collapse
Affiliation(s)
- Robert J Lucas
- Centre for Biological Timing and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Annette E Allen
- Centre for Biological Timing and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Nina Milosavljevic
- Centre for Biological Timing and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Riccardo Storchi
- Centre for Biological Timing and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Tom Woelders
- Centre for Biological Timing and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
41
|
Duda M, Domagalik A, Orlowska-Feuer P, Krzysztynska-Kuleta O, Beldzik E, Smyk MK, Stachurska A, Oginska H, Jeczmien-Lazur JS, Fafrowicz M, Marek T, Lewandowski MH, Sarna T. Melanopsin: From a small molecule to brain functions. Neurosci Biobehav Rev 2020; 113:190-203. [DOI: 10.1016/j.neubiorev.2020.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/29/2022]
|
42
|
Eleftheriou CG, Wright P, Allen AE, Elijah D, Martial FP, Lucas RJ. Melanopsin Driven Light Responses Across a Large Fraction of Retinal Ganglion Cells in a Dystrophic Retina. Front Neurosci 2020; 14:320. [PMID: 32317928 PMCID: PMC7147324 DOI: 10.3389/fnins.2020.00320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/18/2020] [Indexed: 02/02/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin and project to central targets, allowing them to contribute to both image-forming and non-image forming vision. Recent studies have highlighted chemical and electrical synapses between ipRGCs and neurons of the inner retina, suggesting a potential influence from the melanopsin-born signal to affect visual processing at an early stage of the visual pathway. We investigated melanopsin responses in ganglion cell layer (GCL) neurons of both intact and dystrophic mouse retinas using 256 channel multi-electrode array (MEA) recordings. A wide 200 μm inter-electrode spacing enabled a pan-retinal visualization of melanopsin's influence upon GCL activity. Upon initial stimulation of dystrophic retinas with a long, bright light pulse, over 37% of units responded with an increase in firing (a far greater fraction than can be expected from the anatomically characterized number of ipRGCs). This relatively widespread response dissipated with repeated stimulation even at a quite long inter-stimulus interval (ISI; 120 s), to leave a smaller fraction of responsive units (<10%; more in tune with the predicted number of ipRGCs). Visually intact retinas appeared to lack such widespread melanopsin responses indicating that it is a feature of dystrophy. Taken together, our data reveal the potential for anomalously widespread melanopsin responses in advanced retinal degeneration. These could be used to probe the functional reorganization of retinal circuits in degeneration and should be taken into account when using retinally degenerate mice as a model of disease.
Collapse
Affiliation(s)
- Cyril G. Eleftheriou
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, United States
| | - Phillip Wright
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Daniel Elijah
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Franck P. Martial
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Robert J. Lucas
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
43
|
Sondereker KB, Stabio ME, Renna JM. Crosstalk: The diversity of melanopsin ganglion cell types has begun to challenge the canonical divide between image-forming and non-image-forming vision. J Comp Neurol 2020; 528:2044-2067. [PMID: 32003463 DOI: 10.1002/cne.24873] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/15/2022]
Abstract
Melanopsin ganglion cells have defied convention since their discovery almost 20 years ago. In the years following, many types of these intrinsically photosensitive retinal ganglion cells (ipRGCs) have emerged. In the mouse retina, there are currently six known types (M1-M6) of melanopsin ganglion cells, each with unique morphology, mosaics, connections, physiology, projections, and functions. While melanopsin-expressing cells are usually associated with behaviors like circadian photoentrainment and the pupillary light reflex, the characterization of multiple types has demonstrated a reach that may extend far beyond non-image-forming vision. In fact, studies have shown that individual types of melanopsin ganglion cells have the potential to impact image-forming functions like contrast sensitivity and color opponency. Thus, the goal of this review is to summarize the morphological and functional aspects of the six known types of melanopsin ganglion cells in the mouse retina and to highlight their respective roles in non-image-forming and image-forming vision. Although many melanopsin ganglion cell types do project to image-forming brain targets, it is important to note that this is only the first step in determining their influence on image-forming vision. Even so, the visual system has canonically been divided into these two functional realms and melanopsin ganglion cells have begun to challenge the boundary between them, providing an overlap of visual information that is complementary rather than redundant. Further studies on these ganglion cell photoreceptors will no doubt continue to illustrate an ever-expanding role for melanopsin ganglion cells in image-forming vision.
Collapse
Affiliation(s)
| | - Maureen E Stabio
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
44
|
Cui LJ, Chen WH, Liu AL, Han X, Jiang SX, Yuan F, Zhong YM, Yang XL, Weng SJ. nGnG Amacrine Cells and Brn3b-negative M1 ipRGCs are Specifically Labeled in the ChAT-ChR2-EYFP Mouse. Invest Ophthalmol Vis Sci 2020; 61:14. [PMID: 32049344 PMCID: PMC7326507 DOI: 10.1167/iovs.61.2.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Experimental access to specific cell subtypes is essential for deciphering the complexity of retinal networks. Here, we characterized the selective labeling, caused by ectopic transgene expression, of two atypical retinal neurons in the ChAT-Channelrhodopsin-2 (ChR2)-EYFP mouse. Methods Retinal sections and flat-mounts were prepared for double-staining immunohistochemistry with antibodies against EYFP and various neuronal markers. Sagittal/coronal brain slices were made to visualize EYFP signals in central nuclei. Whole-cell recordings were conducted to test the functionality of ChR2. Results Two populations of EYFP-positive retinal cells were observed. The inner nuclear layer (INL)-located one (type I cell) distributed regularly throughout the entire retina, whereas the ganglion cell layer (GCL)-residing one (type II cell) was restricted ventrally. None of them was cholinergic, as evidenced by the complete absence of ChAT immunoreactivity. Type I cells were immunolabeled by the amacrine marker syntaxin. However, the vast majority of them were neither positive to GABA/GAD65, nor to GlyT1/glycine, suggesting that they were non-GABAergic non-glycinergic amacrine cells (nGnG ACs), which was confirmed by double-labeling with the nGnG AC marker PPP1R17. Type II cells were immunopositive to melanopsin, but not to Brn3a or Brn3b. They possessed dendrites stratifying in the outermost inner plexiform layer (IPL) and axons projecting to the suprachiasmatic nucleus (SCN) rather than the olivary pretectal nucleus (OPN), suggesting that they belonged to a Brn3b-negative subset of M1-type intrinsically photosensitive retinal ganglion cells (ipRGCs). Glutamatergic transmission-independent photocurrents were elicited in EYFP-positive cells, indicating the functional expression of ChR2. Conclusions The ChAT-ChR2-EYFP retina exhibits ectopic, but functional, transgene expression in nGnG ACs and SCN-innervating M1 ipRGCs, thus providing an ideal tool to achieve efficient labeling and optogenetic manipulation of these cells.
Collapse
|
45
|
Ahmadi H, Lund‐Andersen H, Kolko M, Bach‐Holm D, Alberti M, Ba‐Ali S. Melanopsin-mediated pupillary light reflex and sleep quality in patients with normal tension glaucoma. Acta Ophthalmol 2020; 98:65-73. [PMID: 31062491 DOI: 10.1111/aos.14133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/10/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE The intrinsically photosensitive retinal ganglion cells (ipRGCs) and sleep quality are impaired in patients with primary open-angle glaucoma (POAG). In this study, we investigated whether ipRGCs and sleep quality were also impaired in patients with normal tension glaucoma (NTG). METHODS We performed pupillometry and sleep quality assessment in 15 patients with NTG and 17 healthy age-matched controls. Pupillometry protocol consisted of monocular stimulation with high illuminance (100 lux) red (633 nm, 300 cd/m2 or 15.23 log quanta/cm2 /s) and blue light (463 nm, 332 cd/m2 or 15.27 log quanta/cm2 /s) and binocular pupil measurements. Prior to light stimulation, patients were dark-adapted for 5 min. The late postillumination pupillary response (PIPRL ate ) to blue light was used as marker of ipRGC activity. Sleep quality was assessed by Pittsburgh Sleep Quality Index (PSQI) questionnaire. RESULTS The PIPRL ate to blue light was significantly reduced in patients with NTG compared to healthy subjects (p < 0.001), indicating impairment of the melanopsin-mediated pupillary pathway. There was no significant difference in the response elicited by red light (p = 0.6). Baseline pupil diameter and pupillary constriction amplitude to both red and blue light were reduced in patients with NTG (p < 0.05). The global score in PSQI was not significantly different between healthy controls and patients with NTG, indicating normal sleep quality (p = 0.6). Furthermore, we found no correlation between sleep parameters and pupillary light reflex parameters. CONCLUSION Patients with NTG exhibited reduced ipRGC activity compared to healthy subjects, while no differences were observed in sleep quality.
Collapse
Affiliation(s)
- Hamid Ahmadi
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Henrik Lund‐Andersen
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Miriam Kolko
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Daniella Bach‐Holm
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Mark Alberti
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Shakoor Ba‐Ali
- Department of Ophthalmology Rigshospitalet Glostrup Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
46
|
Lee SK, Sonoda T, Schmidt TM. M1 Intrinsically Photosensitive Retinal Ganglion Cells Integrate Rod and Melanopsin Inputs to Signal in Low Light. Cell Rep 2019; 29:3349-3355.e2. [PMID: 31825819 PMCID: PMC6951432 DOI: 10.1016/j.celrep.2019.11.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/04/2019] [Accepted: 11/06/2019] [Indexed: 02/01/2023] Open
Abstract
Light influences various behaviors and physiological processes that occur outside of our conscious perception, including circadian photoentrainment, sleep, and even learning and mood. The M1, melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) relay a combination of rod/cone and melanopsin signals to drive these functions. However, little is known about how M1 ipRGCs integrate these signals in low light. We measure the dim light response of M1 ipRGCs and find that they exhibit a wide spectrum of responses to dim, scotopic light stimulation that are driven by a combination of rod pathway input and melanopsin phototransduction. The presence of rod input to M1 ipRGCs correlates with larger and more complex dendritic arbors. Collectively, these results show variability in the rod input to M1 ipRGCs and a surprising contribution of melanopsin to the light responses of M1 ipRGCs at very low light.
Collapse
Affiliation(s)
- Seul Ki Lee
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Takuma Sonoda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
47
|
Caval-Holme F, Zhang Y, Feller MB. Gap Junction Coupling Shapes the Encoding of Light in the Developing Retina. Curr Biol 2019; 29:4024-4035.e5. [PMID: 31708397 PMCID: PMC6927338 DOI: 10.1016/j.cub.2019.10.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/03/2019] [Accepted: 10/16/2019] [Indexed: 11/28/2022]
Abstract
Detection of ambient illumination in the developing retina prior to maturation of conventional photoreceptors is mediated by intrinsically photosensitive retinal ganglion cells (ipRGCs) and is critical for driving several physiological processes, including light aversion, pupillary light reflexes, and photoentrainment of circadian rhythms. The strategies by which ipRGCs encode variations in ambient light intensity at these early ages are not known. Using unsupervised clustering of two-photon calcium responses followed by inspection of anatomical features, we found that the population activity of the neonatal retina could be modeled as six functional groups that were composed of mixtures of ipRGC subtypes and non-ipRGC cell types. By combining imaging, whole-cell recording, pharmacology, and anatomical techniques, we found that functional mixing of cell types is mediated in part by gap junction coupling. Together, these data show that both cell-autonomous intrinsic light responses and gap junction coupling among ipRGCs contribute to the proper encoding of light intensity in the developing retina.
Collapse
Affiliation(s)
- Franklin Caval-Holme
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yizhen Zhang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
48
|
Do MTH. Melanopsin and the Intrinsically Photosensitive Retinal Ganglion Cells: Biophysics to Behavior. Neuron 2019; 104:205-226. [PMID: 31647894 PMCID: PMC6944442 DOI: 10.1016/j.neuron.2019.07.016] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022]
Abstract
The mammalian visual system encodes information over a remarkable breadth of spatiotemporal scales and light intensities. This performance originates with its complement of photoreceptors: the classic rods and cones, as well as the intrinsically photosensitive retinal ganglion cells (ipRGCs). IpRGCs capture light with a G-protein-coupled receptor called melanopsin, depolarize like photoreceptors of invertebrates such as Drosophila, discharge electrical spikes, and innervate dozens of brain areas to influence physiology, behavior, perception, and mood. Several visual responses rely on melanopsin to be sustained and maximal. Some require ipRGCs to occur at all. IpRGCs fulfill their roles using mechanisms that include an unusual conformation of the melanopsin protein, an extraordinarily slow phototransduction cascade, divisions of labor even among cells of a morphological type, and unorthodox configurations of circuitry. The study of ipRGCs has yielded insight into general topics that include photoreceptor evolution, cellular diversity, and the steps from biophysical mechanisms to behavior.
Collapse
Affiliation(s)
- Michael Tri H Do
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Center for Life Science 12061, 3 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Wu XS, Wang YC, Liu TT, Wang L, Sun XH, Wang LQ, Weng SJ, Zhong YM. Morphological alterations of intrinsically photosensitive retinal ganglion cells after ablation of mouse photoreceptors with selective photocoagulation. Exp Eye Res 2019; 188:107812. [PMID: 31550445 DOI: 10.1016/j.exer.2019.107812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/06/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
In this work, we investigated changes in the morphology of intrinsically photosensitive retinal ganglion cells (ipRGCs), M1 subtype, and pupillary light reflex following local and selective ablation of photoreceptors in mice. Laser photocoagulation was used to selectively destroy four patches of photoreceptors per eye at around 4 papillary diameters from the optic disc and at the 3, 6, 9, and 12 o'clock positions between the retinal vessels in the adult mouse retina, leaving cells in the inner retina intact. Morphological parameters of individual M1 cells specifically labeled by the antibody against melanopsin (PA1-780), including dendritic field size, total dendritic length, and dendritic branch number, were examined 1, 2, 4, and 8 weeks after photocoagulation with Neurolucida software. A considerable reduction in these parameters in M1 cells in the "lesioned areas" was found at all the four time points after photocoagulation, as compared with those in the "unlesioned areas". Although M1 cells in the lesioned areas showed significant changes as early as 1 week after laser treatment and the changes gradually increased, reaching a peak value at 2 weeks, morphological restoration was clearly seen in these cells over time. However, no difference in the morphological parameters of M1 cells was observed between the unlesioned areas of laser-treated mice and the corresponding areas of age-matched normal mice without laser lesions. Fluorescence intensity of the somata of melanopsin-positive M1 cells located inside the lesioned areas was significantly decreased at all the four time points after photocoagulation, whereas no changes in pupillary light reflex were detected at different light irradiations, indicating that photocoagulation-induced local photoreceptor loss and alterations of ipRGCs may be insufficient to cause abnormalities in non-image-forming (NIF) visual functions. The results suggest that intact photoreceptors could be crucial for maintaining the expression levels of melanopsin and normal morphology of M1 cells.
Collapse
Affiliation(s)
- Xiao-Sha Wu
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Yong-Chen Wang
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Ting-Ting Liu
- Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai, 200031, PR China
| | - Lu Wang
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Xing-Huai Sun
- Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai, 200031, PR China
| | - Li-Qin Wang
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Shi-Jun Weng
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| | - Yong-Mei Zhong
- Department of Ophthalmology, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science and Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
50
|
Bhandari A, Smith JC, Zhang Y, Jensen AA, Reid L, Goeser T, Fan S, Ghate D, Van Hook MJ. Early-Stage Ocular Hypertension Alters Retinal Ganglion Cell Synaptic Transmission in the Visual Thalamus. Front Cell Neurosci 2019; 13:426. [PMID: 31607867 PMCID: PMC6761307 DOI: 10.3389/fncel.2019.00426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
Axonopathy is a hallmark of many neurodegenerative diseases including glaucoma, where elevated intraocular pressure (ocular hypertension, OHT) stresses retinal ganglion cell (RGC) axons as they exit the eye and form the optic nerve. OHT causes early changes in the optic nerve such as axon atrophy, transport inhibition, and gliosis. Importantly, many of these changes appear to occur prior to irreversible neuronal loss, making them promising points for early diagnosis of glaucoma. It is unknown whether OHT has similarly early effects on the function of RGC output to the brain. To test this possibility, we elevated eye pressure in mice by anterior chamber injection of polystyrene microbeads. Five weeks post-injection, bead-injected eyes showed a modest RGC loss in the peripheral retina, as evidenced by RBPMS antibody staining. Additionally, we observed reduced dendritic complexity and lower spontaneous spike rate of On-αRGCs, targeted for patch clamp recording and dye filling using a Opn4-Cre reporter mouse line. To determine the influence of OHT on retinal projections to the brain, we expressed Channelrhodopsin-2 (ChR2) in melanopsin-expressing RGCs by crossing the Opn4-Cre mouse line with a ChR2-reporter mouse line and recorded post-synaptic responses in thalamocortical relay neurons in the dorsal lateral geniculate nucleus (dLGN) of the thalamus evoked by stimulation with 460 nm light. The use of a Opn4-Cre reporter system allowed for expression of ChR2 in a narrow subset of RGCs responsible for image-forming vision in mice. Five weeks following OHT induction, paired pulse and high-frequency stimulus train experiments revealed that presynaptic vesicle release probability at retinogeniculate synapses was elevated. Additionally, miniature synaptic current frequency was slightly reduced in brain slices from OHT mice and proximal dendrites of post-synaptic dLGN relay neurons, assessed using a Sholl analysis, showed a reduced complexity. Strikingly, these changes occurred prior to major loss of RGCs labeled with the Opn4-Cre mouse, as indicated by immunofluorescence staining of ChR2-expressing retinal neurons. Thus, OHT leads to pre- and post-synaptic functional and structural changes at retinogeniculate synapses. Along with RGC dendritic remodeling and optic nerve transport changes, these retinogeniculate synaptic changes are among the earliest signs of glaucoma.
Collapse
Affiliation(s)
- Ashish Bhandari
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jennie C Smith
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yang Zhang
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States.,Creighton University School of Medicine, Omaha, NE, United States.,Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Aaron A Jensen
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lisa Reid
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Toni Goeser
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shan Fan
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Deepta Ghate
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|