1
|
Tanaka R, Portugues R. On analogies in vertebrate and insect visual systems. Nat Rev Neurosci 2025:10.1038/s41583-025-00932-3. [PMID: 40410391 DOI: 10.1038/s41583-025-00932-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2025] [Indexed: 05/25/2025]
Abstract
Despite the large evolutionary distance between vertebrates and insects, the visual systems of these two taxa bear remarkable similarities that have been noted repeatedly, including by pioneering neuroanatomists such as Ramón y Cajal. Fuelled by the advent of transgenic approaches in neuroscience, studies of visual system anatomy and function in both vertebrates and insects have made dramatic progress during the past two decades, revealing even deeper analogies between their visual systems than were noted by earlier observers. Such across-taxa comparisons have tended to focus on either elementary motion detection or relatively peripheral layers of the visual systems. By contrast, the aims of this Review are to expand the scope of this comparison to pathways outside visual motion detection, as well as to deeper visual structures. To achieve these aims, we primarily discuss examples from recent work in larval zebrafish (Danio rerio) and the fruitfly (Drosophila melanogaster), a pair of genetically tractable model organisms with comparatively sized, small brains. In particular, we argue that the brains of both vertebrates and insects are equipped with third-order visual structures that specialize in shared behavioural tasks, including postural and course stabilization, approach and avoidance, and some other behaviours. These wider analogies between the two distant taxa highlight shared behavioural goals and associated evolutionary constraints and suggest that studies on vertebrate and insect vision have a lot to inspire each other.
Collapse
Affiliation(s)
- Ryosuke Tanaka
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
| | - Ruben Portugues
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Max Planck Fellow Group - Mechanisms of Cognition, MPI Psychiatry, Munich, Germany.
- Bernstein Center for Computational Neuroscience Munich, Munich, Germany.
| |
Collapse
|
2
|
Baden T, Angueyra JM, Bosten JM, Collin SP, Conway BR, Cortesi F, Dedek K, Euler T, Novales Flamarique I, Franklin A, Haverkamp S, Kelber A, Neuhauss SC, Li W, Lucas RJ, Osorio DC, Shekhar K, Tommasini D, Yoshimatsu T, Corbo JC. A standardized nomenclature for the rods and cones of the vertebrate retina. PLoS Biol 2025; 23:e3003157. [PMID: 40333813 PMCID: PMC12057980 DOI: 10.1371/journal.pbio.3003157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Vertebrate photoreceptors have been studied for well over a century, but a fixed nomenclature for referring to orthologous cell types across diverse species has been lacking. Instead, photoreceptors have been variably-and often confusingly-named according to morphology, presence/absence of 'rhodopsin', spectral sensitivity, chromophore usage, and/or the gene family of the opsin(s) they express. Here, we propose a unified nomenclature for vertebrate rods and cones that aligns with the naming systems of other retinal cell classes and that is based on the photoreceptor type's putative evolutionary history. This classification is informed by the functional, anatomical, developmental, and molecular identities of the neuron as a whole, including the expression of deeply conserved transcription factors required for development. The proposed names will be applicable across all vertebrates and indicative of the widest possible range of properties, including their postsynaptic wiring, and hence will allude to their common and species-specific roles in vision. Furthermore, the naming system is open-ended to accommodate the future discovery of as-yet unknown photoreceptor types.
Collapse
Affiliation(s)
- Tom Baden
- Sussex Neuroscience, University of Sussex, Brighton, United Kingdom
| | - Juan M. Angueyra
- Department of Biology and Brain and Behavior Institute, University of Maryland, College Park, Maryland, United States of America
| | - Jenny M. Bosten
- Sussex Neuroscience, University of Sussex, Brighton, United Kingdom
| | - Shaun P. Collin
- School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia,
| | - Bevil R. Conway
- Laboratory of Sensorimotor Research, National Eye Institute, Bethesda, Maryland, United States of America
| | - Fabio Cortesi
- Faculty of Science, School of the Environment, University of Queensland, St Lucia, Australia,
| | - Karin Dedek
- Neurosensory/Animal Navigation, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Thomas Euler
- Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | | | - Anna Franklin
- Sussex Neuroscience, University of Sussex, Brighton, United Kingdom
| | - Silke Haverkamp
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - Caesar, Bonn, Germany
| | - Almut Kelber
- Department of Biology, Lund University, Lund, Sweden
| | | | - Wei Li
- National Eye Institute, Bethesda, Maryland, United States of America
| | - Robert J. Lucas
- Centre for Biological Timing and Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Daniel C. Osorio
- Sussex Neuroscience, University of Sussex, Brighton, United Kingdom
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, California, United States of America
| | - Dario Tommasini
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, California, United States of America
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph C. Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
3
|
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
|
4
|
Günther A, Balaji V, Leberecht B, Forst JJ, Rotov AY, Woldt T, Abdulazhanova D, Mouritsen H, Dedek K. Morphology and connectivity of retinal horizontal cells in two avian species. Front Cell Neurosci 2025; 19:1558605. [PMID: 40103750 PMCID: PMC11914121 DOI: 10.3389/fncel.2025.1558605] [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: 01/10/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
In the outer vertebrate retina, the visual signal is separated into intensity and wavelength information. In birds, seven types of photoreceptors (one rod, four single cones, and two members of the double cone) mediate signals to >20 types of second-order neurons, the bipolar cells and horizontal cells. Horizontal cells contribute to color and contrast processing by providing feedback signals to photoreceptors and feedforward signals to bipolar cells. In fish, reptiles, and amphibians they either encode intensity or show color-opponent responses. Yet, for the bird retina, the number of horizontal cell types is not fully resolved and even more importantly, the synapses between photoreceptors and horizontal cells have never been quantified for any bird species. With a combination of light microscopy and serial EM reconstructions, we found four different types of horizontal cells in two distantly related species, the domestic chicken and the European robin. In agreement with some earlier studies, we confirmed two highly abundant cell types (H1, H2) and two rare cell types (H3, H4), of which H1 is an axon-bearing cell, whereas H2-H4 are axonless. H1 cells made chemical synapses with one type of bipolar cell and an interplexiform amacrine cell at their soma. Dendritic contacts of H1-H4 cells to photoreceptors were type-specific and similar to the turtle retina, which confirms the high degree of evolutionary conservation in the vertebrate outer retina. Our data further suggests that H1 and potentially H2 cells may encode intensity, whereas H3 and H4 may represent color opponent horizontal cells which may contribute to the birds' superb color and/or high acuity vision.
Collapse
Affiliation(s)
- Anja Günther
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany
| | - Vaishnavi Balaji
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Bo Leberecht
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Julia J Forst
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Alexander Y Rotov
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Tobias Woldt
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Dinora Abdulazhanova
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
| | - Henrik Mouritsen
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Neurosensory/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität, Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
5
|
Budoff SA, Poleg-Polsky A. A Complete Spatial Map of Mouse Retinal Ganglion Cells Reveals Density and Gene Expression Specializations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637538. [PMID: 39990332 PMCID: PMC11844403 DOI: 10.1101/2025.02.10.637538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information from the eye to the brain. In mice, several RGC subtypes show nonuniform spatial distributions, potentially mediating specific visual functions. However, the full extent of RGC specialization remains unknown. Here, we used en-face cryosectioning, spatial transcriptomics, and machine learning to map the spatial distribution of all RGC subtypes identified in previous single-cell studies. While two-thirds of RGC subtypes were evenly distributed, others showed strong biases toward ventral or dorso-temporal regions associated with sky vision and the area retinae temporalis (ART), the predicted homolog of the area centralis. Additionally, we observed unexpected spatial variation in gene expression within several subtypes along the dorso-ventral axis or within vs. outside the ART, independent of RGC density profiles. Finally, we found limited correlations between the gene profiles of the ART and the primate macula, suggesting divergent specialization between the mouse and primate central vision.
Collapse
Affiliation(s)
- Samuel A. Budoff
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| | - Alon Poleg-Polsky
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| |
Collapse
|
6
|
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
|
7
|
Lawson E, Khan A, Wessels Q, Taylor AM. Congenital colour vision deficiency in healthcare professionals: a scoping review protocol of the impact on clinical practice and patient safety. BMJ Open 2025; 15:e088563. [PMID: 39832987 PMCID: PMC11748764 DOI: 10.1136/bmjopen-2024-088563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Congenital colour vision deficiency (CVD), known as colour blindness, is a common visual problem affecting around 1 in 12 men and 1 in 200 women. It is known that people who have red-green CVD, the most common phenotype, can have difficulty differentiating colours and this can impact the ability to perform clinical tasks related to patient care. The objective of this scoping review is to understand the extent and type of evidence and the impact on clinical practice and patient safety arising from congenital CVD in healthcare professionals. METHODS AND ANALYSIS The scoping review will follow the methodological framework outlined by the Joanna Briggs Institute and by Arksey and O'Malley and we will adhere to the Preferred Reporting Items for Systematic reviews and Meta-Analyses Scoping Reviews checklist. The following databases will be searched: PubMed, MEDLINE, Web of Science, Scopus, Lens.org and the Trip medical database (filtered for low-income countries) with no limit on earliest date and up to 30 November 2024. Grey literature will be identified by planned searches in the Overton Index. All articles related to 'CVD in healthcare professionals in clinical practice or in training' will be included. The study will also include other professionals who may be involved in healthcare but are not involved in direct patient-facing activities. It will be limited to congenital CVD and will exclude the impact of visual impairment and acquired CVD.There will be an initial search to validate the search strategy. Titles and abstracts will be screened to determine eligibility, and the full text will be reviewed using a data extraction framework. Data will be extracted, collated and then mapped and summarised to present the relevant key findings and outcomes from the papers in tabular and visualised form with a narrative synthesis. ETHICS AND DISSEMINATION The scoping review does not need ethical approval and will provide an overview of the impact of congenital CVD on clinical practice and patient safety. This will determine the future need and direction of research in this area and identify methodological challenges and opportunities. The results will be published open access in a peer-reviewed journal. REGISTRATION INPLASY (DOI: 10.37766/inplasy2024.11.0099).
Collapse
Affiliation(s)
- Euan Lawson
- Lancaster Medical School, Lancaster University, Lancaster, UK
| | - Aqib Khan
- Lancaster Medical School, Lancaster University, Lancaster, UK
| | | | - Adam M Taylor
- Lancaster Medical School, Lancaster University, Lancaster, UK
| |
Collapse
|
8
|
Nguyen MN, Chakraborty D, Messinger J, Sherry DM, Fliesler SJ, Pittler SJ. Modeling Retinitis Pigmentosa 59: Dhdds T206A and Dhdds K42E knock-in mutant mice are phenotypically similar. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632845. [PMID: 39868263 PMCID: PMC11761419 DOI: 10.1101/2025.01.13.632845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
D e h ydro d olichyl d iphosphate s ynthase (DHDDS) is an essential enzyme required for several forms of protein glycosylation in all eukaryotic cells. Surprisingly, three mutant alleles, ( Dhdds K42E/K42E (K42E/K42E), Dhdds T206A/K42E (T206A/K42E), and found in only one patient, Dhdds R98W/K42E (R98W/K42E) have been reported that cause non-syndromic retinitis pigmentosa (RP59), an inherited retinal degeneration (IRD). Because T206A was only observed heterozygously with the K42E allele in RP59 patients, we used CRISPR/CAS9 technology to generate T206A/T206A, and subsequently T206A/K42E alleles in mice to assess the contribution of the T206A allele to the disease phenotype, to model the human disease, and to compare resulting phenotypes to our homozygous K42E mouse model. By postnatal (PN) 12-mo, T206A/K42E mice exhibit significant reduction of inner nuclear layer thickness as was observed in K42E/K42E mice. No change in outer nuclear layer thickness is observed in all mutant phenotypes up to PN 12 mo. Electroretinography (ERG) showed a significantly reduced b-wave without a-wave decrement and by PN 3-mo, ERG c- and d-wave responses were significantly attenuated in all phenotypes. Consistent with a reduction in inner nuclear layer thickness seen by OCT and cell loss observed by histology, bipolar and amacrine cell densities were reduced in all Dhdds mutant phenotypes compared to PN 8-12 mo age-matched controls. These results indicate that the DHDDS T206A allele causes retinal disease independent of the K42E allele, and that there likely is a common disease mechanism involving RP59-associated DHDDS mutations. We conclude that the physiological basis of retinal dysfunction in RP59 involves defective signaling in the inner retina resulting in bipolar/amacrine cell degeneration.
Collapse
|
9
|
Famiglietti EV. Mammalian Retinal Bipolar Cells: Morphological Identification and Systematic Classification in Rabbit Retina with a Comparative Perspective. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613998. [PMID: 39345639 PMCID: PMC11429971 DOI: 10.1101/2024.09.19.613998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Retinal bipolar cells (BCs) convey visual signals from photoreceptors to more than 50 types of rabbit retinal ganglion cells (Famiglietti, 2020). More than 40 years ago, 10-11 types of bipolar cell were recognized in rabbit and cat retinas (Famiglietti, 1981). Twenty years later 10 were identified in mouse, rat, and monkey, while recent molecular genetic studies indicate that there are 15 types of bipolar cell in mouse retina (Shekhar et al., 2016). The present detailed study of more than 800 bipolar cells in ten Golgi-impregnated rabbit retinas indicates that there are 14-16 types of cone bipolar cell and one type of rod bipolar cell in rabbit retina. These have been carefully analyzed in terms of dendritic and axonal morphology, and axon terminal stratification with respect to fiducial starburst amacrine cells. In fortuitous proximity, several types of bipolar cell can be related to identified ganglion cells by stratification and by contacts suggestive of synaptic connection. These results are compared with other studies of rabbit bipolar cells. Homologies with bipolar cells of mouse and monkey are considered in functional terms.
Collapse
|
10
|
James RE, Hamilton NR, Huffman LN, Brown MP, Neckles VN, Pasterkamp RJ, Goff LA, Kolodkin AL. Retinal ganglion cell-derived semaphorin 6A segregates starburst amacrine cell dendritic scaffolds to organize the mouse inner retina. Development 2024; 151:dev204293. [PMID: 39495936 PMCID: PMC11634039 DOI: 10.1242/dev.204293] [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: 08/01/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024]
Abstract
To form functional circuits, neurons must settle in their appropriate cellular locations, and then project and elaborate neurites to contact their target synaptic neuropils. Laminar organization within the vertebrate retinal inner plexiform layer (IPL) facilitates pre- and postsynaptic neurite targeting, yet the precise mechanisms underlying establishment of functional IPL subdomains are not well understood. Here, we explore mechanisms defining the compartmentalization of OFF and ON neurites generally, and OFF and ON direction-selective neurites specifically, within the developing mouse IPL. We show that semaphorin 6A (Sema6A), a repulsive axon guidance cue, is required for delineation of OFF versus ON circuits within the IPL: in the Sema6a null IPL, the boundary between OFF and ON domains is blurred. Furthermore, Sema6A expressed by retinal ganglion cells (RGCs) directs laminar segregation of OFF and ON starburst amacrine cell dendritic scaffolds, which themselves serve as a substrate upon which other retinal neurites elaborate. These results demonstrate that RGCs, the first type of neuron born within the retina, play an active role in functional specialization of the IPL.
Collapse
Affiliation(s)
- Rebecca E. James
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Natalie R. Hamilton
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Lola Nicole Huffman
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Matthew P. Brown
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Victoria N. Neckles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Loyal A. Goff
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
11
|
Ebrahimi MN, Banazadeh M, Alitaneh Z, Jaafari Suha A, Esmaeili A, Hasannejad-Asl B, Siahposht-Khachaki A, Hassanshahi A, Bagheri-Mohammadi S. The distribution of neurotransmitters in the brain circuitry: Mesolimbic pathway and addiction. Physiol Behav 2024; 284:114639. [PMID: 39004195 DOI: 10.1016/j.physbeh.2024.114639] [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: 04/23/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Understanding the central nervous system (CNS) circuitry and its different neurotransmitters that underlie reward is essential to improve treatment for many common health issues, such as addiction. Here, we concentrate on understanding how the mesolimbic circuitry and neurotransmitters are organized and function, and how drug exposure affects synaptic and structural changes in this circuitry. While the role of some reward circuits, like the cerebral dopamine (DA)/glutamate (Glu)/gamma aminobutyric acid (GABA)ergic pathways, in drug reward, is well known, new research using molecular-based methods has shown functional alterations throughout the reward circuitry that contribute to various aspects of addiction, including craving and relapse. A new understanding of the fundamental connections between brain regions as well as the molecular alterations within these particular microcircuits, such as neurotrophic factor and molecular signaling or distinct receptor function, that underlie synaptic and structural plasticity evoked by drugs of abuse has been made possible by the ability to observe and manipulate neuronal activity within specific cell types and circuits. It is exciting that these discoveries from preclinical animal research are now being applied in the clinic, where therapies for human drug dependence, such as deep brain stimulation and transcranial magnetic stimulation, are being tested. Therefore, this chapter seeks to summarize the current understanding of the important brain regions (especially, mesolimbic circuitry) and neurotransmitters implicated in drug-related behaviors and the molecular mechanisms that contribute to altered connectivity between these areas, with the postulation that increased knowledge of the plasticity within the drug reward circuit will lead to new and improved treatments for addiction.
Collapse
Affiliation(s)
- Mohammad Navid Ebrahimi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Alitaneh
- Quantitative and System Biology, Department of Natural Sciences, University of California Merced, USA
| | - Ali Jaafari Suha
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Esmaeili
- Student Research Committee, Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Hasannejad-Asl
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti, University of Medical Sciences, Tehran, Iran
| | - Ali Siahposht-Khachaki
- Immunogenetics Research Center, Department of Physiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amin Hassanshahi
- Department of Physiology, Bam University of Medical Sciences, Bam, Iran
| | - Saeid Bagheri-Mohammadi
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Günther A, Haverkamp S, Irsen S, Watkins PV, Dedek K, Mouritsen H, Briggman KL. Species-specific circuitry of double cone photoreceptors in two avian retinas. Commun Biol 2024; 7:992. [PMID: 39143253 PMCID: PMC11325025 DOI: 10.1038/s42003-024-06697-2] [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: 03/10/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
In most avian retinas, double cones (consisting of a principal and accessory member) outnumber other photoreceptor types and have been associated with various functions, such as encoding luminance, sensing polarized light, and magnetoreception. However, their down-stream circuitry is poorly understood, particularly across bird species. Analysing species differences is important to understand changes in circuitry driven by ecological adaptations. We compare the ultrastructure of double cones and their postsynaptic bipolar cells between a night-migratory European robin and non-migratory chicken. We discover four previously unidentified bipolar cell types in the European robin retina, including midget-like bipolar cells mainly connected to one principal member. A downstream ganglion cell reveals a complete midget-like circuit similar to a circuit in the peripheral primate retina. Additionally, we identify a selective circuit transmitting information from a specific subset of accessory members. Our data highlight species-specific differences in double cone to bipolar cell connectivity, potentially reflecting ecological adaptations.
Collapse
Affiliation(s)
- Anja Günther
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior-caesar, Bonn, Germany.
| | - Silke Haverkamp
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior-caesar, Bonn, Germany
| | - Stephan Irsen
- Electron Microscopy and Analytics, Max Planck Institute for Neurobiology of Behavior-caesar, Bonn, Germany
| | - Paul V Watkins
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior-caesar, Bonn, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Straße 9-11, Oldenburg, Germany
- Research Centre for Neurosensory Sciences, Carl von Ossietzky University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, Oldenburg, Germany
| | - Henrik Mouritsen
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Straße 9-11, Oldenburg, Germany
- Research Centre for Neurosensory Sciences, Carl von Ossietzky University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, Oldenburg, Germany
| | - Kevin L Briggman
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior-caesar, Bonn, Germany.
| |
Collapse
|
15
|
He L, Wang W, Ma L, Huang T. Optimization-Based Pairwise Interaction Point Process (O-PIPP): A Precise and Universal Retinal Mosaic Modeling Approach. Invest Ophthalmol Vis Sci 2024; 65:39. [PMID: 39042401 PMCID: PMC11268446 DOI: 10.1167/iovs.65.8.39] [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: 09/05/2023] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Purpose A retinal mosaic, the spatial organization of a population of homotypic neurons, is thought to sample a specific visual feature into the feedforward visual pathway. The purpose of this study was to propose a universal modeling approach for precisely generating retinal mosaics and overcoming the limitations of previous models, especially in modeling abnormal mosaic patterns under disease conditions. Methods Here, we developed the optimization-based pairwise interaction point process (O-PIPP). It incorporates optimization techniques into previous simulation approaches, enabling directional control of the simulation process according to the user-designed optimization target. For the convenience of the community, we implemented the O-PIPP approach into a Python package and a website application. Results We showed that the O-PIPP can generate more precise neural spatial patterns of healthy and diseased mosaics compared to previous phenomenological approaches. Notably, through modeling the retinal neural circuitry with O-PIPP-simulated retinitis pigmentosa cone mosaics, we elucidated how the cone mosaic rearrangement impacted the information processing of ganglion cells. Conclusions The O-PIPP provides a precise and universal tool to simulate realistic mosaics, which could help to investigate the function of retinal mosaics in vision.
Collapse
Affiliation(s)
- Liuyuan He
- National Key Laboratory for Multimedia Information Processing, School of Computer Science, Peking University, Beijing, China
- National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, China
| | - Wenyao Wang
- National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, China
| | - Lei Ma
- National Key Laboratory for Multimedia Information Processing, School of Computer Science, Peking University, Beijing, China
- National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, China
| | - Tiejun Huang
- National Key Laboratory for Multimedia Information Processing, School of Computer Science, Peking University, Beijing, China
- National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing, China
- Beijing Academy of Artificial Intelligence, Beijing, China
| |
Collapse
|
16
|
Hanke-Gogokhia C, Zapadka TE, Finkelstein S, Klingeborn M, Maugel TK, Singer JH, Arshavsky VY, Demb JB. The Structural and Functional Integrity of Rod Photoreceptor Ribbon Synapses Depends on Redundant Actions of Dynamins 1 and 3. J Neurosci 2024; 44:e1379232024. [PMID: 38641407 PMCID: PMC11209669 DOI: 10.1523/jneurosci.1379-23.2024] [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: 07/21/2023] [Revised: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
Vertebrate vision begins with light absorption by rod and cone photoreceptors, which transmit signals from their synaptic terminals to second-order neurons: bipolar and horizontal cells. In mouse rods, there is a single presynaptic ribbon-type active zone at which the release of glutamate occurs tonically in the dark. This tonic glutamatergic signaling requires continuous exo- and endocytosis of synaptic vesicles. At conventional synapses, endocytosis commonly requires dynamins: GTPases encoded by three genes (Dnm1-3), which perform membrane scission. Disrupting endocytosis by dynamin deletions impairs transmission at conventional synapses, but the impact of disrupting endocytosis and the role(s) of specific dynamin isoforms at rod ribbon synapses are understood incompletely. Here, we used cell-specific knock-outs (KOs) of the neuron-specific Dnm1 and Dnm3 to investigate the functional roles of dynamin isoforms in rod photoreceptors in mice of either sex. Analysis of synaptic protein expression, synapse ultrastructure, and retinal function via electroretinograms (ERGs) showed that dynamins 1 and 3 act redundantly and are essential for supporting the structural and functional integrity of rod ribbon synapses. Single Dnm3 KO showed no phenotype, and single Dnm1 KO only modestly reduced synaptic vesicle density without affecting vesicle size and overall synapse integrity, whereas double Dnm1/Dnm3 KO impaired vesicle endocytosis profoundly, causing enlarged vesicles, reduced vesicle density, reduced ERG responses, synaptic terminal degeneration, and disassembly and degeneration of postsynaptic processes. Concurrently, cone function remained intact. These results show the fundamental redundancy of dynamins 1 and 3 in regulating the structure and function of rod ribbon synapses.
Collapse
Affiliation(s)
- Christin Hanke-Gogokhia
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
| | - Thomas E Zapadka
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
- Cellular & Molecular Physiology, Yale University, New Haven, Connecticut 06511
| | - Stella Finkelstein
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Timothy K Maugel
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Jonathan B Demb
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
- Cellular & Molecular Physiology, Yale University, New Haven, Connecticut 06511
- Department of Neuroscience, Yale University, New Haven, Connecticut 06511
- Wu Tsai Institute, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
17
|
Hellevik AM, Mardoum P, Hahn J, Kölsch Y, D'Orazi FD, Suzuki SC, Godinho L, Lawrence O, Rieke F, Shekhar K, Sanes JR, Baier H, Baden T, Wong RO, Yoshimatsu T. Ancient origin of the rod bipolar cell pathway in the vertebrate retina. Nat Ecol Evol 2024; 8:1165-1179. [PMID: 38627529 DOI: 10.1038/s41559-024-02404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/20/2024] [Indexed: 04/30/2024]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions. The specialized downstream circuit for rod signalling, called the primary rod pathway, is well characterized in mammals, but circuitry for rod signalling in non-mammals is largely unknown. Here we demonstrate that the mammalian primary rod pathway is conserved in zebrafish, which diverged from extant mammals ~400 million years ago. Using single-cell RNA sequencing, we identified two bipolar cell types in zebrafish that are related to mammalian rod bipolar cell (RBCs), the only bipolar type that directly carries rod signals from the outer to the inner retina in the primary rod pathway. By combining electrophysiology, histology and ultrastructural reconstruction of the zebrafish RBCs, we found that, similar to mammalian RBCs, both zebrafish RBC types connect with all rods in their dendritic territory and provide output largely onto amacrine cells. The wiring pattern of the amacrine cells postsynaptic to one RBC type is strikingly similar to that of mammalian RBCs and their amacrine partners, suggesting that the cell types and circuit design of the primary rod pathway emerged before the divergence of teleost fish and mammals. The second RBC type, which forms separate pathways, was either lost in mammals or emerged in fish.
Collapse
Affiliation(s)
- Ayana M Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Philip Mardoum
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA, USA
| | - Yvonne Kölsch
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Florence D D'Orazi
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sachihiro C Suzuki
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Owen Lawrence
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Vision Science Center, University of Washington, Seattle, WA, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Herwig Baier
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO, USA.
- BioRTC, Yobe State University, Damatsuru, Yobe, Nigeria.
| |
Collapse
|
18
|
Friedrichsen K, Hsiang JC, Lin CI, McCoy L, Valkova K, Kerschensteiner D, Morgan JL. Subcellular pathways through VGluT3-expressing mouse amacrine cells provide locally tuned object-motion-selective signals in the retina. Nat Commun 2024; 15:2965. [PMID: 38580652 PMCID: PMC10997783 DOI: 10.1038/s41467-024-46996-0] [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: 06/19/2023] [Accepted: 03/15/2024] [Indexed: 04/07/2024] Open
Abstract
VGluT3-expressing mouse retinal amacrine cells (VG3s) respond to small-object motion and connect to multiple types of bipolar cells (inputs) and retinal ganglion cells (RGCs, outputs). Because these input and output connections are intermixed on the same dendrites, making sense of VG3 circuitry requires comparing the distribution of synapses across their arbors to the subcellular flow of signals. Here, we combine subcellular calcium imaging and electron microscopic connectomic reconstruction to analyze how VG3s integrate and transmit visual information. VG3s receive inputs from all nearby bipolar cell types but exhibit a strong preference for the fast type 3a bipolar cells. By comparing input distributions to VG3 dendrite responses, we show that VG3 dendrites have a short functional length constant that likely depends on inhibitory shunting. This model predicts that RGCs that extend dendrites into the middle layers of the inner plexiform encounter VG3 dendrites whose responses vary according to the local bipolar cell response type.
Collapse
Affiliation(s)
- Karl Friedrichsen
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Graduate Program in Neuroscience, Washington University in St. Louis, St. Louis, USA
| | - Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Graduate Program in Neuroscience, Washington University in St. Louis, St. Louis, USA
| | - Chin-I Lin
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Graduate Program in Neuroscience, Washington University in St. Louis, St. Louis, USA
| | - Liam McCoy
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Katia Valkova
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| | - Josh L Morgan
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
19
|
Hsiang JC, Shen N, Soto F, Kerschensteiner D. Distributed feature representations of natural stimuli across parallel retinal pathways. Nat Commun 2024; 15:1920. [PMID: 38429280 PMCID: PMC10907388 DOI: 10.1038/s41467-024-46348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/22/2024] [Indexed: 03/03/2024] Open
Abstract
How sensory systems extract salient features from natural environments and organize them across neural pathways is unclear. Combining single-cell and population two-photon calcium imaging in mice, we discover that retinal ON bipolar cells (second-order neurons of the visual system) are divided into two blocks of four types. The two blocks distribute temporal and spatial information encoding, respectively. ON bipolar cell axons co-stratify within each block, but separate laminarly between them (upper block: diverse temporal, uniform spatial tuning; lower block: diverse spatial, uniform temporal tuning). ON bipolar cells extract temporal and spatial features similarly from artificial and naturalistic stimuli. In addition, they differ in sensitivity to coherent motion in naturalistic movies. Motion information is distributed across ON bipolar cells in the upper and the lower blocks, multiplexed with temporal and spatial contrast, independent features of natural scenes. Comparing the responses of different boutons within the same arbor, we find that axons of all ON bipolar cell types function as computational units. Thus, our results provide insights into the visual feature extraction from naturalistic stimuli and reveal how structural and functional organization cooperate to generate parallel ON pathways for temporal and spatial information in the mammalian retina.
Collapse
Affiliation(s)
- Jen-Chun Hsiang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Florentina Soto
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
20
|
James RE, Hamilton NR, Huffman LN, Pasterkamp J, Goff LA, Kolodkin AL. Semaphorin 6A in Retinal Ganglion Cells Regulates Functional Specialization of the Inner Retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.18.567662. [PMID: 38014224 PMCID: PMC10680864 DOI: 10.1101/2023.11.18.567662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
To form functional circuits, neurons must settle in their appropriate cellular locations and then project and elaborate neurites to contact their target synaptic neuropils. Laminar organization within the vertebrate retinal inner plexiform layer (IPL) facilitates pre- and postsynaptic neurite targeting, yet, the precise mechanisms underlying establishment of functional IPL subdomains are not well understood. Here we explore mechanisms defining the compartmentalization of OFF and ON neurites generally, and OFF and ON direction-selective neurites specifically, within the developing IPL. We show that semaphorin 6A (Sema6A), a repulsive axon guidance cue, is required for delineation of OFF versus ON circuits within the IPL: in the Sema6a null IPL, the boundary between OFF and ON domains is blurred. Furthermore, Sema6A expressed by retinal ganglion cells (RGCs) directs laminar segregation of OFF and ON starburst amacrine cell (SAC) dendritic scaffolds, which themselves serve as a substrate upon which other retinal neurites elaborate. These results demonstrate for the first time that RGCs, the first neuron-type born within the retina, play an active role in functional specialization of the IPL. Retinal ganglion cell-dependent regulation of OFF and ON starburst amacrine cell dendritic scaffold segregation prevents blurring of OFF versus ON functional domains in the murine inner plexiform layer.
Collapse
|
21
|
Saha A, Zuniga J, Mian K, Zhai H, Derr PJ, Hoon M, Sinha R. Regional variation in the organization and connectivity of the first synapse in the primate night vision pathway. iScience 2023; 26:108113. [PMID: 37915604 PMCID: PMC10616377 DOI: 10.1016/j.isci.2023.108113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/25/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Sensitivity of primate daylight vision varies across the visual field. This is attributed to regional variations in cone photoreceptor density and synaptic connectivity of the underlying circuitry. In contrast, we have limited understanding of how synapse organization of the primate night vision pathway changes across space. Using serial electron microscopy, we reconstructed the first synapse of the night vision pathway between rod photoreceptors and second-order neurons, at multiple locations from the central part of the primate retina, fovea, to the periphery. We find that most facets of the rod synapse connectivity vary across retinal regions. However, rod synaptic divergence and convergence patterns do not change in the same manner across locations. Moreover, patterns of rod synapse organization are tightly correlated with photoreceptor density. Such regional heterogeneities revise the connectivity diagram of the primate rod synapse which will shape synapse function and sensitivity of the night vision pathway across visual space.
Collapse
Affiliation(s)
- Aindrila Saha
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
| | - Juan Zuniga
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Kainat Mian
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Haoshen Zhai
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Paul J. Derr
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - Mrinalini Hoon
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| | - Raunak Sinha
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
22
|
Wu SR, Zoghbi HY. The Atoh1-Cre Knock-In Allele Ectopically Labels a Subpopulation of Amacrine Cells and Bipolar Cells in Mouse Retina. eNeuro 2023; 10:ENEURO.0307-23.2023. [PMID: 37923392 PMCID: PMC10626521 DOI: 10.1523/eneuro.0307-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
The retina has diverse neuronal cell types derived from a common pool of retinal progenitors. Many molecular drivers, mostly transcription factors, have been identified to promote different cell fates. In Drosophila, atonal is required for specifying photoreceptors. In mice, there are two closely related atonal homologs, Atoh1 and Atoh7 While Atoh7 is known to promote the genesis of retinal ganglion cells, there is no study on the function of Atoh1 in retinal development. Here, we crossed Atoh1Cre/+ mice to mice carrying a Cre-dependent TdTomato reporter to track potential Atoh1-lineage neurons in retinas. We characterized a heterogeneous group of TdTomato+ retinal neurons that were detected at the postnatal stage, including glutamatergic amacrine cells, AII amacrine cells, and BC3b bipolar cells. Unexpectedly, we did not observe TdTomato+ retinal neurons in the mice with an Atoh1-FlpO knock-in allele and a Flp-dependent TdTomato reporter, suggesting Atoh1 is not expressed in the mouse retina. Consistent with these data, conditional removal of Atoh1 in the retina did not cause any observable phenotypes. Importantly, we did not detect Atoh1 expression in the retina at multiple ages using mice with Atoh1-GFP knock-in allele. Therefore, we conclude that Atoh1Cre/+ mice have ectopic Cre expression in the retina and that Atoh1 is not required for retinal development.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
| | - Huda Y Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
23
|
Hellevik AM, Mardoum P, Hahn J, Kölsch Y, D’Orazi FD, Suzuki SC, Godinho L, Lawrence O, Rieke F, Shekhar K, Sanes JR, Baier H, Baden T, Wong RO, Yoshimatsu T. Ancient origin of the rod bipolar cell pathway in the vertebrate retina. RESEARCH SQUARE 2023:rs.3.rs-3411693. [PMID: 37886445 PMCID: PMC10602083 DOI: 10.21203/rs.3.rs-3411693/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions. Mammals have a specialized downstream circuit for rod signaling called the primary rod pathway, which comprises specific cell types and wiring patterns that are thought to be unique to this lineage. Thus, it has been long assumed that the primary rod pathway evolved in mammals. Here, we challenge this view by demonstrating that the mammalian primary rod pathway is conserved in zebrafish, which diverged from extant mammals ~400 million years ago. Using single-cell RNA-sequencing, we identified two bipolar cell (BC) types in zebrafish that are related to mammalian rod BCs (RBCs) of the primary rod pathway. By combining electrophysiology, histology, and ultrastructural reconstruction of the zebrafish RBCs, we found that, like mammalian RBCs, both zebrafish RBC types connect with all rods in their dendritic territory, and provide output largely onto amacrine cells. The wiring pattern of the amacrine cells post-synaptic to one RBC type is strikingly similar to that of mammalian RBCs, suggesting that the cell types and circuit design of the primary rod pathway have emerged before the divergence of teleost fish and amniotes. The second RBC type, which forms separate pathways, is either lost in mammals or emerged in fish.
Collapse
Affiliation(s)
- Ayana M Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Philip Mardoum
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
| | - Yvonne Kölsch
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Florence D D’Orazi
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Sachihiro C. Suzuki
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, 80802 Munich, Germany
| | - Owen Lawrence
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
- Vision Science Center, University of Washington, Seattle, WA 98195, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Joshua R Sanes
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology & Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
- BioRTC, Yobe State University, Damatsuru, Yobe 620101, Nigeria
| |
Collapse
|
24
|
Hellevik AM, Mardoum P, Hahn J, Kölsch Y, D’Orazi FD, Suzuki SC, Godinho L, Lawrence O, Rieke F, Shekhar K, Sanes JR, Baier H, Baden T, Wong RO, Yoshimatsu T. Ancient origin of the rod bipolar cell pathway in the vertebrate retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557433. [PMID: 37771914 PMCID: PMC10525478 DOI: 10.1101/2023.09.12.557433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Vertebrates rely on rod photoreceptors for vision in low-light conditions1. Mammals have a specialized downstream circuit for rod signaling called the primary rod pathway, which comprises specific cell types and wiring patterns that are thought to be unique to this lineage2-6. Thus, it has been long assumed that the primary rod pathway evolved in mammals3,5-7. Here, we challenge this view by demonstrating that the mammalian primary rod pathway is conserved in zebrafish, which diverged from extant mammals ~400 million years ago. Using single-cell RNA-sequencing, we identified two bipolar cell (BC) types in zebrafish that are related to mammalian rod BCs (RBCs) of the primary rod pathway. By combining electrophysiology, histology, and ultrastructural reconstruction of the zebrafish RBCs, we found that, like mammalian RBCs8, both zebrafish RBC types connect with all rods and red-cones in their dendritic territory, and provide output largely onto amacrine cells. The wiring pattern of the amacrine cells post-synaptic to one RBC type is strikingly similar to that of mammalian RBCs. This suggests that the cell types and circuit design of the primary rod pathway may have emerged before the divergence of teleost fish and amniotes (mammals, bird, reptiles). The second RBC type in zebrafish, which forms separate pathways from the first RBC type, is either lost in mammals or emerged in fish to serve yet unknown roles.
Collapse
Affiliation(s)
- Ayana M Hellevik
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Philip Mardoum
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
| | - Yvonne Kölsch
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Florence D D’Orazi
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Sachihiro C. Suzuki
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, 80802 Munich, Germany
| | - Owen Lawrence
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
- Vision Science Center, University of Washington, Seattle, WA 98195, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Vision Sciences Graduate Program; California Institute of Quantitative Biosciences (QB3), University of California Berkley, Berkeley, CA 94720, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Joshua R Sanes
- Department of Molecular & Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Department Genes – Circuits – Behavior, 82152 Martinsried, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, 72076, Germany
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology & Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
- BioRTC, Yobe State University, Damatsuru, Yobe 620101, Nigeria
| |
Collapse
|
25
|
Abstract
Because the central nervous system is largely nonrenewing, neurons and their synapses must be maintained over the lifetime of an individual to ensure circuit function. Age is a dominant risk factor for neural diseases, and declines in nervous system function are a common feature of aging even in the absence of disease. These alterations extend to the visual system and, in particular, to the retina. The retina is a site of clinically relevant age-related alterations but has also proven to be a uniquely approachable system for discovering principles that govern neural aging because it is well mapped, contains diverse neuron types, and is experimentally accessible. In this article, we review the structural and molecular impacts of aging on neurons within the inner and outer retina circuits. We further discuss the contribution of non-neuronal cell types and systems to retinal aging outcomes. Understanding how and why the retina ages is critical to efforts aimed at preventing age-related neural decline and restoring neural function.
Collapse
Affiliation(s)
- Jeffrey D Zhu
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Sharma Pooja Tarachand
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Qudrat Abdulwahab
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
26
|
Ichinose T, Hellmer CB, Bohl JM. Presynaptic depolarization differentially regulates dual neurotransmitter release from starburst amacrine cells in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1225824. [PMID: 38444728 PMCID: PMC10914334 DOI: 10.3389/fopht.2023.1225824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The retina is comprised of diverse neural networks, signaling from photoreceptors to ganglion cells to encode images. The synaptic connections between these retinal neurons are crucial points for information transfer; however, the input-output relations of many synapses are understudied. Starburst amacrine cells in the retina are known to contribute to retinal motion detection circuits, providing a unique window for understanding neural computations. We examined the dual transmitter release of GABA and acetylcholine from starburst amacrine cells by optogenetic activation of these cells, and conducted patch clamp recordings from postsynaptic ganglion cells to record excitatory and inhibitory postsynaptic currents (EPSCs and IPSCs). As starburst amacrine cells exhibit distinct kinetics in response to objects moving in a preferred or null direction, we mimicked their depolarization kinetics using optogenetic stimuli by varying slopes of the rising phase. The amplitudes of EPSCs and IPSCs in postsynaptic ganglion cells were reduced as the stimulus rising speed was prolonged. However, the sensitivity of postsynaptic currents to the stimulus slope differed. EPSC amplitudes were consistently reduced as the steepness of the rising phase fell. By contrast, IPSCs were less sensitive to the slope of the stimulus rise phase and maintained their amplitudes until the slope became shallow. These results indicate that distinct synaptic release mechanisms contribute to acetylcholine and GABA release from starburst amacrine cells, which could contribute to the ganglion cells' direction selectivity.
Collapse
Affiliation(s)
- Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | | | - Jeremy M. Bohl
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
27
|
Keeley PW, Patel SS, Reese BE. Cell numbers, cell ratios, and developmental plasticity in the rod pathway of the mouse retina. J Anat 2023; 243:204-222. [PMID: 35292986 PMCID: PMC10335380 DOI: 10.1111/joa.13653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
The precise specification of cellular fate is thought to ensure the production of the correct number of neurons within a population. Programmed cell death may be an additional mechanism controlling cell number, believed to refine the proper ratio of pre- to post-synaptic neurons for a given species. Here, we consider the size of three different neuronal populations in the rod pathway of the mouse retina: rod photoreceptors, rod bipolar cells, and AII amacrine cells. Across a collection of 28 different strains of mice, large variation in the numbers of all three cell types is present. The variation in their numbers is not correlated, so that the ratio of rods to rod bipolar cells, as well as rod bipolar cells to AII amacrine cells, varies as well. Establishing connectivity between such variable pre- and post-synaptic populations relies upon plasticity that modulates process outgrowth and morphological differentiation, which we explore experimentally for both rod bipolar and AII amacrine cells in a mouse retina with elevated numbers of each cell type. While both rod bipolar dendritic and axonal arbors, along with AII lobular arbors, modulate their areal size in relation to local homotypic cell densities, the dendritic appendages of the AII amacrine cells do not. Rather, these processes exhibit a different form of plasticity, regulating the branching density of their overlapping arbors. Each form of plasticity should ensure uniformity in retinal coverage in the presence of the independent specification of afferent and target cell number.
Collapse
Affiliation(s)
- Patrick W. Keeley
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Shivam S. Patel
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Benjamin E. Reese
- Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
- Department of Psychological & Brain SciencesUniversity of California, Santa BarbaraSanta BarbaraCaliforniaUSA
| |
Collapse
|
28
|
Chen K, Wang Y, Huang Y, Liu X, Tian X, Yang Y, Dong A. Cross-species scRNA-seq reveals the cellular landscape of retina and early alterations in type 2 diabetes mice. Genomics 2023; 115:110644. [PMID: 37279838 DOI: 10.1016/j.ygeno.2023.110644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) analysis have provided an unprecedented resolution for the studies on diabetic retinopathy (DR). However, the early changes in the retina in diabetes remain unclear. A total of 8 human and mouse scRNA-seq datasets, containing 276,402 cells were analyzed individually to comprehensively delineate the retinal cell atlas. The neural retinas were isolated from the type 2 diabetes (T2D) and control mice, and scRNA-seq analysis was conducted to evaluate the early effects of diabetes on the retina. Bipolar cell (BC) heterogeneity were identified. We found some stable BCs across multiple datasets, and explored their biological functions. A new RBC subtype (Car8_RBC) in the mouse retina was validated using the multi-color immunohistochemistry. AC149090.1 was significantly upregulated in the rod cells, ON cone BCs (CBCs), OFF CBCs, and RBCs in T2D mice. Additionally, the interneurons, especially BCs, were the most vulnerable cells to diabetes by integrating scRNA-seq and genome-wide association studies (GWAS) analyses. In conclusion, this study delineated a cross-species retinal cell atlas and uncovered the early pathological alterations in the retina of T2D mice.
Collapse
Affiliation(s)
- Kai Chen
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Yinhao Wang
- Department of Ophthalmology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310003, China
| | - Youyuan Huang
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Xinxin Liu
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China.
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China.
| | - Aimei Dong
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China; Department of General Practice, Peking University First Hospital, Beijing 100034, China.
| |
Collapse
|
29
|
Grabner CP, Futagi D, Shi J, Bindokas V, Kitano K, Schwartz EA, DeVries SH. Mechanisms of simultaneous linear and nonlinear computations at the mammalian cone photoreceptor synapse. Nat Commun 2023; 14:3486. [PMID: 37328451 PMCID: PMC10276006 DOI: 10.1038/s41467-023-38943-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/22/2023] [Indexed: 06/18/2023] Open
Abstract
Neurons enhance their computational power by combining linear and nonlinear transformations in extended dendritic trees. Rich, spatially distributed processing is rarely associated with individual synapses, but the cone photoreceptor synapse may be an exception. Graded voltages temporally modulate vesicle fusion at a cone's ~20 ribbon active zones. Transmitter then flows into a common, glia-free volume where bipolar cell dendrites are organized by type in successive tiers. Using super-resolution microscopy and tracking vesicle fusion and postsynaptic responses at the quantal level in the thirteen-lined ground squirrel, Ictidomys tridecemlineatus, we show that certain bipolar cell types respond to individual fusion events in the vesicle stream while other types respond to degrees of locally coincident events, creating a gradient across tiers that are increasingly nonlinear. Nonlinearities emerge from a combination of factors specific to each bipolar cell type including diffusion distance, contact number, receptor affinity, and proximity to glutamate transporters. Complex computations related to feature detection begin within the first visual synapse.
Collapse
Affiliation(s)
- Chad P Grabner
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075, Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Daiki Futagi
- College of Information Science and Engineering, Ritsumeikan University, Shiga, Japan
- Center for Systems Visual Science, Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
- Ritsumeikan Global Innovation Research Organisation, Ritsumeikan University, Shiga, Japan
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jun Shi
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Vytas Bindokas
- Dept of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Katsunori Kitano
- College of Information Science and Engineering, Ritsumeikan University, Shiga, Japan
- Center for Systems Visual Science, Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
| | - Eric A Schwartz
- Dept of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Steven H DeVries
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
30
|
Roy S, Wang D, Rudzite AM, Perry B, Scalabrino ML, Thapa M, Gong Y, Sher A, Field GD. Large-scale interrogation of retinal cell functions by 1-photon light-sheet microscopy. CELL REPORTS METHODS 2023; 3:100453. [PMID: 37159670 PMCID: PMC10163030 DOI: 10.1016/j.crmeth.2023.100453] [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] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 05/11/2023]
Abstract
Visual processing in the retina depends on the collective activity of large ensembles of neurons organized in different layers. Current techniques for measuring activity of layer-specific neural ensembles rely on expensive pulsed infrared lasers to drive 2-photon activation of calcium-dependent fluorescent reporters. We present a 1-photon light-sheet imaging system that can measure the activity in hundreds of neurons in the ex vivo retina over a large field of view while presenting visual stimuli. This allows for a reliable functional classification of different retinal cell types. We also demonstrate that the system has sufficient resolution to image calcium entry at individual synaptic release sites across the axon terminals of dozens of simultaneously imaged bipolar cells. The simple design, large field of view, and fast image acquisition make this a powerful system for high-throughput and high-resolution measurements of retinal processing at a fraction of the cost of alternative approaches.
Collapse
Affiliation(s)
- Suva Roy
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Depeng Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Andra M. Rudzite
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Benjamin Perry
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Miranda L. Scalabrino
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Mishek Thapa
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Yiyang Gong
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Alexander Sher
- Santa Cruz Institute for Particle Physics, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Greg D. Field
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| |
Collapse
|
31
|
Mao X, Stanbouly S, Holley J, Pecaut M, Crapo J. Evidence of Spaceflight-Induced Adverse Effects on Photoreceptors and Retinal Function in the Mouse Eye. Int J Mol Sci 2023; 24:ijms24087362. [PMID: 37108526 PMCID: PMC10138634 DOI: 10.3390/ijms24087362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The goal of the present study was to characterize acute oxidative damage in ocular structure and retinal function after exposure to spaceflight, and to evaluate the efficacy of an antioxidant in reducing spaceflight-induced changes in the retina. Ten-week-old adult C57BL/6 male mice were flown aboard the ISS on Space-X 24 over 35 days, and returned to Earth alive. The mice received a weekly injection of a superoxide dismutase mimic, MnTnBuOE-2-PyP 5+ (BuOE), before launch and during their stay onboard the ISS. Ground control mice were maintained on Earth under identical environmental conditions. Before the launch, intraocular pressure (IOP) was measured using a handheld tonometer and retinal function was evaluated using electroretinogram (ERG). ERG signals were recorded when the mouse eye was under dark-adapted conditions in response to ultraviolet monochromatic light flashes. Within 20 h after splashdown, IOP and ERG assessments were repeated before euthanasia. There were significant increases in body weight for habitat control groups post-flight compared to pre-flight measurements. However, the body weights were similar among flight groups before launch and after splashdown. The IOP measurements were similar between pre- and post-flight groups with no significant differences between BuOE-treated and saline controls. Immunofluorescence evaluation showed increases in retinal oxidative stress and apoptotic cell death after spaceflight. BuOE treatment significantly decreased the level of the oxidative stress biomarker. ERG data showed that the average amplitudes of the a- and b-wave were significantly decreased (39% and 32% by spaceflight, respectively) compared to that of habitat ground controls. These data indicate that spaceflight conditions induce oxidative stress in the retina, which may lead to photoreceptor cell damage and retinal function impairment.
Collapse
Affiliation(s)
- Xiaowen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Jacob Holley
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Michael Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - James Crapo
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, University of Colorado Denver, Denver, CO 80204, USA
| |
Collapse
|
32
|
Hanson L, Ravi-Chander P, Berson D, Awatramani GB. Hierarchical retinal computations rely on hybrid chemical-electrical signaling. Cell Rep 2023; 42:112030. [PMID: 36696265 DOI: 10.1016/j.celrep.2023.112030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/08/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Bipolar cells (BCs) are integral to the retinal circuits that extract diverse features from the visual environment. They bridge photoreceptors to ganglion cells, the source of retinal output. Understanding how such circuits encode visual features requires an accounting of the mechanisms that control glutamate release from bipolar cell axons. Here, we demonstrate orientation selectivity in a specific genetically identifiable type of mouse bipolar cell-type 5A (BC5A). Their synaptic terminals respond best when stimulated with vertical bars that are far larger than their dendritic fields. We provide evidence that this selectivity involves enhanced excitation for vertical stimuli that requires gap junctional coupling through connexin36. We also show that this orientation selectivity is detectable postsynaptically in direction-selective ganglion cells, which were not previously thought to be selective for orientation. Together, these results demonstrate how multiple features are extracted by a single hierarchical network, engaging distinct electrical and chemical synaptic pathways.
Collapse
Affiliation(s)
- Laura Hanson
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5, Canada
| | | | - David Berson
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Gautam B Awatramani
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5, Canada.
| |
Collapse
|
33
|
Park JS, Wei X. Size variations in synaptic terminals among different types of photoreceptors and across the zebrafish retina. Exp Eye Res 2023; 227:109377. [PMID: 36587757 PMCID: PMC9918681 DOI: 10.1016/j.exer.2022.109377] [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: 11/02/2022] [Revised: 12/06/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Photoreceptor synaptic terminals are responsible for transmitting visual information to downstream neurons. In vertebrate retinas, photoreceptor synaptic terminals are of different sizes and structures. The molecular mechanisms that underlie photoreceptor synaptic development are not clearly understood. Here, we have systematically examined the size variations in the synaptic terminals of cone and rod photoreceptors in the adult zebrafish retina. We reveal that the average cone pedicle sizes expand in the order of UV, blue, green, and red cones, echoing the increasing maximally sensitive wavelengths of the opsins expressed in the corresponding cone types. In addition, rod spherules are smaller than all cone pedicles. The terminals of each photoreceptor type also display distinct regional variations across the retina and between males and females. These findings establish the basis for using the zebrafish retina to study the molecular mechanisms that regulate the sizes and structures of photoreceptor terminals for proper visual functions.
Collapse
Affiliation(s)
- Jong-Su Park
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, 15213, Pennsylvania, USA
| | - Xiangyun Wei
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, 15213, Pennsylvania, USA; Department of Molecular Genetics and Microbiology, University of Pittsburgh School of Medicine, Pittsburgh, 15213, Pennsylvania, USA; Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, 15213, Pennsylvania, USA.
| |
Collapse
|
34
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
35
|
Haverkamp S, Mietsch M, Briggman KL. Developmental errors in the common marmoset retina. Front Neuroanat 2022; 16:1000693. [PMID: 36204677 PMCID: PMC9531312 DOI: 10.3389/fnana.2022.1000693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Although retinal organization is remarkably conserved, morphological anomalies can be found to different extents and varieties across animal species with each presenting unique characteristics and patterns of displaced and misplaced neurons. One of the most widely used non-human primates in research, the common marmoset (Callithrix jaccus) could potentially also be of interest for visual research, but is unfortunately not well characterized in this regard. Therefore, the aim of our study was to provide a first time description of structural retinal layering including morphological differences and distinctive features in this species. Retinas from animals (n = 26) of both sexes and different ages were immunostained with cell specific antibodies to label a variety of bipolar, amacrine and ganglion cells. Misplaced ganglion cells with somata in the outermost part of the inner nuclear layer and rod bipolar cells with axon terminals projecting into the outer plexiform layer instead of the inner plexiform layer independent of age or sex of the animals were the most obvious findings, whereas misplaced amacrine cells and misplaced cone bipolar axon terminals occurred to a lesser extent. With this first time description of developmental retinal errors over a wide age range, we provide a basic characterization of the retinal system of the common marmosets, which can be taken into account for future studies in this and other animal species. The finding of misplaced ganglion cells and misplaced bipolar cell axon terminals was not reported before and displays an anatomic variation worthwhile for future analyzes of their physiological and functional impact.
Collapse
Affiliation(s)
- Silke Haverkamp
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior – caesar, Bonn, Germany
- *Correspondence: Silke Haverkamp
| | - Matthias Mietsch
- Laboratory Animal Science Unit, German Primate Center, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Kevin L. Briggman
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior – caesar, Bonn, Germany
| |
Collapse
|
36
|
Bohl JM, Shehu A, Hellmer CB, Ichinose T. Patch clamp recording from bipolar cells in the wholemount mouse retina. STAR Protoc 2022; 3:101482. [PMID: 35769922 PMCID: PMC9234155 DOI: 10.1016/j.xpro.2022.101482] [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] [Indexed: 11/19/2022] Open
Abstract
Bipolar cells are the second-order neurons in the retina that are less accessible for investigating their synaptic responses. Here, we present a protocol to conduct patch clamp recordings from bipolar cells in the wholemount retina from Ai32 mutant mice. We detail whole-cell patch-clamp recording from bipolar cells to examine their light-evoked responses to optogenetic stimulation, followed by imaging terminals of recorded cells to determine bipolar cell type. We describe light stimulus information to activate channelrhodopsin-2 (ChR2). For complete details on the use and execution of this protocol, please refer to Hellmer et al. (2021). Detailed protocol for bipolar cell patch clamp recordings in wholemount mouse retina Bipolar cell subtype identification in live retinal tissue Detailed light stimulus information for channelrhodopsin (ChR2) activation
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Jeremy M Bohl
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Angela Shehu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chase B Hellmer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
37
|
Kulesh B, Reese BE, Keeley PW. Contraction of axonal and dendritic fields in Sox5-deficient cone bipolar cells is accompanied by axonal sprouting and dendritic hyper-innervation of pedicles. Front Neuroanat 2022; 16:944706. [PMID: 36093292 PMCID: PMC9459848 DOI: 10.3389/fnana.2022.944706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022] Open
Abstract
Multiple factors regulate the differentiation of neuronal morphology during development, including interactions with afferents, targets, and homotypic neighbors, as well as cell-intrinsic transcriptional regulation. Retinal bipolar cells provide an exemplary model system for studying the control of these processes, as there are 15 transcriptionally and morphologically distinct types, each extending their dendritic and axonal arbors in respective strata within the synaptic layers of the retina. Here we have examined the role of the transcription factor Sox5 in the control of the morphological differentiation of one type of cone bipolar cell (CBC), the Type 7 cell. We confirm selective expression of SOX5 in this single bipolar cell type, emerging at the close of the first post-natal week, prior to morphological differentiation. Conditional knockout mice were generated by crossing a bipolar cell-specific cre-expressing line with mice carrying floxed Sox5 alleles, as well as the Gustducin-gfp reporter which labels Type 7 CBCs. Loss of SOX5 was confirmed in the bipolar cell stratum, in GFP+ Type 7 cells. Such SOX5-deficient Type 7 cells differentiate axonal and dendritic arbors that are each reduced in areal extent. The axonal arbors exhibit sprouting in the inner plexiform layer (IPL), thereby extending their overall radial extent, while the dendritic arbors connect with fewer cone pedicles in the outer plexiform layer, showing an increase in the average number of dendritic contacts at each pedicle. SOX5-deficient Type 7 CBCs should therefore exhibit smaller receptive fields derived from fewer if now hyper-innervated pedicles, transmitting their signals across a broader depth through the IPL.
Collapse
Affiliation(s)
- Bridget Kulesh
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Benjamin E. Reese
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Patrick W. Keeley
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
- *Correspondence: Patrick W. Keeley
| |
Collapse
|
38
|
Meah A, Boodram V, Bucinca-Cupallari F, Lim H. Axonal architecture of the mouse inner retina revealed by second harmonic generation. PNAS NEXUS 2022; 1:pgac160. [PMID: 36106183 PMCID: PMC9463061 DOI: 10.1093/pnasnexus/pgac160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/11/2022] [Indexed: 01/29/2023]
Abstract
We describe a novel method for visualizing the network of axons in the unlabeled fresh wholemount retina. The intrinsic radiation of second harmonic generation (SHG) was utilized to visualize single axons of all major retinal neurons, i.e., photoreceptors, horizontal cells, bipolar cells, amacrine cells, and the retinal ganglion cells. The cell types of SHG+ axons were determined using transgenic GFP/YFP mice. New findings were obtained with retinal SHG imaging: Müller cells do not maintain uniformly polarized microtubules in the processes; SHG+ axons of bipolar cells terminate in the inner plexiform layer (IPL) in a subtype-specific manner; a subset of amacrine cells, presumably the axon-bearing types, emits SHG; and the axon-like neurites of amacrine cells provide a cytoskeletal scaffolding for the IPL stratification. To demonstrate the utility, retinal SHG imaging was applied to testing whether the inner retina is preserved in glaucoma, using DBA/2 mice as a model of glaucoma and DBA/2-Gpnmb+ as the nonglaucomatous control. It was found that the morphology of the inner retina was largely intact in glaucoma and the presynaptic compartments to the retinal ganglion cells were uncompromised. It proves retinal SHG imaging as a promising technology for studying the physiological and diseased retinas in 3D.
Collapse
Affiliation(s)
- Arafat Meah
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Vinessia Boodram
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Festa Bucinca-Cupallari
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA,The Graduate Centre of the City University of New York, New York, NY 10065, USA
| | | |
Collapse
|
39
|
Italiano ML, Guo T, Lovell NH, Tsai D. Improving the spatial resolution of artificial vision using midget retinal ganglion cell populations modelled at the human fovea. J Neural Eng 2022; 19. [PMID: 35609556 DOI: 10.1088/1741-2552/ac72c2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Retinal prostheses seek to create artificial vision by stimulating surviving retinal neurons of patients with profound vision impairment. Notwithstanding tremendous research efforts, the performance of all implants tested to date has remained rudimentary, incapable of overcoming the threshold for legal blindness. To maximize the perceptual efficacy of retinal prostheses, a device must be capable of controlling retinal neurons with greater spatiotemporal precision. Most studies of retinal stimulation were derived from either non-primate species or the peripheral primate retina. We investigated if artificial stimulation could leverage the high spatial resolution afforded by the neural substrates at the primate fovea and surrounding regions to achieve improved percept qualities. APPROACH We began by developing a new computational model capable of generating anatomically accurate retinal ganglion cell (RGC) populations within the human central retina. Next, multiple RGC populations across the central retina were stimulated in-silico to compare clinical and recently proposed neurostimulation configurations based on their ability to improve perceptual efficacy and reduce activation thresholds. MAIN RESULTS Our model uniquely upholds eccentricity-dependent characteristics such as RGC density and dendritic field diameter, whilst incorporating anatomically accurate features such as axon projection and three-dimensional RGC layering, features often forgone in favor of reduced computational complexity. Following epiretinal stimulation, the RGCs in our model produced response patterns in shapes akin to the complex percepts reported in clinical trials. Our results also demonstrated that even within the neuron-dense central retina, epiretinal stimulation using a multi-return hexapolar electrode arrangement could reliably achieve spatially focused RGC activation and could achieve single-cell excitation in 74% of all tested locations. SIGNIFICANCE This study establishes an anatomically accurate three-dimensional model of the human central retina and demonstrates the potential for an epiretinal hexapolar configuration to achieve consistent, spatially confined retinal responses, even within the neuron-dense foveal region. Our results promote the prospect and optimization of higher spatial resolution in future epiretinal implants.
Collapse
Affiliation(s)
- Michael Lewis Italiano
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Sydney, New South Wales, 2052, AUSTRALIA
| | - Tianruo Guo
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Sydney, New South Wales, 2052, AUSTRALIA
| | - Nigel H Lovell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Sydney, New South Wales, 2052, AUSTRALIA
| | - David Tsai
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Sydney, New South Wales, 2052, AUSTRALIA
| |
Collapse
|
40
|
Ganczer A, Szarka G, Balogh M, Hoffmann G, Tengölics ÁJ, Kenyon G, Kovács-Öller T, Völgyi B. Transience of the Retinal Output Is Determined by a Great Variety of Circuit Elements. Cells 2022; 11:cells11050810. [PMID: 35269432 PMCID: PMC8909309 DOI: 10.3390/cells11050810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Retinal ganglion cells (RGCs) encrypt stimulus features of the visual scene in action potentials and convey them toward higher visual centers in the brain. Although there are many visual features to encode, our recent understanding is that the ~46 different functional subtypes of RGCs in the retina share this task. In this scheme, each RGC subtype establishes a separate, parallel signaling route for a specific visual feature (e.g., contrast, the direction of motion, luminosity), through which information is conveyed. The efficiency of encoding depends on several factors, including signal strength, adaptational levels, and the actual efficacy of the underlying retinal microcircuits. Upon collecting inputs across their respective receptive field, RGCs perform further analysis (e.g., summation, subtraction, weighting) before they generate the final output spike train, which itself is characterized by multiple different features, such as the number of spikes, the inter-spike intervals, response delay, and the rundown time (transience) of the response. These specific kinetic features are essential for target postsynaptic neurons in the brain in order to effectively decode and interpret signals, thereby forming visual perception. We review recent knowledge regarding circuit elements of the mammalian retina that participate in shaping RGC response transience for optimal visual signaling.
Collapse
Affiliation(s)
- Alma Ganczer
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Márton Balogh
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Gyula Hoffmann
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ádám Jonatán Tengölics
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Garrett Kenyon
- Los Alamos National Laboratory, Computer & Computational Science Division, Los Alamos, NM 87545, USA;
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
41
|
Fournel R, Veruki ML, Hartveit E. Digital reconstruction and quantitative morphometric analysis of bipolar cells in live rat retinal slices. J Comp Neurol 2022; 530:1700-1728. [PMID: 35152437 PMCID: PMC9310816 DOI: 10.1002/cne.25308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/06/2022] [Accepted: 01/27/2022] [Indexed: 11/27/2022]
Abstract
Bipolar cells convey signals from photoreceptors in the outer retina to amacrine and ganglion cells in the inner retina. In mammals, there are typically 10–15 types of cone bipolar cells and one type of rod bipolar cell. Different types of cone bipolar cells are thought to code and transmit different features of a complex visual stimulus, thereby generating parallel channels that uniquely filter and transform the photoreceptor outputs. Differential synaptic connectivity and expression of ligand‐ and voltage‐gated ion channels are thought to be important mechanisms for processing and filtering visual signals. Whereas the biophysical basis for such mechanisms has been investigated more extensively in rat retina, there is a lack of quantitative morphological data necessary for advancing the structure–function correlation in this species, as recent connectomics investigations have focused on mouse retina. Here, we performed whole‐cell recordings from cone and rod bipolar cells in rat retinal slices, filled the cells with fluorescent dyes, and acquired image stacks by multiphoton excitation microscopy. Following deconvolution, we performed digital reconstruction and morphometric analysis of 25 cone and 14 rod bipolar cells. Compared to previous descriptions, the extent and complexity of branching of the axon terminal was surprisingly high. By precisely quantifying the level of stratification of the axon terminals in the inner plexiform layer, we have generated a reference system for reliable classification of individual cells in future studies focused on correlating physiological and morphological properties. The implemented workflow can be extended to the development of morphologically realistic compartmental models for these neurons.
Collapse
Affiliation(s)
- Rémi Fournel
- University of Bergen Department of Biomedicine Bergen Norway
| | | | - Espen Hartveit
- University of Bergen Department of Biomedicine Bergen Norway
| |
Collapse
|
42
|
Behrens C, Yadav SC, Korympidou MM, Zhang Y, Haverkamp S, Irsen S, Schaedler A, Lu X, Liu Z, Lause J, St-Pierre F, Franke K, Vlasits A, Dedek K, Smith RG, Euler T, Berens P, Schubert T. Retinal horizontal cells use different synaptic sites for global feedforward and local feedback signaling. Curr Biol 2022; 32:545-558.e5. [PMID: 34910950 PMCID: PMC8886496 DOI: 10.1016/j.cub.2021.11.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 10/19/2021] [Accepted: 11/23/2021] [Indexed: 11/19/2022]
Abstract
In the outer plexiform layer (OPL) of the mammalian retina, cone photoreceptors (cones) provide input to more than a dozen types of cone bipolar cells (CBCs). In the mouse, this transmission is modulated by a single horizontal cell (HC) type. HCs perform global signaling within their laterally coupled network but also provide local, cone-specific feedback. However, it is unknown how HCs provide local feedback to cones at the same time as global forward signaling to CBCs and where the underlying synapses are located. To assess how HCs simultaneously perform different modes of signaling, we reconstructed the dendritic trees of five HCs as well as cone axon terminals and CBC dendrites in a serial block-face electron microscopy volume and analyzed their connectivity. In addition to the fine HC dendritic tips invaginating cone axon terminals, we also identified "bulbs," short segments of increased dendritic diameter on the primary dendrites of HCs. These bulbs are in an OPL stratum well below the cone axon terminal base and make contacts with other HCs and CBCs. Our results from immunolabeling, electron microscopy, and glutamate imaging suggest that HC bulbs represent GABAergic synapses that do not receive any direct photoreceptor input. Together, our data suggest the existence of two synaptic strata in the mouse OPL, spatially separating cone-specific feedback and feedforward signaling to CBCs. A biophysical model of a HC dendritic branch and voltage imaging support the hypothesis that this spatial arrangement of synaptic contacts allows for simultaneous local feedback and global feedforward signaling by HCs.
Collapse
Affiliation(s)
- Christian Behrens
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Shubhash Chandra Yadav
- Neurosensorics/Animal Navigation, Institute for Biology and Environmental Sciences, University of Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26111 Oldenburg, Germany
| | - Maria M Korympidou
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Yue Zhang
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Silke Haverkamp
- Department of Computational Neuroethology, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Stephan Irsen
- Electron Microscopy and Analytics, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Anna Schaedler
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Xiaoyu Lu
- Systems, Synthetic, and Physical Biology Program, Rice University, 6500 Main St., Houston, TX 77005, USA
| | - Zhuohe Liu
- Department of Electrical and Computer Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Jan Lause
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - François St-Pierre
- Systems, Synthetic, and Physical Biology Program, Rice University, 6500 Main St., Houston, TX 77005, USA; Department of Electrical and Computer Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA; Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Katrin Franke
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Anna Vlasits
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Karin Dedek
- Neurosensorics/Animal Navigation, Institute for Biology and Environmental Sciences, University of Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26111 Oldenburg, Germany
| | - Robert G Smith
- Department of Neuroscience, University of Pennsylvania, 422 Curie Blvd, Philadelphia, PA 19104, USA
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Tübingen AI Center, University of Tübingen, Maria-von-Linden-Straße 6, 72076 Tübingen, Germany
| | - Timm Schubert
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany.
| |
Collapse
|
43
|
Dendro-somatic synaptic inputs to ganglion cells contradict receptive field and connectivity conventions in the mammalian retina. Curr Biol 2022; 32:315-328.e4. [PMID: 34822767 PMCID: PMC8792273 DOI: 10.1016/j.cub.2021.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 01/26/2023]
Abstract
The morphology of retinal neurons strongly influences their physiological function. Ganglion cell (GC) dendrites ramify in distinct strata of the inner plexiform layer (IPL) so that GCs responding to light increments (ON) or decrements (OFF) receive appropriate excitatory inputs. This vertical stratification prescribes response polarity and ensures consistent connectivity between cell types, whereas the lateral extent of GC dendritic arbors typically dictates receptive field (RF) size. Here, we identify circuitry in mouse retina that contradicts these conventions. AII amacrine cells are interneurons understood to mediate "crossover" inhibition by relaying excitatory input from the ON layer to inhibitory outputs in the OFF layer. Ultrastructural and physiological analyses show, however, that some AIIs deliver powerful inhibition to OFF GC somas and proximal dendrites in the ON layer, rendering the inhibitory RFs of these GCs smaller than their dendritic arbors. This OFF pathway, avoiding entirely the OFF region of the IPL, challenges several tenets of retinal circuitry. These results also indicate that subcellular synaptic organization can vary within a single population of neurons according to their proximity to potential postsynaptic targets.
Collapse
|
44
|
Sharpe ZJ, Shehu A, Ichinose T. Asymmetric Distributions of Achromatic Bipolar Cells in the Mouse Retina. Front Neuroanat 2022; 15:786142. [PMID: 35095431 PMCID: PMC8792968 DOI: 10.3389/fnana.2021.786142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
In the retina, evolutionary changes can be traced in the topography of photoreceptors. The shape of the visual streak depends on the height of the animal and its habitat, namely, woods, prairies, or mountains. Also, the distribution of distinct wavelength-sensitive cones is unique to each animal. For example, UV and green cones reside in the ventral and dorsal regions in the mouse retina, respectively, whereas in the rat retina these cones are homogeneously distributed. In contrast with the abundant investigation on the distribution of photoreceptors and the third-order neurons, the distribution of bipolar cells has not been well understood. We utilized two enhanced green fluorescent protein (EGFP) mouse lines, Lhx4-EGFP (Lhx4) and 6030405A18Rik-EGFP (Rik), to examine the topographic distributions of bipolar cells in the retina. First, we characterized their GFP-expressing cells using type-specific markers. We found that GFP was expressed by type 2, type 3a, and type 6 bipolar cells in the Rik mice and by type 3b, type 4, and type 5 bipolar cells in the Lhx4 mice. All these types are achromatic. Then, we examined the distributions of bipolar cells in the four cardinal directions and three different eccentricities of the retinal tissue. In the Rik mice, GFP-expressing bipolar cells were more highly observed in the nasal region than those in the temporal retina. The number of GFP cells was not different along with the ventral-dorsal axis. In contrast, in the Lhx4 mice, GFP-expressing cells occurred at a higher density in the ventral region than in the dorsal retina. However, no difference was observed along the nasal-temporal axis. Furthermore, we examined which type of bipolar cells contributed to the asymmetric distributions in the Rik mice. We found that type 3a bipolar cells occurred at a higher density in the temporal region, whereas type 6 bipolar cells were denser in the nasal region. The asymmetricity of these bipolar cells shaped the uneven distribution of the GFP cells in the Rik mice. In conclusion, we found that a subset of achromatic bipolar cells is asymmetrically distributed in the mouse retina, suggesting their unique roles in achromatic visual processing.
Collapse
|
45
|
West ER, Lapan SW, Lee C, Kajderowicz KM, Li X, Cepko CL. Spatiotemporal patterns of neuronal subtype genesis suggest hierarchical development of retinal diversity. Cell Rep 2022; 38:110191. [PMID: 34986354 DOI: 10.1016/j.celrep.2021.110191] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 10/21/2021] [Accepted: 12/08/2021] [Indexed: 11/26/2022] Open
Abstract
How do neuronal subtypes emerge during development? Recent molecular studies have profiled existing neuronal diversity, but neuronal subtype genesis remains elusive. The 15 types of mouse retinal bipolar interneurons are characterized by distinct functions, morphologies, and transcriptional profiles. Here, we develop a comprehensive spatiotemporal map of bipolar subtype genesis in the murine retina. Combining multiplexed detection of 16 RNA markers with timed delivery of 5-ethynyl uridine (EdU) and bromodeoxyuridine (BrdU), we analyze more than 30,000 single cells in full retinal sections to classify all bipolar subtypes and their birthdates. We find that bipolar subtype birthdates are ordered and follow a centrifugal developmental axis. Spatial analysis reveals a striking wave pattern of bipolar subtype birthdates, and lineage analyses suggest clonal restriction on homotypic subtype production. These results inspire a hierarchical developmental model, with ordered subtype genesis within lineages. Our results provide insight into neuronal subtype development and establish a framework for studying subtype diversification.
Collapse
Affiliation(s)
- Emma R West
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Sylvain W Lapan
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - ChangHee Lee
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kathrin M Kajderowicz
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Constance L Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
46
|
Ichinose T, Habib S. ON and OFF Signaling Pathways in the Retina and the Visual System. FRONTIERS IN OPHTHALMOLOGY 2022; 2:989002. [PMID: 36926308 PMCID: PMC10016624 DOI: 10.3389/fopht.2022.989002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Visual processing starts at the retina of the eye, and signals are then transferred primarily to the visual cortex and the tectum. In the retina, multiple neural networks encode different aspects of visual input, such as color and motion. Subsequently, multiple neural streams in parallel convey unique aspects of visual information to cortical and subcortical regions. Bipolar cells, which are the second order neurons of the retina, separate visual signals evoked by light and dark contrasts and encode them to ON and OFF pathways, respectively. The interplay between ON and OFF neural signals is the foundation for visual processing for object contrast which underlies higher order stimulus processing. ON and OFF pathways have been classically thought to signal in a mirror-symmetric manner. However, while these two pathways contribute synergistically to visual perception in some instances, they have pronounced asymmetries suggesting independent operation in other cases. In this review, we summarize the role of the ON-OFF dichotomy in visual signaling, aiming to contribute to the understanding of visual recognition.
Collapse
Affiliation(s)
- Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
- Correspondence: Tomomi Ichinose, MD, PhD,
| | - Samar Habib
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Medical Parasitology, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
47
|
Hellmer CB, Hall LM, Bohl JM, Sharpe ZJ, Smith RG, Ichinose T. Cholinergic feedback to bipolar cells contributes to motion detection in the mouse retina. Cell Rep 2021; 37:110106. [PMID: 34910920 PMCID: PMC8793255 DOI: 10.1016/j.celrep.2021.110106] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 08/11/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Retinal bipolar cells are second-order neurons that transmit basic features of the visual scene to postsynaptic partners. However, their contribution to motion detection has not been fully appreciated. Here, we demonstrate that cholinergic feedback from starburst amacrine cells (SACs) to certain presynaptic bipolar cells via alpha-7 nicotinic acetylcholine receptors (α7-nAChRs) promotes direction-selective signaling. Patch clamp recordings reveal that distinct bipolar cell types making synapses at proximal SAC dendrites also express α7-nAChRs, producing directionally skewed excitatory inputs. Asymmetric SAC excitation contributes to motion detection in On-Off direction-selective ganglion cells (On-Off DSGCs), predicted by computational modeling of SAC dendrites and supported by patch clamp recordings from On-Off DSGCs when bipolar cell α7-nAChRs is eliminated pharmacologically or by conditional knockout. Altogether, these results show that cholinergic feedback to bipolar cells enhances direction-selective signaling in postsynaptic SACs and DSGCs, illustrating how bipolar cells provide a scaffold for postsynaptic microcircuits to cooperatively enhance retinal motion detection.
Collapse
Affiliation(s)
- Chase B Hellmer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA; Present address: Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Leo M Hall
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA; Present address: Department of Internal Medicine, St. Mary Mercy Livonia Hospital, Livonia, MI 48154, USA
| | - Jeremy M Bohl
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zachary J Sharpe
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Robert G Smith
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
48
|
Ptito M, Bleau M, Bouskila J. The Retina: A Window into the Brain. Cells 2021; 10:cells10123269. [PMID: 34943777 PMCID: PMC8699497 DOI: 10.3390/cells10123269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Affiliation(s)
- Maurice Ptito
- School of Optometry, University of Montreal, Montreal, QC H3T 1P1, Canada; (M.B.); (J.B.)
- Department of Neuroscience, Copenhagen University, 2200 Copenhagen, Denmark
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Correspondence:
| | - Maxime Bleau
- School of Optometry, University of Montreal, Montreal, QC H3T 1P1, Canada; (M.B.); (J.B.)
| | - Joseph Bouskila
- School of Optometry, University of Montreal, Montreal, QC H3T 1P1, Canada; (M.B.); (J.B.)
| |
Collapse
|
49
|
Pang JJ, Gao F, Wu SM. Dual-Cell Patch-Clamp Recording Revealed a Mechanism for a Ribbon Synapse to Process Both Digital and Analog Inputs and Outputs. Front Cell Neurosci 2021; 15:722533. [PMID: 34720878 PMCID: PMC8552968 DOI: 10.3389/fncel.2021.722533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/13/2021] [Indexed: 12/02/2022] Open
Abstract
A chemical synapse is either an action potential (AP) synapse or a graded potential (GP) synapse but not both. This study investigated how signals passed the glutamatergic synapse between the rod photoreceptor and its postsynaptic hyperpolarizing bipolar cells (HBCs) and light responses of retinal neurons with dual-cell and single-cell patch-clamp recording techniques. The results showed that scotopic lights evoked GPs in rods, whose depolarizing Phase 3 associated with the light offset also evoked APs of a duration of 241.8 ms and a slope of 4.5 mV/ms. The depolarization speed of Phase 3 (Speed) was 0.0001–0.0111 mV/ms and 0.103–0.469 mV/ms for rods and cones, respectively. On pairs of recorded rods and HBCs, only the depolarizing limbs of square waves applied to rods evoked clear currents in HBCs which reversed at −6.1 mV, indicating cation currents. We further used stimuli that simulated the rod light response to stimulate rods and recorded the rod-evoked excitatory current (rdEPSC) in HBCs. The normalized amplitude (R/Rmax), delay, and rising slope of rdEPSCs were differentially exponentially correlated with the Speed (all p < 0.001). For the Speed < 0.1 mV/ms, R/Rmax grew while the delay and duration reduced slowly; for the Speed between 0.1 and 0.4 mV/ms, R/Rmax grew fast while the delay and duration dramatically decreased; for the Speed > 0.4 mV/ms, R/Rmax reached the plateau, while the delay and duration approached the minimum, resembling digital signals. The rdEPSC peak was left-shifted and much faster than currents in rods. The scotopic-light-offset-associated major and minor cation currents in retinal ganglion cells (RGCs), the gigantic excitatory transient currents (GTECs) in HBCs, and APs and Phase 3 in rods showed comparable light-intensity-related locations. The data demonstrate that the rod-HBC synapse is a perfect synapse that can differentially decode and code analog and digital signals to process enormously varied rod and coupled-cone inputs.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Fan Gao
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Samuel M Wu
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
50
|
Young BK, Ramakrishnan C, Ganjawala T, Wang P, Deisseroth K, Tian N. An uncommon neuronal class conveys visual signals from rods and cones to retinal ganglion cells. Proc Natl Acad Sci U S A 2021; 118:e2104884118. [PMID: 34702737 PMCID: PMC8612366 DOI: 10.1073/pnas.2104884118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/01/2023] Open
Abstract
Neurons in the central nervous system (CNS) are distinguished by the neurotransmitter types they release, their synaptic connections, morphology, and genetic profiles. To fully understand how the CNS works, it is critical to identify all neuronal classes and reveal their synaptic connections. The retina has been extensively used to study neuronal development and circuit formation. Here, we describe a previously unidentified interneuron in mammalian retina. This interneuron shares some morphological, physiological, and molecular features with retinal bipolar cells, such as receiving input from photoreceptors and relaying visual signals to retinal ganglion cells. It also shares some features with amacrine cells (ACs), particularly Aii-ACs, such as their neurite morphology in the inner plexiform layer, the expression of some AC-specific markers, and possibly the release of the inhibitory neurotransmitter glycine. Thus, we unveil an uncommon interneuron, which may play an atypical role in vision.
Collapse
Affiliation(s)
- Brent K Young
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
| | | | - Tushar Ganjawala
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202
| | - Ping Wang
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Ning Tian
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132;
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, UT 84148
| |
Collapse
|