1
|
Santana NNM, da Silva MMO, Silva EHA, dos Santos SF, Bezerra LL, Escarião WKM, Vasiljevic GAM, Fiuza FP, Cavalcante JS, Engelberth RC. Neuronal number and somal volume in calbindin-expressing neurons of the marmoset dorsal lateral geniculate nucleus are preserved during aging. PLoS One 2025; 20:e0323906. [PMID: 40408448 PMCID: PMC12101782 DOI: 10.1371/journal.pone.0323906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/15/2025] [Indexed: 05/25/2025] Open
Abstract
Compelling evidence links age-related brain dysfunction and neurodegenerative processes to persistent disruptions in intracellular calcium (Ca2+) signaling, a central hypothesis in the Ca2+ theory of aging. Calbindin (CB), a classical Ca2+ buffer, has been implicated in region-specific susceptibility to aging-related effects. Specifically, CB-immunopositive (CB+) neurons have demonstrated an age-dependent decline in neuronal number across various cortical and subcortical regions. However, it remains unclear whether this decrease occur in the dorsal lateral geniculate nucleus (DLG), a crucial relay and modulatory center for visual processing. Additionally, the potential impact of aging on the cellular volume of CB+ neurons in the DLG has not been fully elucidated, albeit an age-dependent neuronal hypertrophy of this region has been reported. To address these questions, we investigated CB+ neurons in the DLG of six marmosets (Callithrix jacchus), aged between 29-143 months. Using design-based stereological techniques, we estimated the total number and somal volume of CB+ neurons in DLG layers. Our results revealed no signs of CB+ neuronal number loss and somal volumetric changes in aged DLG, particularly within the koniocellular layers, a stratum that primarily expresses CB and play a critical role in blue/yellow color vision. Altogether, our findings suggest a preserved neuronal number and cellular volume of the CB+ population during aging process in the marmoset DLG. Moreover, they provide a valuable basis for future investigations into the neuroprotective role of CB in visual processing during aging and open avenues for strategies designed to preserve vulnerable neuronal populations in age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Nelyane N. M. Santana
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Maria M. O. da Silva
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eryck H. A. Silva
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sâmarah F. dos Santos
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Lyzandro L.F. Bezerra
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Wellydo K. M. Escarião
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Gabriel A. M. Vasiljevic
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Felipe P. Fiuza
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Jeferson S. Cavalcante
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Rovena Clara Engelberth
- Department of Physiology and Behavior, Laboratory of Neurochemical Studies, Bioscience Center, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
2
|
Guder P, Scheungrab M, Kohnert P, Kolyfetis G, Wanner G, Heß M. Horizontal cell connectivity in the anchovy retina-a 3D electron microscopic study. BMC Biol 2025; 23:137. [PMID: 40390032 PMCID: PMC12090589 DOI: 10.1186/s12915-025-02242-7] [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: 11/15/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Block-face scanning electron microscopy has opened a new era of connectomics research, in which it is possible to make dense reconstructions of all cells in a clipping of a neuronal network, such as the retina, resolving synaptic contacts. Anchovies, exceptionally abundant marine teleosts, have retinae with regions for triple cone-based color vision and a region with specialized cone photoreceptors, so-called polycones, made of long and short cones with axially oriented outer segment lamellae for polarization contrast vision. This modality, discovered in the 1970s, is unique in vertebrates, but the neural wiring for contrast generation in deeper retinal layers is unknown so far. RESULTS To elucidate the retinal connectomics of the European anchovy Engraulis encrasicolus (Linnaeus, 1758), in a first project, we investigated the shapes and cone-specific wiring rules of 3 horizontal cell types using volume electron microscopy and subsequent computer-aided reconstruction: H1 cells contact both cone types of the polycone, H2 cells contact only the short cones, and H3 cells are exclusively connected to rods. In addition, a distinctive double band of Müller fibers and a layer of H1 axon terminals were structurally clarified. CONCLUSIONS The findings suggest that (1) the monochromatic polarization contrast system based on fine structure specializations in the outer retina is connected to an inherited (bichromatic) color contrast mechanism in the inner retina, (2) the anchovy polycones arose from red (now long) and green (now short) cones, and (3) the blue single cones disappeared in the relevant retinal region.
Collapse
Affiliation(s)
- Petra Guder
- Staatliches Museum Für Naturkunde Karlsruhe, Erbprinzenstraße 13, Karlsruhe, 76133, Germany
| | - Max Scheungrab
- Biozentrum LMU München, Großhaderner Straße 2, Planegg-Martinsried, 82152, Germany
| | - Peter Kohnert
- Zoologische Staatssammlung München, Münchhausenstraße 21, Munich, 81247, Germany
| | - Georgios Kolyfetis
- Neurobiology University Konstanz, Universitätsstraße 10, Konstanz, 78464, Germany
| | - Gerhard Wanner
- Biozentrum LMU München, Großhaderner Straße 2, Planegg-Martinsried, 82152, Germany
| | - Martin Heß
- Biozentrum LMU München, Großhaderner Straße 2, Planegg-Martinsried, 82152, Germany.
- GeoBio Center LMU, Richard Wagner Straße 10, Munich, 80333, Germany.
| |
Collapse
|
3
|
Komatsu H, Hosoda N, Ikuno T. Polarity-tunable dye-sensitized optoelectronic artificial synapses for physical reservoir computing-based machine vision. Sci Rep 2025; 15:16488. [PMID: 40355457 PMCID: PMC12069640 DOI: 10.1038/s41598-025-00693-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
Conventional machine vision systems process huge time-series data per second, presenting significant challenges for edge-device applications due to limitations in data transfer and storage. Inspired by the human visual system, artificial optoelectronic synapses replicating synaptic responses have emerged as promising solutions. However, achieving color recognition comparable to human vision remains challenging. Moreover, most optoelectronic artificial synapses rely on photocurrent-based operation, producing low current values and necessitating external circuits. This study reports a self-powered optoelectronic artificial synapse capable of distinguishing wavelengths with a resolution of 10 nm by integrating dye-sensitized solar cells. The device exhibits synaptic responses to light pulses and bipolar responses when exposed to different wavelengths. The wavelength-dependent bipolar behavior enables exceptional separation capabilities, achieving six-bit resolution with 64 distinct states and supporting multiple logic operations, including AND, OR, and XOR, within a single device. Additionally, the device leverages distinct responses to red, green, and blue light irradiation for physical reservoir computing, facilitating the classification of color-coded human motion with an accuracy of 82%. These findings advance the development of optoelectronic artificial synapses for precise, human-eye-like color discrimination.
Collapse
Affiliation(s)
- Hiroaki Komatsu
- Department of Applied Electronics, Graduate School of Advanced Engineering, Tokyo University of Science, Katsushika, Tokyo, 125-8585, Japan
| | - Norika Hosoda
- Department of Applied Electronics, Graduate School of Advanced Engineering, Tokyo University of Science, Katsushika, Tokyo, 125-8585, Japan
| | - Takashi Ikuno
- Department of Applied Electronics, Graduate School of Advanced Engineering, Tokyo University of Science, Katsushika, Tokyo, 125-8585, Japan.
| |
Collapse
|
4
|
Blanco-Formoso M, Galluzzi F, Vacca F, Gianiorio T, Piergentili I, Cook AB, Welzen PLW, van Hest JCM, Di Marco S, Tantussi F, Benfenati F, Colombo E, De Angelis F. Spiropyran-based glutamate nanovalve for neuronal stimulation. MATERIALS HORIZONS 2025. [PMID: 40314589 DOI: 10.1039/d5mh00082c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
One of the main challenges in medical applications is achieving precise spatial and temporal control over the release of active molecules, such as neurotransmitters. To address this issue, we engineered a nanovalve that can deliver active molecules on demand by activating or deactivating a light-sensitive chemical barrier. This valve is composed of a polymer containing a spiropyran moiety, which can switch from a hydrophobic to a hydrophilic state upon photo-stimulation. Accordingly, the nanovalve either blocks or allows molecular diffusion through a solid-state nanopore array. Here, we demonstrate that the system blocks up to 96% of the translocation of the neurotransmitter glutamate and that the on-demand release of glutamate upon light stimulation reaches 60 μM h-1, mimicking a physiological synaptic release rate. We proved its cytocompatibility and analyzed its potential for the stimulation of primary neurons and blind retinal explants by patch-clamp experiments. These results represent a milestone for the development of biomimetic neuroprostheses restoring chemical synaptic transmission lost by degeneration or delivering drugs in a light-controlled fashion.
Collapse
Affiliation(s)
- M Blanco-Formoso
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
- CINBIO Universidade de Vigo, Vigo, Spain
| | - F Galluzzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Genova, Italy
| | - F Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - T Gianiorio
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- Department of Neuroscience (DINOGMI), University of Genoa, Genova, Italy
| | - I Piergentili
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - A B Cook
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - P L W Welzen
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - J C M van Hest
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - S Di Marco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F Tantussi
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| | - F Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - E Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F De Angelis
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
5
|
Ghosh M, Palaniswamy HP, G N, Joylin S, T.S S, Sanjana M, Radhika R, Theruveethi N. Impact of domestic white LED light on cognitive functions and amelioration of blue light blocking lens (BBL) on healthy adults. Neurobiol Sleep Circadian Rhythms 2025; 18:100119. [PMID: 40342909 PMCID: PMC12060505 DOI: 10.1016/j.nbscr.2025.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
White light-emitting diodes (WLEDs) can affect cognition and working memory. Blue light-blocking lenses (BBL) may help alleviate this. We aim to study the relationship between WLED and the ameliorative effect of BBL. We included 15 healthy participants based on the PSQI and Mini-Cog™ screening. The eligible participants underwent a baseline recording of event-related potential (ERP) of P300 using electroencephalography (EEG) while performing a 2-back task, followed by exposure to WLED (600 lux) that was given (45° with 80 cm apart from the participant's eye plane) for 30 min. A similar protocol was maintained when BBL was worn with WLED exposure. The participants' mean PSQI and Mini-Cog™ scores (n = 15) were 3 and 5, respectively. The behavioral functioning of participants using a 2-back task revealed enhancement in working memory cognition by fastening the response time (ms) from base to post-WLED to post-WLED + BBL (p < 0.001). Still, no significant difference (p > 0.05) in accuracy (%) was observed. The learning effect in the control group using a 2-back task revealed no statistically significant difference (p > 0.05) in both accuracy (%) and response time (ms). Additionally, no significant change (p > 0.05) was found within the three light groups in latency (ms) and amplitude (μV) at the P300 region of ERP in the prefrontal cortex. The existing results found that domestic WLED exposure significantly leads to a faster response time in working memory performance in the prefrontal cortex, thus remaining alert. BBL is not protective in the nonvisual senses when exposed to WLED for 30 min.
Collapse
Affiliation(s)
- Mousumi Ghosh
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
- Department of Optometry, Dr B.C.Roy Academy of Professional Courses, Durgapur, West Bengal, India
| | - Hari Prakash Palaniswamy
- Department of Speech and Hearing, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nishitha. G
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Stelyna Joylin
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
- Department of Optometry, Srinivas Institute of Allied Health Sciences, Srinivas University, Mangalore, India
| | - Shwetha. T.S
- Department of Clinical Psychology, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - M. Sanjana
- Department of Speech and Hearing, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - R.P. Radhika
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nagarajan Theruveethi
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, India
| |
Collapse
|
6
|
Soto F, Lin CI, Jo A, Chou SY, Harding EG, Ruzycki PA, Seabold GK, Petralia RS, Kerschensteiner D. Molecular mechanism establishing the OFF pathway in vision. Nat Commun 2025; 16:3708. [PMID: 40251167 PMCID: PMC12008213 DOI: 10.1038/s41467-025-59046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
Parallel ON and OFF (positive- and negative-contrast) pathways fundamental to vision arise at the complex synapse of cone photoreceptors. Cone pedicles form spatially segregated functionally opposite connections with ON and OFF bipolar cells. Here, we discover that mammalian cones express LRFN2, a cell-adhesion molecule, which localizes to the pedicle base. LRFN2 stabilizes basal contacts between cone pedicles and OFF bipolar cell dendrites to guide pathway-specific partner choices, encompassing multiple cell types. In addition, LRFN2 trans-synaptically organizes glutamate receptor clusters, determining the contrast preferences of the OFF pathway. ON and OFF pathways converge in the inner retina to regulate bipolar cell outputs. We analyze LRFN2's contributions to ON-OFF interactions, pathway asymmetries, and neural and behavioral responses to approaching predators. Our results reveal that LRFN2 controls the formation of the OFF pathway in vision, supports parallel processing in a single synapse, and shapes contrast coding and the detection of visual threats.
Collapse
Affiliation(s)
- Florentina Soto
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| | - Chin-I Lin
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Graduate Program in Neuroscience, Division of Biological & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew Jo
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Ssu-Yu Chou
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Ellen G Harding
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip A Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Gail K Seabold
- Laboratory of Neurochemistry, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Ronald S Petralia
- Laboratory of Neurochemistry, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA.
- Bright Center for Human Vision, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Hoon M. Neural circuit assembly: A sticky cue that connects neurons with a preference for hue. Curr Biol 2025; 35:R255-R258. [PMID: 40199249 DOI: 10.1016/j.cub.2025.02.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Neurons rely on molecular interactions typically mediated by transmembrane adhesion proteins to locate appropriate partners and establish connections. A recent study finds that a member of the cadherin family of cell adhesion proteins organizes color-preferring connections in the part of the retinal neural circuit designated for encoding light decrements.
Collapse
Affiliation(s)
- Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
8
|
Zhang Y, Lin S, Yu L, Lin X, Qu S, Ye Q, Yu M, Chen W, Wu W. Gene therapy shines light on congenital stationary night blindness for future cures. J Transl Med 2025; 23:392. [PMID: 40181393 PMCID: PMC11969737 DOI: 10.1186/s12967-025-06392-8] [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: 11/13/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
Congenital Stationary Night Blindness (CSNB) is a non-progressive hereditary eye disease that primarily affects the retinal signal processing, resulting in significantly reduced vision under low-light conditions. CSNB encompasses various subtypes, each with distinct genetic patterns and pathogenic genes. Over the past few decades, gene therapy for retinal genetic disorders has made substantial progress; however, effective clinical therapies for CSNB are yet to be discovered. With the continuous advancement of gene-therapy tools, there is potential for these methods to become effective treatments for CSNB. Nonetheless, challenges remain in the treatment of CSNB, including issues related to delivery vectors, therapeutic efficacy, and possible side effects. This article reviews the clinical diagnosis, pathogenesis, and associated mutated genes of CSNB, discusses existing animal models, and explores the application of gene therapy technologies in retinal genetic disorders, as well as the current state of research on gene therapy for CSNB.
Collapse
Affiliation(s)
- Yi Zhang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Siqi Lin
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Lingqi Yu
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Xiang Lin
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Shuai Qu
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Qingyang Ye
- Hangzhou Bipolar Biotechnology Co., Ltd., Hangzhou, 311199, China
| | - Mengting Yu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350028, China
| | - Wenfeng Chen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
| | - Wenjie Wu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350028, China.
| |
Collapse
|
9
|
Kim DE, Kim S, Kim M, Min BK, Im M. Retinal degeneration increases inter-trial variabilities of light-evoked spiking activities in ganglion cells. Exp Eye Res 2025; 253:110305. [PMID: 39983973 DOI: 10.1016/j.exer.2025.110305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information to the brain in the form of spike trains, which form visual perception. The reliabilities of spike timing and count are thought to play a crucial role in generating stable percepts. However, the effect of retinal degeneration on spike reproducibility remains underexplored. In this study, we examined longitudinal changes of both spike timing and count across different RGC types in response to repeated presentations of an identical light stimulus in retinal degeneration 10 (rd10) mice (B6.CXBl-Pde6brd10/J), a well-established model of retinitis pigmentosa (RP). We recorded the spiking responses of RGC populations to repeated white flashes using 256-channel multi-electrode array (MEA) at four rd10 age groups representing various stages of retinal degeneration. Our experimental results revealed a significant reduction in both spike timing and count consistencies compared to those in wild-type RGC recordings. Furthermore, the inter-trial variability patterns of different RGC types were found to differ throughout the degeneration process. For instance, when the spike time tiling coefficient (STTC) was used to evaluate inter-trial spike timing consistency, contrast-sensitive RGCs (ON, OFF, and ON-OFF types) exhibited a systematic decrease in spike timing consistency as degeneration progressed, whereas the remaining units did not show similar trends. Thus, we concluded that light-evoked spike trains become less consistent as degeneration progresses, with variability in spike timing and spike count varying across cell types. Given the critical role of spiking reliability in visual perception, our findings highlight the importance of accounting for cell type-specific degeneration patterns and inter-trial spiking inconsistencies when developing visual rehabilitation therapies to achieve enhanced performance. The underlying mechanism(s) driving the inter-trial spiking inconsistencies warrant further investigation.
Collapse
Affiliation(s)
- Da Eun Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Sein Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea
| | - Minju Kim
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea
| | - Byoung-Kyong Min
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, KIST (Korea Institute of Science and Technology), Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science & Technology, Seoul, Republic of Korea; KHU-KIST Department of Converging Science & Technology, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Laniado DD, Maron Y, Gemmer JA, Sabbah S. A spherical code of retinal orientation selectivity enables decoding in ensembled and retinotopic operation. Cell Rep 2025; 44:115373. [PMID: 40023844 DOI: 10.1016/j.celrep.2025.115373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/04/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
Selectivity to orientations of edges is seen at the earliest stages of visual processing in retinal orientation-selective ganglion cells (OSGCs), which are thought to prefer vertical or horizontal orientation. However, because stationary edges are projected on the hemispherical retina as lines of longitude or latitude, how edge orientation is encoded and decoded by the brain is unknown. Here, by mapping the orientation selectivity (OS) of thousands of OSGCs at known retinal locations in mice, we identify three OSGC types whose preferences match two longitudinal fields and a fourth type matching two latitudinal fields, with the members of each field pair being non-orthogonal. A geometric decoder reveals that two OS sensors yield optimal orientation decoding when approaching the deviation from orthogonality we observe for OSGC field pairs. Retinotopically organized decoding generates type-specific variation in decoding efficiency across the visual field. OS tuning is greater in the dorsal retina, possibly reflecting an evolutionary adaptation to an environmental gradient of edges.
Collapse
Affiliation(s)
- Dimitrios D Laniado
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Yariv Maron
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - John A Gemmer
- Department of Mathematics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Shai Sabbah
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel.
| |
Collapse
|
11
|
Leitão Guerra RL, Leitão Guerra CL, Meirelles MGB, Barbosa GCS, Novais EA, Badaró E, Lucatto LFA, Roisman L. Exploring retinal conditions through blue light reflectance imaging. Prog Retin Eye Res 2025; 105:101326. [PMID: 39756669 DOI: 10.1016/j.preteyeres.2024.101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Blue light reflectance (BLR) imaging offers a non-invasive, cost-effective method for evaluating retinal structures by analyzing the reflectance and absorption characteristics of the inner retinal layers. By leveraging blue light's interaction with retinal tissues, BLR enhances visualization beyond the retinal nerve fiber layer, improving detection of structures such as the outer plexiform layer and macular pigment. Its diagnostic utility has been demonstrated in distinct retinal conditions, including hyperreflectance in early macular telangiectasia, hyporeflectance in non-perfused areas indicative of ischemia, identification of pseudodrusen patterns (notably the ribbon type), and detection of peripheral retinal tears and degenerative retinoschisis in eyes with reduced retinal pigment epithelial pigmentation. Best practices for image acquisition and interpretation are discussed, emphasizing standardization to minimize variability. Common artifacts and mitigation strategies are also addressed, ensuring image reliability. BLR's clinical utility, limitations, and future research directions are highlighted, particularly its potential in automated image analysis and quantitative assessment. Different BLR acquisition methods, such as fundus photography, confocal scanning laser ophthalmoscopy, and broad line fundus imaging, are evaluated for their respective advantages and limitations. As research advances, BLR's integration into multimodal workflows is expected to improve early detection and precise monitoring of retinal diseases.
Collapse
Affiliation(s)
- Ricardo Luz Leitão Guerra
- Department of Ophthalmology Leitão Guerra - Oftalmologia (Salvador, Brazil), Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil; Orbit Ophthalmo Learning, Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Cezar Luz Leitão Guerra
- Department of Ophthalmology Leitão Guerra - Oftalmologia (Salvador, Brazil), Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Mariana Gouveia Bastos Meirelles
- Department of Ophthalmology Leitão Guerra - Oftalmologia (Salvador, Brazil), Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Gabriel Castilho Sandoval Barbosa
- Department of Ophthalmology, University of São Paulo, Av. Dr. Arnaldo, 455 - Cerqueira César, CEP: 01.246-903, São Paulo, (SP), Brazil
| | - Eduardo Amorim Novais
- Orbit Ophthalmo Learning, Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Emmerson Badaró
- Orbit Ophthalmo Learning, Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Luiz Filipe Adami Lucatto
- Orbit Ophthalmo Learning, Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| | - Luiz Roisman
- Orbit Ophthalmo Learning, Rua Rio de São Pedro, no 256 Graça, CEP 40.150-350, Salvador, (BA), Brazil
| |
Collapse
|
12
|
Brais-Brunet S, Boudoux C, Dehaes M. Morphological characterization of retinal development from birth to adulthood via retinal thickness assessment in mice: A systematic review. Exp Eye Res 2025; 251:110229. [PMID: 39755351 DOI: 10.1016/j.exer.2024.110229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
The morphology and thickness of the retinal layers are valuable biomarkers for retinal health and development. The retinal layers in mice are similar to those in humans; thus, a mouse is appropriate for studying the retina. The objectives of this systematic review were: (1) to describe normal retinal morphology quantitatively using retinal layer thickness measured from birth to age 6 months in healthy mice; and (2) to describe morphological changes in physiological retinal development over time using the longitudinal (in vivo) and cross-sectional (ex vivo) data from the included studies. A PubMed search was conducted for articles published from to 1980-2024 that included quantitative data. Prior to sexual maturity, an increase in the total retinal and inner plexiform layer thicknesses were observed, with a decrease in the inner nuclear layer thickness. After sexual maturity, an asymptotic decrease in thickness was observed up to age 6 months in all layers; during this period, no significant changes were observed in the outer nuclear layer or nerve fiber layer/ganglion cell layer complex. Potential sources of variability and inconsistency among the studies included differences in imaging modality, animal strain, measurement timing, and retinal segmentation/assignment techniques. These findings highlight the importance of including a control group in experimental designs and providing comparative data for further investigations.
Collapse
Affiliation(s)
- Simon Brais-Brunet
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada; Research Center, CHU Sainte-Justine University Hospital Centre, Montréal, Canada
| | - Caroline Boudoux
- Research Center, CHU Sainte-Justine University Hospital Centre, Montréal, Canada; Department of Engineering Physics, Polytechnique Montréal, Montréal, Canada
| | - Mathieu Dehaes
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada; Research Center, CHU Sainte-Justine University Hospital Centre, Montréal, Canada; Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montréal, Montréal, Canada.
| |
Collapse
|
13
|
Zheng J, Meister M. The unbearable slowness of being: Why do we live at 10 bits/s? Neuron 2025; 113:192-204. [PMID: 39694032 PMCID: PMC11758279 DOI: 10.1016/j.neuron.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024]
Abstract
This article is about the neural conundrum behind the slowness of human behavior. The information throughput of a human being is about 10 bits/s. In comparison, our sensory systems gather data at ∼109 bits/s. The stark contrast between these numbers remains unexplained and touches on fundamental aspects of brain function: what neural substrate sets this speed limit on the pace of our existence? Why does the brain need billions of neurons to process 10 bits/s? Why can we only think about one thing at a time? The brain seems to operate in two distinct modes: the "outer" brain handles fast high-dimensional sensory and motor signals, whereas the "inner" brain processes the reduced few bits needed to control behavior. Plausible explanations exist for the large neuron numbers in the outer brain, but not for the inner brain, and we propose new research directions to remedy this.
Collapse
Affiliation(s)
- Jieyu Zheng
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Markus Meister
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
14
|
Ziraldo G, Cupini S, Sesti V, Delfino E, Lanzani G, Bertarelli C, Benfenati F, Di Marco S. A membrane-targeted photoswitch restores physiological ON/OFF responses to light in the degenerate retina. Nat Commun 2025; 16:600. [PMID: 39799138 PMCID: PMC11724966 DOI: 10.1038/s41467-025-55882-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
The lack of effective therapies for visual restoration in Retinitis pigmentosa and macular degeneration has led to the development of new strategies, such as optogenetics and retinal prostheses. However, visual restoration is poor due to the massive light-evoked activation of retinal neurons, regardless of the segregation of visual information in ON and OFF channels, which is essential for contrast sensitivity and spatial resolution. Here, we show that Ziapin2, a membrane photoswitch that modulates neuronal capacitance and excitability in a light-dependent manner, is capable of reinstating, in mouse and rat genetic models of photoreceptor degeneration, brisk and sluggish ON, OFF, and ON-OFF responses in retinal ganglion cells evoked by full-field stimuli, with reactivation of their excitatory and inhibitory conductances. Intravitreally injected Ziapin2 in fully blind rd10 mice restores light-driven behavior and optomotor reflexes. The results indicate that Ziapin2 is a promising molecule for reinstating physiological visual responses in the late stages of retinal degeneration.
Collapse
Affiliation(s)
- Gaia Ziraldo
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genova, Italy
| | - Sara Cupini
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Valentina Sesti
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta", Politecnico di Milano, Milano, Italy
- Center for Nanoscience and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Emanuela Delfino
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Guglielmo Lanzani
- Center for Nanoscience and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Chiara Bertarelli
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta", Politecnico di Milano, Milano, Italy
- Center for Nanoscience and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Stefano Di Marco
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
15
|
Gonschorek D, Goldin MA, Oesterle J, Schwerd-Kleine T, Arlinghaus R, Zhao Z, Schubert T, Marre O, Euler T. Nitric oxide modulates contrast suppression in a subset of mouse retinal ganglion cells. eLife 2025; 13:RP98742. [PMID: 39783858 PMCID: PMC11717361 DOI: 10.7554/elife.98742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Neuromodulators have major influences on the regulation of neural circuit activity across the nervous system. Nitric oxide (NO) has been shown to be a prominent neuromodulator in many circuits and has been extensively studied in the retina. Here, it has been associated with the regulation of light adaptation, gain control, and gap junctional coupling, but its effect on the retinal output, specifically on the different types of retinal ganglion cells (RGCs), is still poorly understood. In this study, we used two-photon Ca2+ imaging and multi-electrode array (MEA) recordings to measure light-evoked activity of RGCs in the ganglion cell layer in the ex vivo mouse retina. This approach allowed us to investigate the neuromodulatory effects of NO on a cell type-level. Our findings reveal that NO selectively modulates the suppression of temporal responses in a distinct subset of contrast-suppressed RGC types, increasing their activity without altering the spatial properties of their receptive fields. Given that under photopic conditions, NO release is triggered by quick changes in light levels, we propose that these RGC types signal fast contrast changes to higher visual regions. Remarkably, we found that about one-third of the RGC types, recorded using two-photon Ca2+ imaging, exhibited consistent, cell type-specific adaptational response changes throughout an experiment, independent of NO. By employing a sequential-recording paradigm, we could disentangle those additional adaptational response changes from drug-induced modulations. Taken together, our research highlights the selective neuromodulatory effects of NO on RGCs and emphasizes the need of considering non-pharmacological activity changes, like adaptation, in such study designs.
Collapse
Affiliation(s)
- Dominic Gonschorek
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Matías A Goldin
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Jonathan Oesterle
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Hertie Institute for AI in Brain Health, Tübingen AI Center, University of TübingenTübingenGermany
| | - Tom Schwerd-Kleine
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Ryan Arlinghaus
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Zhijian Zhao
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Timm Schubert
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Olivier Marre
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Thomas Euler
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
- Bernstein Center for Computational Neuroscience, University of TübingenTübingenGermany
| |
Collapse
|
16
|
Frederiksen R, Peng YR, Sampath AP, Fain GL. Evolution of rod bipolar cells and rod vision. J Physiol 2025. [PMID: 39775947 DOI: 10.1113/jp287652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Bipolar cells are vertebrate retinal interneurons conveying signals from rod and cone photoreceptors to amacrine and ganglion cells. Bipolar cells are found in all vertebrates and have many structural and molecular affinities with photoreceptors; they probably appeared very early during vertebrate evolution in conjunction with rod and cone progenitors. There are two types of bipolar cells, responding to central illumination with depolarization (ON) or hyperpolarization (OFF). In most vertebrate species, rod signals are conveyed to specialized rod bipolar cells, which sum signals from many rods and facilitate detection at the visual threshold. Lamprey, which diverged from all other vertebrates in the late Cambrian, have both rod ON and rod OFF bipolar cells, but mammals have only rod ON cells. Rod signals in mammals are conveyed to output neurons indirectly via AII (or A2) amacrine cells, which synapse onto cone ON and cone OFF bipolar-cells and then to ganglion cells. These findings raise the question of when during retinal evolution rod OFF bipolar cells were lost. Because physiological recordings have been made from rod OFF bipolar cells in both cartilaginous fishes (dogfish) and urodeles (salamanders), rod OFF bipolar cells and their circuits must have been retained in vertebrate progenitors at least until the Devonian. Recent evidence showing that zebrafish retina processes rod signals similar to those in mammals indicates that rod OFF bipolar cells were lost at least twice. The sole utilization of rod ON bipolar cells may have provided a selective advantage from increased signal-to-noise discrimination near the visual threshold. KEY POINTS: Rods and cones have many structural and molecular similarities to bipolar cells, which are retinal interneurons conveying signals from photoreceptors to the retinal output. Bipolar cells can be either ON (centre depolarizing) or OFF (centre hyperpolarizing) and either rod or cone dominant. Lamprey, which diverged from all other vertebrates 500 million years ago, have both ON and OFF bipolar cells, which can each be either rod or cone dominant. We argue that this configuration of separate rod/cone bipolar-cell pathways is representative of early vertebrates. Rod ON and rod OFF bipolars persisted at least until the progenitors of amphibians in the Devonian, but mammals and teleost fishes have only rod ON bipolar cells and convey rod OFF signals via a specialized amacrine cell. We argue that rod OFF bipolar cells were lost in at least two different lineages during vertebrate evolution, probably to increase the signal-to-noise of rod vision.
Collapse
Affiliation(s)
- Rikard Frederiksen
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | - Gordon L Fain
- Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
17
|
Wutikeli H, Yu Y, Zhang T, Cao J, Nawy S, Shen Y. Role of Elavl-like RNA-binding protein in retinal development and signal transduction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167518. [PMID: 39307290 DOI: 10.1016/j.bbadis.2024.167518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/25/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
RNA-binding proteins (RBPs) play central roles in post-transcriptional gene regulation. However, the function of RBP in retinal progenitor cell differentiation and synaptic signal transmission are largely unexplored. Previously we have shown that Elavl2 regulates amacrine cell (AC) differentiation during retinogenesis, by directly binding to Nr4a2 and Barhl2. Elavl2 is expressed in early neuronal progenitors to mature neurons, and Elavl4 expression begins slightly later, during cortical neuron development as a paralog. Here, Retinal-specific Elavl2 and Elavl4 double knockout mice were made to further explore the role of Elavl2 and Elavl4 in retinal development and signal transduction. We disclose that Elavl4 binds to Satb1 to regulate Neurod1, then promoting retinal progenitor and amacrine cells differentiation. We were also surprised to find that Elavl2 interacted with GABAB receptors at the RNA and protein levels. In conclusion, Elavl2 and Elavl4 regulate amacrine cells differentiation through different pathways, leading to decreased scotopic vision. Our findings reveal the roles of Elavl2 and Elavl4 in retinal amacrine cells differentiation in modulating visual functions.
Collapse
Affiliation(s)
- Huxitaer Wutikeli
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Yao Yu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Tianlu Zhang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | | | - Scott Nawy
- University of California Berkeley, Department of Molecular and Cell Biology, Berkeley, CA, USA
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.
| |
Collapse
|
18
|
Avaroth Bhaskaran R, Vondráčková Z, Koladiya A, Čapek M, Dionigi F, Begall S, Burda H, Peichl L, Němec P. Distinct retinal ganglion cell types in strictly subterranean, naturally microphthalmic mammals. Proc Biol Sci 2025; 292:20242586. [PMID: 39809306 PMCID: PMC11732417 DOI: 10.1098/rspb.2024.2586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
African mole-rats (Bathyergidae, Rodentia) are subterranean rodents that live in extensive dark underground tunnel systems and rarely emerge aboveground. They can discriminate between light and dark but show no overt visually driven behaviours except for light-avoidance responses. Their eyes and central visual system are strongly reduced but not degenerated. Here, we focus on retinal ganglion cells (RGCs). Sighted mammals have numerous RGC types with distinct morphological and functional properties that encode different aspects of a visual scene. We analysed the morphological diversity of 216 intracellularly dye-injected RGCs in the giant mole-rat (Fukomys mechowii) and 48 RGCs in Ansell's mole-rat (Fukomys anselli). Using a hierarchical cluster analysis on 11 morphological parameters, we show that both species possess at least five RGC types with distinct dendritic field sizes and branching patterns. These resemble some RGC types of the mouse and rat, but mole-rat RGCs feature overall sparser and more asymmetric branching patterns. The dendritic trees of most RGCs in all clusters are monostratified in the inner plexiform layer, but bistratified and multistratified/diffuse cells also exist. Thus, although RGC morphologies have become disorganized, the basic retinal organization principle of parallel information processing by distinct RGC types is retained.
Collapse
Affiliation(s)
| | - Zuzana Vondráčková
- Department of Zoology, Faculty of Science, Charles University, Prague128 43, Czech Republic
| | - Abhishek Koladiya
- Department of Pediatrics, Stanford University School of Medicine, StanfordCA 95305, USA
| | - Martin Čapek
- Light Microscopy, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague142 20, Czech Republic
- Laboratory of Biomathematics, Institute of Physiology of the Czech Academy of Sciences, Prague142 20, Czech Republic
| | - Francesco Dionigi
- Department of Zoology, Faculty of Science, Charles University, Prague128 43, Czech Republic
| | - Sabine Begall
- Department of General Zoology, Faculty of Biology, University of Duisburg-Essen, Essen45141, Germany
| | - Hynek Burda
- Department of General Zoology, Faculty of Biology, University of Duisburg-Essen, Essen45141, Germany
- Department of Game Management and Wildlife Biology, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Prague165 00, Czech Republic
| | - Leo Peichl
- Dr. Senckenbergische Anatomie, Institute for Clinical Neuroanatomy, Frankfurt am Main60590, Germany
- Max Planck Institute for Brain Research, Frankfurt am Main60438, Germany
| | - Pavel Němec
- Department of Zoology, Faculty of Science, Charles University, Prague128 43, Czech Republic
| |
Collapse
|
19
|
Wang J, Zhang L, Cavallini M, Pahlevan A, Sun J, Morshedian A, Fain GL, Sampath AP, Peng YR. Molecular characterization of the sea lamprey retina illuminates the evolutionary origin of retinal cell types. Nat Commun 2024; 15:10761. [PMID: 39737973 PMCID: PMC11685597 DOI: 10.1038/s41467-024-55019-x] [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: 11/23/2023] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
The lamprey, a primitive jawless vertebrate whose ancestors diverged from all other vertebrates over 500 million years ago, offers a unique window into the ancient formation of the retina. Using single-cell RNA-sequencing, we characterize retinal cell types in the lamprey and compare them to those in mouse, chicken, and zebrafish. We find six cell classes and 74 distinct cell types, many shared with other vertebrate species. The conservation of cell types indicates their emergence early in vertebrate evolution, highlighting primordial designs of retinal circuits for the rod pathway, ON-OFF discrimination, and direction selectivity. The diversification of amacrine and some ganglion cell types appears, however, to be distinct in the lamprey. We further infer genetic regulators in specifying retinal cell classes and identify ancestral regulatory elements across species, noting decreased conservation in specifying amacrine cells. Altogether, our characterization of the lamprey retina illuminates the evolutionary origin of visual processing in the retina.
Collapse
Affiliation(s)
- Junqiang Wang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lin Zhang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martina Cavallini
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ali Pahlevan
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Junwei Sun
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ala Morshedian
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Gordon L Fain
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Goerdt L, Swain TA, Kar D, McGwin G, Berlin A, Clark ME, Owsley C, Sloan KR, Curcio CA. Band Visibility in High-Resolution Optical Coherence Tomography Assessed With a Custom Review Tool and Updated, Histology-Derived Nomenclature. Transl Vis Sci Technol 2024; 13:19. [PMID: 39671227 PMCID: PMC11645748 DOI: 10.1167/tvst.13.12.19] [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: 08/13/2024] [Accepted: 11/16/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose For structure-function research at the transition of aging to age-related macular degeneration, we refined the current consensus optical coherence tomography (OCT) nomenclature and evaluated a novel review software for investigational high-resolution OCT imaging (HR-OCT; <3 µm axial resolution). Method Volume electron microscopy, immunolocalizations, histology, and investigational devices informed a refined OCT nomenclature for a custom ImageJ-based review tool to assess retinal band visibility. We examined effects on retinal band visibility of automated real-time averaging (ART) 9 and 100 (11 eyes of 10 healthy young adults), aging (10 young vs 22 healthy aged), and age-related macular degeneration (AMD; 22 healthy aged, 17 early (e)AMD, 15 intermediate (i)AMD). Intrareader reliability was assessed. Results Bands not included in consensus nomenclature are now visible using HR-OCT: inner plexiform layer (IPL) 1-5, outer plexiform layer (OPL) 1-2, outer segment interdigitation zone 1-2 (OSIZ, including hyporeflective outer segments), and retinal pigment epithelium (RPE) 1-5. Cohen's kappa was 0.54-0.88 for inner and 0.67-0.83 for outer retinal bands in a subset of 10 eyes. IPL-3-5 and OPL-2 visibility benefitted from increased ART. OSIZ-2 and RPE-1,2,3,5 visibility was worse in aged eyes than in young eyes. OSIZ-1-2, RPE-1, and RPE-5 visibility decreased in eAMD and iAMD compared to healthy aged eyes. Conclusions We reliably identified 28 retinal bands using a novel review tool for HR-OCT. Image averaging improved inner retinal band visibility. Aging and AMD development impacted outer retinal band visibility. Translational Significance Detailed knowledge of anatomic structures visible on OCT will enhance precision in research, including AI training and structure-function analyses.
Collapse
Affiliation(s)
- Lukas Goerdt
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Thomas A. Swain
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Deepayan Kar
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Gerald McGwin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, AL, USA
| | - Andreas Berlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Ophthalmology, University of Würzburg, Würzburg, Germany
| | - Mark E. Clark
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Cynthia Owsley
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Kenneth R. Sloan
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| |
Collapse
|
21
|
Korympidou MM, Strauss S, Schubert T, Franke K, Berens P, Euler T, Vlasits AL. GABAergic amacrine cells balance biased chromatic information in the mouse retina. Cell Rep 2024; 43:114953. [PMID: 39509269 DOI: 10.1016/j.celrep.2024.114953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
The retina extracts chromatic information present in an animal's environment. How this information is processed in the retina is not well understood. In the mouse, chromatic information is not collected equally throughout the retina. Green and UV signals are primarily detected in the dorsal and ventral retina, respectively. However, at the output of the retina, chromatic tuning is more mixed throughout the retina. This suggests that lateral processing by inhibitory amacrine cells shapes chromatic information at the retinal output. We systematically surveyed the chromatic responses of dendritic processes of the 40+ GABAergic amacrine cell types. We identified 25 functional types with distinct chromatic and achromatic properties. We used pharmacology and a biologically inspired deep learning model to explore how inhibition and excitation shape the properties of functional types. Our data suggest that amacrine cells balance the biased spectral tuning of excitation, thereby supporting diversity of chromatic information at the retinal output.
Collapse
Affiliation(s)
- Maria M Korympidou
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, 72076 Tübingen, Germany
| | - Sarah Strauss
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, 72076 Tübingen, Germany; Hertie Institute for AI in Brain Health, University of Tübingen, 72076 Tübingen, Germany
| | - Timm Schubert
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Katrin Franke
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA 94303, USA
| | - Philipp Berens
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Hertie Institute for AI in Brain Health, University of Tübingen, 72076 Tübingen, Germany; Tübingen AI Center, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany.
| | - Anna L Vlasits
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Department of Ophthalmology & Visual Sciences, University of Illinois, Chicago, IL 60603, USA.
| |
Collapse
|
22
|
Choi C, Lee GJ, Chang S, Song YM, Kim DH. Inspiration from Visual Ecology for Advancing Multifunctional Robotic Vision Systems: Bio-inspired Electronic Eyes and Neuromorphic Image Sensors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2412252. [PMID: 39402806 DOI: 10.1002/adma.202412252] [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: 08/19/2024] [Revised: 09/18/2024] [Indexed: 11/29/2024]
Abstract
In robotics, particularly for autonomous navigation and human-robot collaboration, the significance of unconventional imaging techniques and efficient data processing capabilities is paramount. The unstructured environments encountered by robots, coupled with complex missions assigned to them, present numerous challenges necessitating diverse visual functionalities, and consequently, the development of multifunctional robotic vision systems has become indispensable. Meanwhile, rich diversity inherent in animal vision systems, honed over evolutionary epochs to meet their survival demands across varied habitats, serves as a profound source of inspirations. Here, recent advancements in multifunctional robotic vision systems drawing inspiration from natural ocular structures and their visual perception mechanisms are delineated. First, unique imaging functionalities of natural eyes across terrestrial, aerial, and aquatic habitats and visual signal processing mechanism of humans are explored. Then, designs and functionalities of bio-inspired electronic eyes are explored, engineered to mimic key components and underlying optical principles of natural eyes. Furthermore, neuromorphic image sensors are discussed, emulating functional properties of synapses, neurons, and retinas and thereby enhancing accuracy and efficiency of robotic vision tasks. Next, integration examples of electronic eyes with mobile robotic/biological systems are introduced. Finally, a forward-looking outlook on the development of bio-inspired electronic eyes and neuromorphic image sensors is provided.
Collapse
Affiliation(s)
- Changsoon Choi
- Center for Quantum Technology, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Gil Ju Lee
- School of Electrical and Electronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Sehui Chang
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Young Min Song
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- AI Graduate School, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Department of Semiconductor Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
23
|
Santistevan NJ, Ford CT, Gilsdorf CS, Grinblat Y. Behavioral and transcriptomic analyses of mecp2 function in zebrafish. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32981. [PMID: 38551133 DOI: 10.1002/ajmg.b.32981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 11/15/2024]
Abstract
Rett syndrome (RTT), a human neurodevelopmental disorder characterized by severe cognitive and motor impairments, is caused by dysfunction of the conserved transcriptional regulator Methyl-CpG-binding protein 2 (MECP2). Genetic analyses in mouse Mecp2 mutants, which exhibit key features of human RTT, have been essential for deciphering the mechanisms of MeCP2 function; nonetheless, our understanding of these complex mechanisms is incomplete. Zebrafish mecp2 mutants exhibit mild behavioral deficits but have not been analyzed in depth. Here, we combine transcriptomic and behavioral assays to assess baseline and stimulus-evoked motor responses and sensory filtering in zebrafish mecp2 mutants from 5 to 7 days post-fertilization (dpf). We show that zebrafish mecp2 function is required for normal thigmotaxis but is dispensable for gross movement, acoustic startle response, and sensory filtering (habituation and sensorimotor gating), and reveal a previously unknown role for mecp2 in behavioral responses to visual stimuli. RNA-seq analysis identified a large gene set that requires mecp2 function for correct transcription at 4 dpf, and pathway analysis revealed several pathways that require MeCP2 function in both zebrafish and mammals. These findings show that MeCP2's function as a transcriptional regulator is conserved across vertebrates and supports using zebrafish to complement mouse modeling in elucidating these conserved mechanisms.
Collapse
Affiliation(s)
- Nicholas J Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Colby T Ford
- School of Data Science, University of North Carolina, Charlotte, North Carolina, USA
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, North Carolina, USA
- Tuple, LLC, Charlotte, North Carolina, USA
| | - Cole S Gilsdorf
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
24
|
Nelson APK, Mole J, Pombo G, Gray RJ, Ruffle JK, Chan E, Rees GE, Cipolotti L, Nachev P. The minimal computational substrate of fluid intelligence. Cortex 2024; 179:62-76. [PMID: 39141936 DOI: 10.1016/j.cortex.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/08/2024] [Accepted: 07/01/2024] [Indexed: 08/16/2024]
Abstract
The quantification of cognitive powers rests on identifying a behavioural task that depends on them. Such dependence cannot be assured, for the powers a task invokes cannot be experimentally controlled or constrained a priori, resulting in unknown vulnerability to failure of specificity and generalisability. Evaluating a compact version of Raven's Advanced Progressive Matrices (RAPM), a widely used clinical test of fluid intelligence, we show that LaMa, a self-supervised artificial neural network trained solely on the completion of partially masked images of natural environmental scenes, achieves representative human-level test scores a prima vista, without any task-specific inductive bias or training. Compared with cohorts of healthy and focally lesioned participants, LaMa exhibits human-like variation with item difficulty, and produces errors characteristic of right frontal lobe damage under degradation of its ability to integrate global spatial patterns. LaMa's narrow training and limited capacity suggest matrix-style tests may be open to computationally simple solutions that need not necessarily invoke the substrates of reasoning.
Collapse
Affiliation(s)
- Amy P K Nelson
- High Dimensional Neurology Group, UCL Queen Square Institute of Neurology, University College London, Russell Square House, Bloomsbury, London, UK.
| | - Joe Mole
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK; UCL Queen Square Institute of Neurology, London, UK
| | - Guilherme Pombo
- High Dimensional Neurology Group, UCL Queen Square Institute of Neurology, University College London, Russell Square House, Bloomsbury, London, UK
| | - Robert J Gray
- High Dimensional Neurology Group, UCL Queen Square Institute of Neurology, University College London, Russell Square House, Bloomsbury, London, UK
| | - James K Ruffle
- High Dimensional Neurology Group, UCL Queen Square Institute of Neurology, University College London, Russell Square House, Bloomsbury, London, UK
| | - Edgar Chan
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK; UCL Queen Square Institute of Neurology, London, UK
| | - Geraint E Rees
- UCL Queen Square Institute of Neurology, London, UK; University College London, Gower Street, London, UK
| | - Lisa Cipolotti
- Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, London, UK; UCL Queen Square Institute of Neurology, London, UK
| | - Parashkev Nachev
- High Dimensional Neurology Group, UCL Queen Square Institute of Neurology, University College London, Russell Square House, Bloomsbury, London, UK.
| |
Collapse
|
25
|
do Nascimento THO, Pereira-Figueiredo D, Veroneze L, Nascimento AA, De Logu F, Nassini R, Campello-Costa P, Faria-Melibeu ADC, Souza Monteiro de Araújo D, Calaza KC. Functions of TRPs in retinal tissue in physiological and pathological conditions. Front Mol Neurosci 2024; 17:1459083. [PMID: 39386050 PMCID: PMC11461470 DOI: 10.3389/fnmol.2024.1459083] [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: 07/03/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
The Transient Receptor Potential (TRP) constitutes a family of channels subdivided into seven subfamilies: Ankyrin (TRPA), Canonical (TRPC), Melastatin (TRPM), Mucolipin (TRPML), no-mechano-potential C (TRPN), Polycystic (TRPP), and Vanilloid (TRPV). Although they are structurally similar to one another, the peculiarities of each subfamily are key to the response to stimuli and the signaling pathway that each one triggers. TRPs are non-selective cation channels, most of which are permeable to Ca2+, which is a well-established second messenger that modulates several intracellular signaling pathways and is involved in physiological and pathological conditions in various cell types. TRPs depolarize excitable cells by increasing the influx of Ca2+, Na+, and other cations. Most TRP families are activated by temperature variations, membrane stretching, or chemical agents and, therefore, are defined as polymodal channels. All TPRs are expressed, at some level, in the central nervous system (CNS) and ocular-related structures, such as the retina and optic nerve (ON), except the TRPP in the ON. TRPC, TRPM, TRPV, and TRPML are found in the retinal pigmented cells, whereas only TRPA1 and TRPM are detected in the uvea. Accordingly, several studies have focused on the search to unravel the role of TRPs in physiological and pathological conditions related to the eyes. Thus, this review aims to shed light on endogenous and exogenous modulators, triggered cell signaling pathways, and localization and roles of each subfamily of TRP channels in physiological and pathological conditions in the retina, optic nerve, and retinal pigmented epithelium of vertebrates.
Collapse
Affiliation(s)
- Thaianne Hanah Oliveira do Nascimento
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Biomedical Sciences, Biology Institute, Fluminense Federal University Niterói, Rio de Janeiro, Brazil
| | - Danniel Pereira-Figueiredo
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Neurosciences, Biology Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Louise Veroneze
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Neurosciences, Biology Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Amanda Alves Nascimento
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Neurosciences, Biology Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Paula Campello-Costa
- Laboratory of Neuroplasticity, Program of Neurosciences, Department of Neurobiology, Biology Institute, Niteroi, Brazil
| | - Adriana da Cunha Faria-Melibeu
- Laboratory of Neurobiology of Development, Program of Neurosciences, Department of Neurobiology, Biology Institute, Niteroi, Brazil
| | | | - Karin Costa Calaza
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Biomedical Sciences, Biology Institute, Fluminense Federal University Niterói, Rio de Janeiro, Brazil
- Laboratory Neurobiology of the Retina, Department of Neurobiology and Program of Neurosciences, Biology Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Kim SY, Park CH, Moon BH, Seabold GK. Murine Retina Outer Plexiform Layer Development and Transcriptome Analysis of Pre-Synapses in Photoreceptors. Life (Basel) 2024; 14:1103. [PMID: 39337887 PMCID: PMC11433150 DOI: 10.3390/life14091103] [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: 07/08/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited knowledge about rod versus (vs.) cone synapse development and the associated genes. The transcription factor neural retina leucine zipper (NRL) determines the rod vs. cone photoreceptor cell fate and is critical for rod differentiation. Nrl knockout mice fail to form rods, generating all cone or S-cone-like (SCL) photoreceptors in the retina, whereas ectopic expression of Nrl using a cone-rod homeobox (Crx) promoter (CrxpNrl) forms all rods. Here, we examined rod and cone pre-synapse development, including axonal elongation, terminal shaping, and synaptic lamination in the outer plexiform layer (OPL) in the presence or absence of Nrl. We show that NRL loss and knockdown result in delayed OPL maturation and plasticity with aberrant dendrites of bipolar neurons. The integrated analyses of the transcriptome in developing rods and SCLs with NRL CUT&RUN and synaptic gene ontology analyses identified G protein subunit beta (Gnb) 1 and p21 (RAC1) activated kinase 5 (Pak5 or Pak7) transcripts were upregulated in developing rods and down-regulated in developing SCLs. Notably, Gnb1 and Gnb5 are rod dominant, and Gnb3 is enriched in cones. NRL binds to the genes of Gnb1, Gnb3, and Gnb5. NRL also regulates pre-synapse ribbon genes, and their expression is altered in rods and SCLs. Our study of histological and gene analyses provides new insights into the morphogenesis of photoreceptor pre-synapse development and regulation of associated genes in the developing retina.
Collapse
Affiliation(s)
- Soo-Young Kim
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Haewon Park
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bo-Hyun Moon
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Gail K Seabold
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Carrero L, Antequera D, Municio C, Carro E. Circadian rhythm disruption and retinal dysfunction: a bidirectional link in Alzheimer's disease? Neural Regen Res 2024; 19:1967-1972. [PMID: 38227523 DOI: 10.4103/1673-5374.390962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/07/2023] [Indexed: 01/17/2024] Open
Abstract
Dysfunction in circadian rhythms is a common occurrence in patients with Alzheimer's disease. A predominant function of the retina is circadian synchronization, carrying information to the brain through the retinohypothalamic tract, which projects to the suprachiasmatic nucleus. Notably, Alzheimer's disease hallmarks, including amyloid-β, are present in the retinas of Alzheimer's disease patients, followed/associated by structural and functional disturbances. However, the mechanistic link between circadian dysfunction and the pathological changes affecting the retina in Alzheimer's disease is not fully understood, although some studies point to the possibility that retinal dysfunction could be considered an early pathological process that directly modulates the circadian rhythm.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Eva Carro
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
28
|
Stevens-Sostre WA, Hoon M. Cellular and Molecular Mechanisms Regulating Retinal Synapse Development. Annu Rev Vis Sci 2024; 10:377-402. [PMID: 39292551 PMCID: PMC12022667 DOI: 10.1146/annurev-vision-102122-105721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Synapse formation within the retinal circuit ensures that distinct neuronal types can communicate efficiently to process visual signals. Synapses thus form the core of the visual computations performed by the retinal circuit. Retinal synapses are diverse but can be broadly categorized into multipartner ribbon synapses and 1:1 conventional synapses. In this article, we review our current understanding of the cellular and molecular mechanisms that regulate the functional establishment of mammalian retinal synapses, including the role of adhesion proteins, synaptic proteins, extracellular matrix and cytoskeletal-associated proteins, and activity-dependent cues. We outline future directions and areas of research that will expand our knowledge of these mechanisms. Understanding the regulators moderating synapse formation and function not only reveals the integrated developmental processes that establish retinal circuits, but also divulges the identity of mechanisms that could be engaged during disease and degeneration.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
29
|
Sigulinsky CL, Pfeiffer RL, Jones BW. Retinal Connectomics: A Review. Annu Rev Vis Sci 2024; 10:263-291. [PMID: 39292552 DOI: 10.1146/annurev-vision-102122-110414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
The retina is an ideal model for understanding the fundamental rules for how neural networks are constructed. The compact neural networks of the retina perform all of the initial processing of visual information before transmission to higher visual centers in the brain. The field of retinal connectomics uses high-resolution electron microscopy datasets to map the intricate organization of these networks and further our understanding of how these computations are performed by revealing the fundamental topologies and allowable networks behind retinal computations. In this article, we review some of the notable advances that retinal connectomics has provided in our understanding of the specific cells and the organization of their connectivities within the retina, as well as how these are shaped in development and break down in disease. Using these anatomical maps to inform modeling has been, and will continue to be, instrumental in understanding how the retina processes visual signals.
Collapse
Affiliation(s)
- Crystal L Sigulinsky
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, USA;
| | - Rebecca L Pfeiffer
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, USA;
| | - Bryan William Jones
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah, USA;
| |
Collapse
|
30
|
Wei H, Yao W. Computational Modeling of Ganglion Cell Bicolor Opponent Receptive Fields and FPGA Adaptation for Parallel Arrays. Biomimetics (Basel) 2024; 9:526. [PMID: 39329548 PMCID: PMC11430245 DOI: 10.3390/biomimetics9090526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
The biological system is not a perfect system, but it is a relatively complete system. It is difficult to realize the lower power consumption and high parallelism that characterize biological systems if lower-level information pathways are ignored. In this paper, we focus on the K, M and P pathways of visual signal processing from the retina to the lateral geniculate nucleus (LGN). We model the visual system at a fine-grained level to ensure efficient information transmission while minimizing energy use. We also implement a circuit-level distributed parallel computing model on FPGAs. The results show that we are able to transfer information with low energy consumption and high parallelism. The Artix-7 family of xc7a200tsbv484-1 FPGAs can reach a maximum frequency of 200 MHz and a maximum parallelism of 600, and a single receptive field model consumes only 0.142 W of power. This can be useful for building assistive vision systems for small and light devices.
Collapse
Affiliation(s)
- Hui Wei
- Laboratory of Algorithms for Cognitive Models, School of Computer Science, Fudan University, Shanghai 200438, China;
| | | |
Collapse
|
31
|
Müllner FE, Roska B. Individual thalamic inhibitory interneurons are functionally specialized toward distinct visual features. Neuron 2024; 112:2765-2782.e9. [PMID: 38917805 PMCID: PMC11348917 DOI: 10.1016/j.neuron.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Inhibitory interneurons in the dorsolateral geniculate nucleus (dLGN) are situated at the first central synapse of the image-forming visual pathway, but little is known about their function. Given their anatomy, they are expected to be multiplexors, integrating many different retinal channels along their dendrites. Here, using targeted single-cell-initiated rabies tracing, we found that mouse dLGN interneurons exhibit a degree of retinal input specialization similar to thalamocortical neurons. Some are anatomically highly specialized, for example, toward motion-selective information. Two-photon calcium imaging performed in vivo revealed that interneurons are also functionally specialized. In mice lacking retinal horizontal direction selectivity, horizontal direction selectivity is reduced in interneurons, suggesting a causal link between input and functional specialization. Functional specialization is not only present at interneuron somata but also extends into their dendrites. Altogether, inhibitory interneurons globally display distinct visual features which reflect their retinal input specialization and are ideally suited to perform feature-selective inhibition.
Collapse
Affiliation(s)
- Fiona E Müllner
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
32
|
Yao Y, Song Q, Zhang J, Wen Y, Dou X. Retina-Brain Homology: The Correlation Between Ophthalmic or Retinal Artery Occlusion and Ischemic Stroke. Eye Brain 2024; 16:25-38. [PMID: 39156910 PMCID: PMC11328846 DOI: 10.2147/eb.s454977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/23/2024] [Indexed: 08/20/2024] Open
Abstract
The retina's similar structure and function to the brain make it a unique visual "window" for studying cerebral disorders. Ophthalmic artery occlusion (OAO) or retinal artery occlusion (RAO) is a severe ophthalmic emergency that significantly affects visual acuity. Studies have demonstrated that patients with OAO or RAO face a notably higher risk of future acute ischemic stroke (AIS). However, ophthalmologists often overlook multidisciplinary approach involving the neurologist, to evaluate the risk of AIS and devise clinical treatment strategies for patients with OAO or RAO. Unlike the successful use of thrombolysis in AIS, the application of thrombolysis for OAO or RAO remains limited and controversial due to insufficient reliable evidence. In this review, we aim to summarize the anatomical and functional connections between the retina and the brain, and the clinical connection between OAO or RAO and AIS, compare and review recent advances in the effectiveness and safety of intravenous and intra-arterial thrombolysis therapy in patients with OAO or RAO, and discuss future research directions for OAO or RAO. Our goal is to advance the development of multidisciplinary diagnosis and treatment strategies for the disease, as well as to establish expedited pathways or thrombolysis guidelines for vascular intervention.
Collapse
Affiliation(s)
- Yufeng Yao
- Shantou University Medical College, Shantou, Guangdong Province, People’s Republic of China
- Department of Ophthalmology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Qiyuan Song
- Department of Ophthalmology, Shenzhen Children’s Hospital, Shenzhen, Guangdong Province, People’s Republic of China
| | - Jingnan Zhang
- Department of Ophthalmology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
- Shenzhen University Medical College, No.1066 Xueyuan Road, Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Yingying Wen
- Department of Ophthalmology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
- Shenzhen University Medical College, No.1066 Xueyuan Road, Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Xiaoyan Dou
- Department of Ophthalmology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, People’s Republic of China
| |
Collapse
|
33
|
Kumar D, Khan B, Okcay Y, Sis ÇÖ, Abdallah A, Murray F, Sharma A, Uemura M, Taliyan R, Heinbockel T, Rahman S, Goyal R. Dynamic endocannabinoid-mediated neuromodulation of retinal circadian circuitry. Ageing Res Rev 2024; 99:102401. [PMID: 38964508 DOI: 10.1016/j.arr.2024.102401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Circadian rhythms are biological rhythms that originate from the "master circadian clock," called the suprachiasmatic nucleus (SCN). SCN orchestrates the circadian rhythms using light as a chief zeitgeber, enabling humans to synchronize their daily physio-behavioral activities with the Earth's light-dark cycle. However, chronic/ irregular photic disturbances from the retina via the retinohypothalamic tract (RHT) can disrupt the amplitude and the expression of clock genes, such as the period circadian clock 2, causing circadian rhythm disruption (CRd) and associated neuropathologies. The present review discusses neuromodulation across the RHT originating from retinal photic inputs and modulation offered by endocannabinoids as a function of mitigation of the CRd and associated neuro-dysfunction. Literature indicates that cannabinoid agonists alleviate the SCN's ability to get entrained to light by modulating the activity of its chief neurotransmitter, i.e., γ-aminobutyric acid, thus preventing light-induced disruption of activity rhythms in laboratory animals. In the retina, endocannabinoid signaling modulates the overall gain of the retinal ganglion cells by regulating the membrane currents (Ca2+, K+, and Cl- channels) and glutamatergic neurotransmission of photoreceptors and bipolar cells. Additionally, endocannabinoids signalling also regulate the high-voltage-activated Ca2+ channels to mitigate the retinal ganglion cells and intrinsically photosensitive retinal ganglion cells-mediated glutamate release in the SCN, thus regulating the RHT-mediated light stimulation of SCN neurons to prevent excitotoxicity. As per the literature, cannabinoid receptors 1 and 2 are becoming newer targets in drug discovery paradigms, and the involvement of endocannabinoids in light-induced CRd through the RHT may possibly mitigate severe neuropathologies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| | - Bareera Khan
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India
| | - Yagmur Okcay
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Çağıl Önal Sis
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Aya Abdallah
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Fiona Murray
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Ashish Sharma
- School of Medicine, Washington University, St. Louis, USA
| | - Maiko Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology Science, Pilani, Rajasthan 333301, India.
| | - Thomas Heinbockel
- Howard University College of Medicine, Department of Anatomy, Washington, DC 20059, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy South Dakota State University, Brookings, SD, USA.
| | - Rohit Goyal
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| |
Collapse
|
34
|
Lee SCS, Wei AJ, Martin PR, Grünert U. Thorny and Tufted Retinal Ganglion Cells Express the Transcription Factor Forkhead Proteins Foxp1 and Foxp2 in Marmoset (Callithrix jacchus). J Comp Neurol 2024; 532:e25663. [PMID: 39235164 DOI: 10.1002/cne.25663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
The transcription factor forkhead/winged-helix domain proteins Foxp1 and Foxp2 have previously been studied in mouse retina, where they are expressed in retinal ganglion cells named F-mini and F-midi. Here we show that both transcription factors are expressed by small subpopulations (on average less than 10%) of retinal ganglion cells in the retina of the marmoset monkey (Callithrix jacchus). The morphology of Foxp1- and Foxp2-expressing cells was revealed by intracellular DiI injections of immunofluorescent cells. Foxp1- and Foxp2-expressing cells comprised multiple types of wide-field ganglion cells, including broad thorny cells, narrow thorny cells, and tufted cells. The large majority of Foxp2-expressing cells were identified as tufted cells. Tufted cells stratify broadly in the middle of the inner plexiform layer. They resemble broad thorny cells but their proximal dendrites are bare of branches and the distal dendrites branch frequently forming dense dendritic tufts. Double labeling with calretinin, a previously established marker for broad thorny and narrow thorny cells, showed that only a small proportion of ganglion cells co-expressed calretinin and Foxp1 or Foxp2 supporting the idea that the two markers are differentially expressed in retinal ganglion cells of marmoset retina.
Collapse
Affiliation(s)
- Sammy C S Lee
- Save Sight Institute and Discipline of Ophthalmology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anlai J Wei
- Save Sight Institute and Discipline of Ophthalmology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul R Martin
- Save Sight Institute and Discipline of Ophthalmology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ulrike Grünert
- Save Sight Institute and Discipline of Ophthalmology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Choi J, Joisher HNV, Gill HK, Lin L, Cepko C. Characterization of the development of the high-acuity area of the chick retina. Dev Biol 2024; 511:39-52. [PMID: 38548147 DOI: 10.1016/j.ydbio.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
The fovea is a small region within the central retina that is responsible for our high acuity daylight vision. Chickens also have a high acuity area (HAA), and are one of the few species that enables studies of the mechanisms of HAA development, due to accessible embryonic tissue and methods to readily perturb gene expression. To enable such studies, we characterized the development of the chick HAA using single molecule fluorescent in situ hybridization (smFISH), along with more classical methods. We found that Fgf8 provides a molecular marker for the HAA throughout development and into adult stages, allowing studies of the cellular composition of this area over time. The radial dimension of the ganglion cell layer (GCL) was seen to be the greatest at the HAA throughout development, beginning during the period of neurogenesis, suggesting that genesis, rather than cell death, creates a higher level of retinal ganglion cells (RGCs) in this area. In contrast, the HAA acquired its characteristic high density of cone photoreceptors post-hatching, which is well after the period of neurogenesis. We also confirmed that rod photoreceptors are not present in the HAA. Analyses of cell death in the developing photoreceptor layer, where rods would reside, did not show apoptotic cells, suggesting that lack of genesis, rather than death, created the "rod-free zone" (RFZ). Quantification of each cone photoreceptor subtype showed an ordered mosaic of most cone subtypes. The changes in cellular densities and cell subtypes between the developing and mature HAA provide some answers to the overarching strategy used by the retina to create this area and provide a framework for future studies of the mechanisms underlying its formation.
Collapse
Affiliation(s)
- Jiho Choi
- Department of Genetics, Blavatnik Institute, USA; Department of Ophthalmology, Harvard Medical School, USA; Howard Hughes Medical Institute, USA
| | - Heer N V Joisher
- Department of Genetics, Blavatnik Institute, USA; Department of Ophthalmology, Harvard Medical School, USA; Howard Hughes Medical Institute, USA
| | | | - Lucas Lin
- Department of Genetics, Blavatnik Institute, USA; Department of Ophthalmology, Harvard Medical School, USA; Howard Hughes Medical Institute, USA
| | - Constance Cepko
- Department of Genetics, Blavatnik Institute, USA; Department of Ophthalmology, Harvard Medical School, USA; Howard Hughes Medical Institute, USA.
| |
Collapse
|
36
|
Xu Y, Tummala SR, Chen X, Vardi N. VDAC in Retinal Health and Disease. Biomolecules 2024; 14:654. [PMID: 38927058 PMCID: PMC11201675 DOI: 10.3390/biom14060654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The retina, a tissue of the central nervous system, is vital for vision as its photoreceptors capture light and transform it into electrical signals, which are further processed before they are sent to the brain to be interpreted as images. The retina is unique in that it is continuously exposed to light and has the highest metabolic rate and demand for energy amongst all the tissues in the body. Consequently, the retina is very susceptible to oxidative stress. VDAC, a pore in the outer membrane of mitochondria, shuttles metabolites between mitochondria and the cytosol and normally protects cells from oxidative damage, but when a cell's integrity is greatly compromised it initiates cell death. There are three isoforms of VDAC, and existing evidence indicates that all three are expressed in the retina. However, their precise localization and function in each cell type is unknown. It appears that most retinal cells express substantial amounts of VDAC2 and VDAC3, presumably to protect them from oxidative stress. Photoreceptors express VDAC2, HK2, and PKM2-key proteins in the Warburg pathway that also protect these cells. Consistent with its role in initiating cell death, VDAC is overexpressed in the retinal degenerative diseases retinitis pigmentosa, age related macular degeneration (AMD), and glaucoma. Treatment with antioxidants or inhibiting VDAC oligomerization reduced its expression and improved cell survival. Thus, VDAC may be a promising therapeutic candidate for the treatment of these diseases.
Collapse
Affiliation(s)
- Ying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (Y.X.); (X.C.)
| | - Shanti R. Tummala
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Xiongmin Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (Y.X.); (X.C.)
| | - Noga Vardi
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Baden T. The vertebrate retina: a window into the evolution of computation in the brain. Curr Opin Behav Sci 2024; 57:None. [PMID: 38899158 PMCID: PMC11183302 DOI: 10.1016/j.cobeha.2024.101391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 06/21/2024]
Abstract
Animal brains are probably the most complex computational machines on our planet, and like everything in biology, they are the product of evolution. Advances in developmental and palaeobiology have been expanding our general understanding of how nervous systems can change at a molecular and structural level. However, how these changes translate into altered function - that is, into 'computation' - remains comparatively sparsely explored. What, concretely, does it mean for neuronal computation when neurons change their morphology and connectivity, when new neurons appear or old ones disappear, or when transmitter systems are slowly modified over many generations? And how does evolution use these many possible knobs and dials to constantly tune computation to give rise to the amazing diversity in animal behaviours we see today? Addressing these major gaps of understanding benefits from choosing a suitable model system. Here, I present the vertebrate retina as one perhaps unusually promising candidate. The retina is ancient and displays highly conserved core organisational principles across the entire vertebrate lineage, alongside a myriad of adjustments across extant species that were shaped by the history of their visual ecology. Moreover, the computational logic of the retina is readily interrogated experimentally, and our existing understanding of retinal circuits in a handful of species can serve as an anchor when exploring the visual circuit adaptations across the entire vertebrate tree of life, from fish deep in the aphotic zone of the oceans to eagles soaring high up in the sky.
Collapse
|
38
|
Rathbun DL, Jalligampala A, Zrenner E, Hosseinzadeh Z. Improvements for recording retinal function with Microelectrode Arrays. MethodsX 2024; 12:102543. [PMID: 38313698 PMCID: PMC10834997 DOI: 10.1016/j.mex.2023.102543] [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: 12/02/2023] [Accepted: 12/28/2023] [Indexed: 02/06/2024] Open
Abstract
A microelectrode array (MEA) is a configuration of multiple electrodes that enables the concurrent targeting of multiple sites for extracellular recording and stimulation. By utilizing light pulses or electrical stimulations, MEA recordings unveil the complex patterns of electrical activities that arise from the signaling processes within the retinal network. Here, we present a stepwise approach for using microelectrode arrays (MEAs) for recording action potentials from the mouse retina in response to electrical and light stimuli. We provide detailed techniques accompanied by description of a custom optical system, example recordings, troubleshooting guidelines, and data processing methods including spike sorting and code resources for analyzing light and electrical responses. The comprehensive nature of this paper aims to guide researchers in utilizing MEAs effectively for investigating retinal functionality. In particular, it can be easy to have a MEA experiment fail, but hard to identify the source of the failure. This paper is meant to demystify that process. It includes:•A description of MEA setup, recording, and spike data validation.•A troubleshooting guide for common failure modes in MEA recordings from mouse retina.•Spike detection and sorting to precisely extract distinctive action potential.
Collapse
Affiliation(s)
- D L Rathbun
- Department of Ophthalmology, Detroit Institute of Ophthalmology, Henry Ford Health System, Detroit, MI 48202, USA
| | - A Jalligampala
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, KY 40202, USA
| | - E Zrenner
- Institute for Ophthalmic Research, Eberhard Karls University, 72076 Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience (CIN), 72076 Tübingen, Germany
| | - Z Hosseinzadeh
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Liu Y, Zhang J, Jiang Z, Qin M, Xu M, Zhang S, Ma G. Organization of corticocortical and thalamocortical top-down inputs in the primary visual cortex. Nat Commun 2024; 15:4495. [PMID: 38802410 PMCID: PMC11130321 DOI: 10.1038/s41467-024-48924-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Unified visual perception requires integration of bottom-up and top-down inputs in the primary visual cortex (V1), yet the organization of top-down inputs in V1 remains unclear. Here, we used optogenetics-assisted circuit mapping to identify how multiple top-down inputs from higher-order cortical and thalamic areas engage V1 excitatory and inhibitory neurons. Top-down inputs overlap in superficial layers yet segregate in deep layers. Inputs from the medial secondary visual cortex (V2M) and anterior cingulate cortex (ACA) converge on L6 Pyrs, whereas ventrolateral orbitofrontal cortex (ORBvl) and lateral posterior thalamic nucleus (LP) inputs are processed in parallel in Pyr-type-specific subnetworks (Pyr←ORBvl and Pyr←LP) and drive mutual inhibition between them via local interneurons. Our study deepens understanding of the top-down modulation mechanisms of visual processing and establishes that V2M and ACA inputs in L6 employ integrated processing distinct from the parallel processing of LP and ORBvl inputs in L5.
Collapse
Affiliation(s)
- Yanmei Liu
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahe Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhishan Jiang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meiling Qin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Siyu Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Guofen Ma
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
40
|
Kartsaki E, Hilgen G, Sernagor E, Cessac B. How Does the Inner Retinal Network Shape the Ganglion Cells Receptive Field? A Computational Study. Neural Comput 2024; 36:1041-1083. [PMID: 38669693 DOI: 10.1162/neco_a_01663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/02/2024] [Indexed: 04/28/2024]
Abstract
We consider a model of basic inner retinal connectivity where bipolar and amacrine cells interconnect and both cell types project onto ganglion cells, modulating their response output to the brain visual areas. We derive an analytical formula for the spatiotemporal response of retinal ganglion cells to stimuli, taking into account the effects of amacrine cells inhibition. This analysis reveals two important functional parameters of the network: (1) the intensity of the interactions between bipolar and amacrine cells and (2) the characteristic timescale of these responses. Both parameters have a profound combined impact on the spatiotemporal features of retinal ganglion cells' responses to light. The validity of the model is confirmed by faithfully reproducing pharmacogenetic experimental results obtained by stimulating excitatory DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) expressed on ganglion cells and amacrine cells' subclasses, thereby modifying the inner retinal network activity to visual stimuli in a complex, entangled manner. Our mathematical model allows us to explore and decipher these complex effects in a manner that would not be feasible experimentally and provides novel insights in retinal dynamics.
Collapse
Affiliation(s)
- Evgenia Kartsaki
- Université Côte d'Azur, Inria, Biovision Team and Neuromod Institute, Sophia Antipolis, France
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K.
| | - Gerrit Hilgen
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
- Health and Life Sciences, Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, U.K.
| | - Evelyne Sernagor
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K.
| | - Bruno Cessac
- Université Côte d'Azur, Inria, Biovision Team and Neuromod Institute, Sophia Antipolis, France
| |
Collapse
|
41
|
Carleton M, Oesch NW. Asymmetric Activation of ON and OFF Pathways in the Degenerated Retina. eNeuro 2024; 11:ENEURO.0110-24.2024. [PMID: 38719453 PMCID: PMC11097263 DOI: 10.1523/eneuro.0110-24.2024] [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: 03/13/2024] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/18/2024] Open
Abstract
Retinal prosthetics are one of the leading therapeutic strategies to restore lost vision in patients with retinitis pigmentosa and age-related macular degeneration. Much work has described patterns of spiking in retinal ganglion cells (RGCs) in response to electrical stimulation, but less work has examined the underlying retinal circuitry that is activated by electrical stimulation to drive these responses. Surprisingly, little is known about the role of inhibition in generating electrical responses or how inhibition might be altered during degeneration. Using whole-cell voltage-clamp recordings during subretinal electrical stimulation in the rd10 and wild-type (wt) retina, we found electrically evoked synaptic inputs differed between ON and OFF RGC populations, with ON cells receiving mostly excitation and OFF cells receiving mostly inhibition and very little excitation. We found that the inhibition of OFF bipolar cells limits excitation in OFF RGCs, and a majority of both pre- and postsynaptic inhibition in the OFF pathway arises from glycinergic amacrine cells, and the stimulation of the ON pathway contributes to inhibitory inputs to the RGC. We also show that this presynaptic inhibition in the OFF pathway is greater in the rd10 retina, compared with that in the wt retina.
Collapse
Affiliation(s)
- Maya Carleton
- Department of Psychology, University of California San Diego, La Jolla, California 92093
| | - Nicholas W Oesch
- Department of Psychology, University of California San Diego, La Jolla, California 92093
- Department of Ophthalmology, University of California San Diego, La Jolla, California 92093
- Neuroscience Graduate Program, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
42
|
Touahri Y, Hanna J, Tachibana N, Okawa S, Liu H, David LA, Olender T, Vasan L, Pak A, Mehta DN, Chinchalongporn V, Balakrishnan A, Cantrup R, Dixit R, Mattar P, Saleh F, Ilnytskyy Y, Murshed M, Mains PE, Kovalchuk I, Lefebvre JL, Leong HS, Cayouette M, Wang C, Del Sol A, Brand M, Reese BE, Schuurmans C. Pten regulates endocytic trafficking of cell adhesion and Wnt signaling molecules to pattern the retina. Cell Rep 2024; 43:114005. [PMID: 38551961 PMCID: PMC11290456 DOI: 10.1016/j.celrep.2024.114005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/30/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024] Open
Abstract
The retina is exquisitely patterned, with neuronal somata positioned at regular intervals to completely sample the visual field. Here, we show that phosphatase and tensin homolog (Pten) controls starburst amacrine cell spacing by modulating vesicular trafficking of cell adhesion molecules and Wnt proteins. Single-cell transcriptomics and double-mutant analyses revealed that Pten and Down syndrome cell adhesion molecule Dscam) are co-expressed and function additively to pattern starburst amacrine cell mosaics. Mechanistically, Pten loss accelerates the endocytic trafficking of DSCAM, FAT3, and MEGF10 off the cell membrane and into endocytic vesicles in amacrine cells. Accordingly, the vesicular proteome, a molecular signature of the cell of origin, is enriched in exocytosis, vesicle-mediated transport, and receptor internalization proteins in Pten conditional knockout (PtencKO) retinas. Wnt signaling molecules are also enriched in PtencKO retinal vesicles, and the genetic or pharmacological disruption of Wnt signaling phenocopies amacrine cell patterning defects. Pten thus controls vesicular trafficking of cell adhesion and signaling molecules to establish retinal amacrine cell mosaics.
Collapse
Affiliation(s)
- Yacine Touahri
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada
| | - Joseph Hanna
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nobuhiko Tachibana
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hedy Liu
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Luke Ajay David
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Thomas Olender
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Lakshmy Vasan
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alissa Pak
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dhruv Nimesh Mehta
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada
| | - Vorapin Chinchalongporn
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Robert Cantrup
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Rajiv Dixit
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Pierre Mattar
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Fermisk Saleh
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Monzur Murshed
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3G 1A6, Canada
| | - Paul E Mains
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Julie L Lefebvre
- Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada; Program for Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Immunology, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Marjorie Brand
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Benjamin E Reese
- Department of Psychological and Brain Sciences, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106-5060, USA
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
43
|
Müller R. Bioinspiration from bats and new paradigms for autonomy in natural environments. BIOINSPIRATION & BIOMIMETICS 2024; 19:033001. [PMID: 38452384 DOI: 10.1088/1748-3190/ad311e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/07/2024] [Indexed: 03/09/2024]
Abstract
Achieving autonomous operation in complex natural environment remains an unsolved challenge. Conventional engineering approaches to this problem have focused on collecting large amounts of sensory data that are used to create detailed digital models of the environment. However, this only postpones solving the challenge of identifying the relevant sensory information and linking it to action control to the domain of the digital world model. Furthermore, it imposes high demands in terms of computing power and introduces large processing latencies that hamper autonomous real-time performance. Certain species of bats that are able to navigate and hunt their prey in dense vegetation could be a biological model system for an alternative approach to addressing the fundamental issues associated with autonomy in complex natural environments. Bats navigating in dense vegetation rely on clutter echoes, i.e. signals that consist of unresolved contributions from many scatters. Yet, the animals are able to extract the relevant information from these input signals with brains that are often less than 1 g in mass. Pilot results indicate that information relevant to location identification and passageway finding can be directly obtained from clutter echoes, opening up the possibility that the bats' skill can be replicated in man-made autonomous systems.
Collapse
Affiliation(s)
- Rolf Müller
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, United States of America
| |
Collapse
|
44
|
Ghanbarpour M, Haghiri S, Hazzazi F, Assaad M, Chaudhary MA, Ahmadi A. Investigation on Vision System: Digital FPGA Implementation in Case of Retina Rod Cells. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2024; 18:299-307. [PMID: 37824307 DOI: 10.1109/tbcas.2023.3323324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The development of prostheses and treatments for illnesses and recovery has recently been centered on hardware modeling for various delicate biological components, including the nervous system, brain, eyes, and heart. The retina, being the thinnest and deepest layer of the eye, is of particular interest. In this study, we employ the Nyquist-Based Approximation of Retina Rod Cell (NBAoRRC) approach, which has been adapted to utilize Look-Up Tables (LUTs) rather than original functions, to implement rod cells in the retina using cost-effective hardware. In modern mathematical models, numerous nonlinear functions are used to represent the activity of these cells. However, these nonlinear functions would require a substantial amount of hardware for direct implementation and may not meet the required speed constraints. The proposed method eliminates the need for multiplication functions and utilizes a fast, cost-effective rod cell device. Simulation results demonstrate the extent to which the proposed model aligns with the behavior of the primary rod cell model, particularly in terms of dynamic behavior. Based on the results of hardware implementation using the Field-Programmable Gate Arrays (FPGA) board Virtex-5, the proposed model is shown to be reliable, consume 30 percent less power than the primary model, and have reduced hardware resource requirements. Based on the results of hardware implementation using the reconfigurable FPGA board Virtex-5, the proposed model is reliable, uses 30% less power consumption than the primary model in the worth state of the set of approximation method, and has a reduced hardware resource requirement. In fact, using the proposed model, this reduction in the power consumption can be achieved. Finally, in this article, by using the LUT which is systematically sampled (Nyquist rate), we were able to remove all costly operators in terms of hardware (digital) realization and achieve very good results in the field of digital implementation in two scales of network and single neuron.
Collapse
|
45
|
Zhang X, Wang X, Zhu J, Chen K, Ullah R, Tong J, Shen Y. Retinal VIP-amacrine cells: their development, structure, and function. Eye (Lond) 2024; 38:1065-1076. [PMID: 38066110 PMCID: PMC11009269 DOI: 10.1038/s41433-023-02844-x] [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: 07/18/2022] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 04/13/2024] Open
Abstract
Amacrine cells (ACs) are the most structurally and functionally diverse neuron type in the retina. Different ACs have distinct functions, such as neuropeptide secretion and inhibitory connection. Vasoactive intestinal peptide (VIP) -ergic -ACs are retina gamma-aminobutyric acid (GABA) -ergic -ACs that were discovered long ago. They secrete VIP and form connections with bipolar cells (BCs), other ACs, and retinal ganglion cells (RGCs). They have a specific structure, density, distribution, and function. They play an important role in myopia, light stimulated responses, retinal vascular disease and other ocular diseases. Their significance in the study of refractive development and disease is increasing daily. However, a systematic review of the structure and function of retinal VIP-ACs is lacking. We discussed the detailed characteristics of VIP-ACs from every aspect across species and providing systematic knowledge base for future studies. Our review led to the main conclusion that retinal VIP-ACs develop early, and although their morphology and distribution across species are not the same, they have similar functions in a wide range of ocular diseases based on their function of secreting neuropeptides and forming inhibitory connections with other cells.
Collapse
Affiliation(s)
- Xuhong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jiru Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China
| | - Rahim Ullah
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Medical School, Zhejiang University, Hangzhou, China.
| |
Collapse
|
46
|
Namgung SD, Kim RM, Han JH, Nam KT. Circular polarization sensitive opto-neuromorphic operation at plasmonic hot electron transistor using chiral gold nanoparticles. NANOTECHNOLOGY 2024; 35:245201. [PMID: 38461550 DOI: 10.1088/1361-6528/ad321e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/10/2024] [Indexed: 03/12/2024]
Abstract
Opto-neuromorphic operation is critical for biological system to recognize the visual objects and mimicking such operation is important for artificial prosthesis as well as machine vision system for industrial applications. To sophisticatedly mimic biological system, regulation of learning and memorizing efficiency is needed, however engineered synthetic platform has been lack of controllability, which makes huge gap between biological system and synthetic platform. Here we demonstrated controllable learning and memorizing opto-neuromorphic operation at plasmonic hot electron transistor. Especially, circularly polarized light (CPL) sensitive synaptic characteristics and learning experience capability are enabled by incorporating chiral plasmonic nanoparticle. Furthermore, gate voltage gives rise to controllable neuromorphic operation due to hot electron injection and trapping effect, resulting in high remaining synaptic weight of ∼70% at negative gate voltage under CPL excitation. We believe that this discovery makes significant leap toward on-demand in-sensor computing as well as toward bio-realistic device.
Collapse
Affiliation(s)
- Seok Daniel Namgung
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Ryeong Myeong Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Division of Biomedical Metrology, Medical Metrology Group, Korea Research Institute of Standards and Science (KRISS) Daejeon 34113, Republic of Korea
| | - Jeong Hyun Han
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Ki Tae Nam
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
47
|
Abubaker MB, Hsu FY, Feng KL, Chu LA, de Belle JS, Chiang AS. Asymmetric neurons are necessary for olfactory learning in the Drosophila brain. Curr Biol 2024; 34:946-957.e4. [PMID: 38320552 DOI: 10.1016/j.cub.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/31/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024]
Abstract
Animals have complementary parallel memory systems that process signals from various sensory modalities. In the brain of the fruit fly Drosophila melanogaster, mushroom body (MB) circuitry is the primary associative neuropil, critical for all stages of olfactory memory. Here, our findings suggest that active signaling from specific asymmetric body (AB) neurons is also crucial for this process. These AB neurons respond to odors and electric shock separately and exhibit timing-sensitive neuronal activity in response to paired stimulation while leaving a decreased memory trace during retrieval. Our experiments also show that rutabaga-encoded adenylate cyclase, which mediates coincidence detection, is required for learning and short-term memory in both AB and MB. We observed additive effects when manipulating rutabaga co-expression in both structures. Together, these results implicate the AB in playing a critical role in associative olfactory learning and short-term memory.
Collapse
Affiliation(s)
| | - Fu-Yu Hsu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Lin Feng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Li-An Chu
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA; School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA; MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan; Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
48
|
Chen B, Zou J, Xie L, Cai Y, Li B, Tan W, Huang J, Li F, Xu H. WNT-inhibitory factor 1-mediated glycolysis protects photoreceptor cells in diabetic retinopathy. J Transl Med 2024; 22:245. [PMID: 38448948 PMCID: PMC10918886 DOI: 10.1186/s12967-024-05046-5] [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: 11/14/2023] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND In diabetic retinopathy (DR), hypoxia-inducible factor (HIF-1α) induces oxidative stress by upregulating glycolysis. This process leads to neurodegeneration, particularly photoreceptor cell damage, which further contributes to retinal microvascular deterioration. Further, the regulation of Wnt-inhibitory factor 1 (WIF1), a secreted Wnt signaling antagonist, has not been fully characterized in neurodegenerative eye diseases. We aimed to explore the impact of WIF1 on photoreceptor function within the context of DR. METHOD Twelve-week-old C57BL/KsJ-db/db mice were intravitreally injected with WIF1 overexpression lentivirus. After 4 weeks, optical coherence tomography (OCT), transmission electron microscopy (TEM), H&E staining, and electroretinography (ERG) were used to assess the retinal tissue and function. The potential mechanism of action of WIF1 in photoreceptor cells was explored using single-cell RNA sequencing. Under high-glucose conditions, 661 W cells were used as an in vitro DR model. WIF1-mediated signaling pathway components were assessed using quantitative real-time PCR, immunostaining, and western blotting. RESULT Typical diabetic manifestations were observed in db/db mice. Notably, the expression of WIF1 was decreased at the mRNA and protein levels. These pathological manifestations and visual function improved after WIF1 overexpression in db/db mice. TEM demonstrated that WIF1 restored damaged mitochondria, the Golgi apparatus, and photoreceptor outer segments. Moreover, ERG indicated the recovery of a-wave potential amplitude. Single-cell RNA sequencing and in vitro experiments suggested that WIF1 overexpression prevented the expression of glycolytic enzymes and lactate production by inhibiting the canonical Wnt signaling pathway, HIF-1α, and Glut1, thereby reducing retinal and cellular reactive oxygen species levels and maintaining 661 W cell viability. CONCLUSIONS WIF1 exerts an inhibitory effect on the Wnt/β-catenin-HIF-1α-Glut1 glycolytic pathway, thereby alleviating oxidative stress levels and mitigating pathological structural characteristics in retinal photoreceptor cells. This mechanism helps preserve the function of photoreceptor cells in DR and indicates that WIF1 holds promise as a potential therapeutic candidate for DR and other neurodegenerative ocular disorders.
Collapse
Affiliation(s)
- Bolin Chen
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jing Zou
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lihui Xie
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yinjun Cai
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bowen Li
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wei Tan
- Department of Ophthalmology, Xiangtan Central Hospital, Xiangtan, 411199, Hunan, China
| | - Jinhaohao Huang
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fangling Li
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huizhuo Xu
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, No 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
49
|
Matsuo Y, Kawakami A, Matsuo R. Visual afferents from an eye in the terrestrial slug Limax valentianus. J Comp Neurol 2024; 532:e25600. [PMID: 38433660 DOI: 10.1002/cne.25600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Terrestrial gastropods have a lens-bearing eye on the tip of their tentacles. There are two morphologically distinct photoreceptors, called Type-I and Type-II photoreceptors, in the retina. Type-I photoreceptors are equipped with highly developed photoreceptive microvilli in their outer rhabdomeric segment, whereas Type-II photoreceptors have short and fewer microvilli. Although both types of photoreceptors send afferent projections directly to the brain, their destinations in the brain, called optic neuropiles, have not been sufficiently investigated. Our recent studies revealed that there are commissural fibers in the cerebral ganglia that transmit photic information acquired by bilateral eyes. Moreover, some of the retinal photoreceptors are connected by gap junctions to the photosensitive brain neurons, suggesting the functional interaction of the photic information between the eye and brain photoreceptors, as well as between bilateral eyes. However, it has not been clarified which type of retinal photoreceptors send commissural projections to the contralateral hemiganglion nor interact with the brain photoreceptors. In the present study, we demonstrated by molecular histological analyses and tracer injections that (1) Type-I and Type-II photoreceptors send glutamatergic afferent projections to the medial and lateral lobes of the ipsilateral optic neuropile, respectively, (2) direct synaptic interaction between bilateral optic nerves occurs in the medial lobe of the optic neuropile, and (3) brain photosensory neurons form gap junctions with the medial lobe of the contralateral optic neuropile. These results reveal an ordered pattern of afferent projections from the retina and provide insight into the different functional roles of retinal photoreceptors.
Collapse
Affiliation(s)
- Yuko Matsuo
- Department of Environmental Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Airi Kawakami
- Department of Environmental Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| | - Ryota Matsuo
- Department of Environmental Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka, Japan
| |
Collapse
|
50
|
Grannonico M, Miller DA, Liu M, Krause MA, Savier E, Erisir A, Netland PA, Cang J, Zhang HF, Liu X. Comparative In Vivo Imaging of Retinal Structures in Tree Shrews, Humans, and Mice. eNeuro 2024; 11:ENEURO.0373-23.2024. [PMID: 38538082 PMCID: PMC10972737 DOI: 10.1523/eneuro.0373-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Rodent models, such as mice and rats, are commonly used to examine retinal ganglion cell damage in eye diseases. However, as nocturnal animals, rodent retinal structures differ from primates, imposing significant limitations in studying retinal pathology. Tree shrews (Tupaia belangeri) are small, diurnal paraprimates that exhibit superior visual acuity and color vision compared with mice. Like humans, tree shrews have a dense retinal nerve fiber layer (RNFL) and a thick ganglion cell layer (GCL), making them a valuable model for investigating optic neuropathies. In this study, we applied high-resolution visible-light optical coherence tomography to characterize the tree shrew retinal structure in vivo and compare it with that of humans and mice. We quantitatively characterize the tree shrew's retinal layer structure in vivo, specifically examining the sublayer structures within the inner plexiform layer (IPL) for the first time. Next, we conducted a comparative analysis of retinal layer structures among tree shrews, mice, and humans. We then validated our in vivo findings in the tree shrew inner retina using ex vivo confocal microscopy. The in vivo and ex vivo analyses of the shrew retina build the foundation for future work to accurately track and quantify the retinal structural changes in the IPL, GCL, and RNFL during the development and progression of human optic diseases.
Collapse
Affiliation(s)
- Marta Grannonico
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - David A Miller
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Mingna Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Michael A Krause
- Departments of Ophthalmology, University of Virginia, Charlottesville, Virginia 22904
| | - Elise Savier
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Alev Erisir
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| | - Peter A Netland
- Departments of Ophthalmology, University of Virginia, Charlottesville, Virginia 22904
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|