1
|
Müllner FE, Roska B. Individual thalamic inhibitory interneurons are functionally specialized toward distinct visual features. Neuron 2024; 112:2765-2782.e9. [PMID: 38917805 PMCID: PMC11348917 DOI: 10.1016/j.neuron.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Inhibitory interneurons in the dorsolateral geniculate nucleus (dLGN) are situated at the first central synapse of the image-forming visual pathway, but little is known about their function. Given their anatomy, they are expected to be multiplexors, integrating many different retinal channels along their dendrites. Here, using targeted single-cell-initiated rabies tracing, we found that mouse dLGN interneurons exhibit a degree of retinal input specialization similar to thalamocortical neurons. Some are anatomically highly specialized, for example, toward motion-selective information. Two-photon calcium imaging performed in vivo revealed that interneurons are also functionally specialized. In mice lacking retinal horizontal direction selectivity, horizontal direction selectivity is reduced in interneurons, suggesting a causal link between input and functional specialization. Functional specialization is not only present at interneuron somata but also extends into their dendrites. Altogether, inhibitory interneurons globally display distinct visual features which reflect their retinal input specialization and are ideally suited to perform feature-selective inhibition.
Collapse
Affiliation(s)
- Fiona E Müllner
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
2
|
Gomez-Frittelli J, Devienne G, Travis L, Kyloh MA, Duan X, Hibberd TJ, Spencer NJ, Huguenard JR, Kaltschmidt JA. Synaptic cell adhesion molecule Cdh6 identifies a class of sensory neurons with novel functions in colonic motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606748. [PMID: 39149241 PMCID: PMC11326146 DOI: 10.1101/2024.08.06.606748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Intrinsic sensory neurons are an essential part of the enteric nervous system (ENS) and play a crucial role in gastrointestinal tract motility and digestion. Neuronal subtypes in the ENS have been distinguished by their electrophysiological properties, morphology, and expression of characteristic markers, notably neurotransmitters and neuropeptides. Here we investigated synaptic cell adhesion molecules as novel cell type markers in the ENS. Our work identifies two Type II classic cadherins, Cdh6 and Cdh8, specific to sensory neurons in the mouse colon. We show that Cdh6+ neurons demonstrate all other distinguishing classifications of enteric sensory neurons including marker expression of Calcb and Nmu, Dogiel type II morphology and AH-type electrophysiology and I H current. Optogenetic activation of Cdh6+ sensory neurons in distal colon evokes retrograde colonic motor complexes (CMCs), while pharmacologic blockade of rhythmicity-associated current I H disrupts the spontaneous generation of CMCs. These findings provide the first demonstration of selective activation of a single neurochemical and functional class of enteric neurons, and demonstrate a functional and critical role for sensory neurons in the generation of CMCs.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford University; Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
| | - Gabrielle Devienne
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Melinda A Kyloh
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San Francisco; San Francisco, CA, USA
| | - Tim J Hibberd
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - John R Huguenard
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Julia A Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
3
|
Merkulyeva N, Mikhalkin A, Veshchitskii A. Inner Structure of the Lateral Geniculate Complex of Adult and Newborn Acomys cahirinus. Int J Mol Sci 2024; 25:7855. [PMID: 39063096 PMCID: PMC11277159 DOI: 10.3390/ijms25147855] [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: 07/05/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Acomys cahirinus is a unique Rodentia species with several distinctive physiological traits, such as precocial development and remarkable regenerative abilities. These characteristics render A. cahirinus increasingly valuable for regenerative and developmental physiology studies. Despite this, the structure and postnatal development of the central nervous system in A. cahirinus have been inadequately explored, with only sporadic data available. This study is the first in a series of papers addressing these gaps. Our first objective was to characterize the structure of the main visual thalamic region, the lateral geniculate complex, using several neuronal markers (including Ca2+-binding proteins, glutamic acid decarboxylase enzyme, and non-phosphorylated domains of heavy-chain neurofilaments) to label populations of principal neurons and interneurons in adult and newborn A. cahirinus. As typically found in other rodents, we identified three subdivisions in the geniculate complex: the dorsal and ventral lateral geniculate nuclei (LGNd and LGNv) and the intergeniculate leaflet (IGL). Additionally, we characterized internal diversity in the LGN nuclei. The "shell" and "core" regions of the LGNd were identified using calretinin in adults and newborns. In adults, the inner and outer parts of the LGNv were identified using calbindin, calretinin, parvalbumin, GAD67, and SMI-32, whereas in newborns, calretinin and SMI-32 were employed for this purpose. Our findings revealed more pronounced developmental changes in LGNd compared to LGNv and IGL, suggesting that LGNd is less mature at birth and more influenced by visual experience.
Collapse
Affiliation(s)
- Natalia Merkulyeva
- Neuromorphology Laboratory, Pavlov Institute of Physiology of Russian Academy of Sciences, St. Petersburg 199034, Russia; (A.M.); (A.V.)
| | | | | |
Collapse
|
4
|
Li J, Choi J, Cheng X, Ma J, Pema S, Sanes JR, Mardon G, Frankfort BJ, Tran NM, Li Y, Chen R. Comprehensive single-cell atlas of the mouse retina. iScience 2024; 27:109916. [PMID: 38812536 PMCID: PMC11134544 DOI: 10.1016/j.isci.2024.109916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/18/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has advanced our understanding of cellular heterogeneity by characterizing cell types across tissues and species. While several mouse retinal scRNA-seq datasets exist, each dataset is either limited in cell numbers or focused on specific cell classes, thereby hindering comprehensive gene expression analysis across all retina types. To fill the gap, we generated the largest retinal scRNA-seq dataset to date, comprising approximately 190,000 single cells from C57BL/6J mouse retinas, enriched for rare population cells via antibody-based magnetic cell sorting. Integrating this dataset with public datasets, we constructed the Mouse Retina Cell Atlas (MRCA) for wild-type mice, encompassing over 330,000 cells, characterizing 12 major classes and 138 cell types. The MRCA consolidates existing knowledge, identifies new cell types, and is publicly accessible via CELLxGENE, UCSC Cell Browser, and the Broad Single Cell Portal, providing a user-friendly resource for the mouse retina research community.
Collapse
Affiliation(s)
- Jin Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jongsu Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xuesen Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justin Ma
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shahil Pema
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Graeme Mardon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Ophthalmology and Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin J. Frankfort
- Departments of Ophthalmology and Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nicholas M. Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Toma K, Zhao M, Zhang S, Wang F, Graham HK, Zou J, Modgil S, Shang WH, Tsai NY, Cai Z, Liu L, Hong G, Kriegstein AR, Hu Y, Körbelin J, Zhang R, Liao YJ, Kim TN, Ye X, Duan X. Perivascular neurons instruct 3D vascular lattice formation via neurovascular contact. Cell 2024; 187:2767-2784.e23. [PMID: 38733989 PMCID: PMC11223890 DOI: 10.1016/j.cell.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024]
Abstract
The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.
Collapse
Affiliation(s)
- Kenichi Toma
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Mengya Zhao
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Shaobo Zhang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Fei Wang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Hannah K Graham
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Jun Zou
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Shweta Modgil
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wenhao H Shang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Y Tsai
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Zhishun Cai
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Liping Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Guiying Hong
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Arnold R Kriegstein
- Department of Neurology and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ruobing Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tyson N Kim
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Xin Ye
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology and Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
DePiero VJ, Deng Z, Chen C, Savier EL, Chen H, Wei W, Cang J. Transformation of Motion Pattern Selectivity from Retina to Superior Colliculus. J Neurosci 2024; 44:e1704232024. [PMID: 38569924 PMCID: PMC11097260 DOI: 10.1523/jneurosci.1704-23.2024] [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: 09/11/2023] [Revised: 03/07/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
The superior colliculus (SC) is a prominent and conserved visual center in all vertebrates. In mice, the most superficial lamina of the SC is enriched with neurons that are selective for the moving direction of visual stimuli. Here, we study how these direction selective neurons respond to complex motion patterns known as plaids, using two-photon calcium imaging in awake male and female mice. The plaid pattern consists of two superimposed sinusoidal gratings moving in different directions, giving an apparent pattern direction that lies between the directions of the two component gratings. Most direction selective neurons in the mouse SC respond robustly to the plaids and show a high selectivity for the moving direction of the plaid pattern but not of its components. Pattern motion selectivity is seen in both excitatory and inhibitory SC neurons and is especially prevalent in response to plaids with large cross angles between the two component gratings. However, retinal inputs to the SC are ambiguous in their selectivity to pattern versus component motion. Modeling suggests that pattern motion selectivity in the SC can arise from a nonlinear transformation of converging retinal inputs. In contrast, the prevalence of pattern motion selective neurons is not seen in the primary visual cortex (V1). These results demonstrate an interesting difference between the SC and V1 in motion processing and reveal the SC as an important site for encoding pattern motion.
Collapse
Affiliation(s)
- Victor J DePiero
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
| | - Zixuan Deng
- Committee on Neurobiology, University of Chicago, Chicago, Illinois 60637
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
| | - Elise L Savier
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Department of Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Hui Chen
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
| | - Wei Wei
- Department of Neurobiology, Neuroscience Institute, University of Chicago, Chicago, Illinois 60637
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
7
|
Lin F, Li Y, Wang J, Jardines S, King R, Chrenek MA, Wiggs JL, Boatright JH, Geisert EE. POU6F2, a risk factor for glaucoma, myopia and dyslexia, labels specific populations of retinal ganglion cells. Sci Rep 2024; 14:10096. [PMID: 38698014 PMCID: PMC11066091 DOI: 10.1038/s41598-024-60444-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/23/2024] [Indexed: 05/05/2024] Open
Abstract
Pou6f2 is a genetic connection between central corneal thickness (CCT) in the mouse and a risk factor for developing primary open-angle glaucoma. POU6F2 is also a risk factor for several conditions in humans, including glaucoma, myopia, and dyslexia. Recent findings demonstrate that POU6F2-positive retinal ganglion cells (RGCs) comprise a number of RGC subtypes in the mouse, some of which also co-stain for Cdh6 and Hoxd10. These POU6F2-positive RGCs appear to be novel of ON-OFF directionally selective ganglion cells (ooDSGCs) that do not co-stain with CART or SATB2 (typical ooDSGCs markers). These POU6F2-positive cells are sensitive to damage caused by elevated intraocular pressure. In the DBA/2J mouse glaucoma model, heavily-labeled POU6F2 RGCs decrease by 73% at 8 months of age compared to only 22% loss of total RGCs (labeled with RBPMS). Additionally, Pou6f2-/- mice suffer a significant loss of acuity and spatial contrast sensitivity along with an 11.4% loss of total RGCs. In the rhesus macaque retina, POU6F2 labels the large parasol ganglion cells that form the magnocellular (M) pathway. The association of POU6F2 with the M-pathway may reveal in part its role in human glaucoma, myopia, and dyslexia.
Collapse
Affiliation(s)
- Fangyu Lin
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Ying Li
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Jiaxing Wang
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Sandra Jardines
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Rebecca King
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Micah A Chrenek
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Janey L Wiggs
- Massachusetts Eye and Ear, Harvard Medical School Boston, Boston, MA, USA
| | - Jeffrey H Boatright
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | - Eldon E Geisert
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Tworig JM, Morrie RD, Bistrong K, Somaiya RD, Hsu S, Liang J, Cornejo KG, Feller MB. Differential Expression Analysis Identifies Candidate Synaptogenic Molecules for Wiring Direction-Selective Circuits in the Retina. J Neurosci 2024; 44:e1461232024. [PMID: 38514178 PMCID: PMC11063823 DOI: 10.1523/jneurosci.1461-23.2024] [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: 08/01/2023] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
An organizational feature of neural circuits is the specificity of synaptic connections. A striking example is the direction-selective (DS) circuit of the retina. There are multiple subtypes of DS retinal ganglion cells (DSGCs) that prefer motion along one of four preferred directions. This computation is mediated by selective wiring of a single inhibitory interneuron, the starburst amacrine cell (SAC), with each DSGC subtype preferentially receiving input from a subset of SAC processes. We hypothesize that the molecular basis of this wiring is mediated in part by unique expression profiles of DSGC subtypes. To test this, we first performed paired recordings from isolated mouse retinas of both sexes to determine that postnatal day 10 (P10) represents the age at which asymmetric synapses form. Second, we performed RNA sequencing and differential expression analysis on isolated P10 ON-OFF DSGCs tuned for either nasal or ventral motion and identified candidates which may promote direction-specific wiring. We then used a conditional knock-out strategy to test the role of one candidate, the secreted synaptic organizer cerebellin-4 (Cbln4), in the development of DS tuning. Using two-photon calcium imaging, we observed a small deficit in directional tuning among ventral-preferring DSGCs lacking Cbln4, though whole-cell voltage-clamp recordings did not identify a significant change in inhibitory inputs. This suggests that Cbln4 does not function primarily via a cell-autonomous mechanism to instruct wiring of DS circuits. Nevertheless, our transcriptomic analysis identified unique candidate factors for gaining insights into the molecular mechanisms that instruct wiring specificity in the DS circuit.
Collapse
Affiliation(s)
- Joshua M Tworig
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Ryan D Morrie
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Karina Bistrong
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720
| | - Rachana D Somaiya
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Shaw Hsu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Jocelyn Liang
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Karen G Cornejo
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720
| |
Collapse
|
9
|
Ma J, Chu TK, Polo Prieto M, Park Y, Li Y, Chen R, Mardon G, Frankfort BJ, Tran NM. Sample multiplexing for retinal single-cell RNA-sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.589797. [PMID: 38712294 PMCID: PMC11071429 DOI: 10.1101/2024.04.23.589797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Rare cell populations can be challenging to characterize using microfluidic single-cell RNA sequencing (scRNA-seq) platforms. Typically, the population of interest must be enriched and pooled from multiple biological specimens for efficient collection. However, these practices preclude the resolution of sample origin together with phenotypic data and are problematic in experiments in which biological or technical variation is expected to be high (e.g., disease models, genetic perturbation screens, or human samples). One solution is sample multiplexing whereby each sample is tagged with a unique sequence barcode that is resolved bioinformatically. We have established a scRNA-seq sample multiplexing pipeline for mouse retinal ganglion cells using cholesterol-modified-oligos and utilized the enhanced precision to investigate cell type distribution and transcriptomic variance across retinal samples. As single cell transcriptomics are becoming more widely used to research development and disease, sample multiplexing represents a useful method to enhance the precision of scRNA-seq analysis.
Collapse
|
10
|
de Malmazet D, Kühn NK, Li C, Farrow K. Retinal origin of orientation but not direction selective maps in the superior colliculus. Curr Biol 2024; 34:1222-1233.e7. [PMID: 38417446 PMCID: PMC10980837 DOI: 10.1016/j.cub.2024.02.001] [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: 08/18/2023] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 03/01/2024]
Abstract
Neurons in the mouse superior colliculus ("colliculus") are arranged in ordered spatial maps. While orientation-selective (OS) neurons form a concentric map aligned to the center of vision, direction-selective (DS) neurons are arranged in patches with changing preferences across the visual field. It remains unclear whether these maps are a consequence of feedforward input from the retina or local computations in the colliculus. To determine whether these maps originate in the retina, we mapped the local and global distribution of OS and DS retinal ganglion cell axon boutons using in vivo two-photon calcium imaging. We found that OS boutons formed patches that matched the distribution of OS neurons within the colliculus. DS boutons displayed fewer regional specializations, better reflecting the organization of DS neurons in the retina. Both eyes convey similar orientation but different DS inputs to the colliculus, as shown in recordings from retinal explants. These data demonstrate that orientation and direction maps within the colliculus are independent, where orientation maps are likely inherited from the retina, but direction maps require additional computations.
Collapse
Affiliation(s)
- Daniel de Malmazet
- Neuro-Electronics Research Flanders, Leuven 3001, Belgium; KU Leuven, Department of Biology & Leuven Brain Institute, Leuven 3000, Belgium; MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Norma K Kühn
- Neuro-Electronics Research Flanders, Leuven 3001, Belgium; KU Leuven, Department of Biology & Leuven Brain Institute, Leuven 3000, Belgium; VIB, Leuven 3001, Belgium
| | - Chen Li
- Neuro-Electronics Research Flanders, Leuven 3001, Belgium; KU Leuven, Department of Biology & Leuven Brain Institute, Leuven 3000, Belgium
| | - Karl Farrow
- Neuro-Electronics Research Flanders, Leuven 3001, Belgium; KU Leuven, Department of Biology & Leuven Brain Institute, Leuven 3000, Belgium; VIB, Leuven 3001, Belgium; imec, Leuven 3001, Belgium.
| |
Collapse
|
11
|
Krizan J, Song X, Fitzpatrick MJ, Shen N, Soto F, Kerschensteiner D. Predation without direction selectivity. Proc Natl Acad Sci U S A 2024; 121:e2317218121. [PMID: 38483997 PMCID: PMC10962952 DOI: 10.1073/pnas.2317218121] [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: 10/04/2023] [Accepted: 01/27/2024] [Indexed: 03/19/2024] Open
Abstract
Across the animal kingdom, visual predation relies on motion-sensing neurons in the superior colliculus (SC) and its orthologs. These neurons exhibit complex stimulus preferences, including direction selectivity, which is thought to be critical for tracking the unpredictable escape routes of prey. The source of direction selectivity in the SC is contested, and its contributions to predation have not been tested experimentally. Here, we use type-specific cell removal to show that narrow-field (NF) neurons in the mouse SC guide predation. In vivo recordings demonstrate that direction-selective responses of NF cells are independent of recently reported stimulus-edge effects. Monosynaptic retrograde tracing reveals that NF cells receive synaptic input from direction-selective ganglion cells. When we eliminate direction selectivity in the retina of adult mice, direction-selective responses in the SC, including in NF cells, are lost. However, eliminating retinal direction selectivity does not affect the hunting success or strategies of mice, even when direction selectivity is removed after mice have learned to hunt, and despite abolishing the gaze-stabilizing optokinetic reflex. Thus, our results identify the retinal source of direction selectivity in the SC. They show that NF cells in the SC guide predation, an essential spatial orienting task, independent of their direction selectivity, revealing behavioral multiplexing of complex neural feature preferences and highlighting the importance of feature-selective manipulations for neuroethology.
Collapse
Affiliation(s)
- Jenna Krizan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Graduate program in Neuroscience, Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Xiayingfang Song
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Graduate program in Biomedical Engineering, Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Michael J. Fitzpatrick
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Graduate program in Neuroscience, Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Ning Shen
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Florentina Soto
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO63110
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| |
Collapse
|
12
|
Roy S, Yao X, Rathinavelu J, Field GD. GABAergic Inhibition Controls Receptive Field Size, Sensitivity, and Contrast Preference of Direction Selective Retinal Ganglion Cells Near the Threshold of Vision. J Neurosci 2024; 44:e1979232023. [PMID: 38182419 PMCID: PMC10941243 DOI: 10.1523/jneurosci.1979-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Information about motion is encoded by direction-selective retinal ganglion cells (DSGCs). These cells reliably transmit this information across a broad range of light levels, spanning moonlight to sunlight. Previous work indicates that adaptation to low light levels causes heterogeneous changes to the direction tuning of ON-OFF (oo)DSGCs and suggests that superior-preferring ON-OFF DSGCs (s-DSGCs) are biased toward detecting stimuli rather than precisely signaling direction. Using a large-scale multielectrode array, we measured the absolute sensitivity of ooDSGCs and found that s-DSGCs are 10-fold more sensitive to dim flashes of light than other ooDSGCs. We measured their receptive field (RF) sizes and found that s-DSGCs also have larger receptive fields than other ooDSGCs; however, the size difference does not fully explain the sensitivity difference. Using a conditional knock-out of gap junctions and pharmacological manipulations, we demonstrate that GABA-mediated inhibition contributes to the difference in absolute sensitivity and receptive field size at low light levels, while the connexin36-mediated gap junction coupling plays a minor role. We further show that under scotopic conditions, ooDSGCs exhibit only an ON response, but pharmacologically removing GABA-mediated inhibition unmasks an OFF response. These results reveal that GABAergic inhibition controls and differentially modulates the responses of ooDSGCs under scotopic conditions.
Collapse
Affiliation(s)
- Suva Roy
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| | - Xiaoyang Yao
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Jay Rathinavelu
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Greg D Field
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, California 90095
| |
Collapse
|
13
|
Chander PR, Hanson L, Chundekkad P, Awatramani GB. Neural Circuits Underlying Multifeature Extraction in the Retina. J Neurosci 2024; 44:e0910232023. [PMID: 37957014 PMCID: PMC10919202 DOI: 10.1523/jneurosci.0910-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Classic ON-OFF direction-selective ganglion cells (DSGCs) that encode the four cardinal directions were recently shown to also be orientation-selective. To clarify the mechanisms underlying orientation selectivity, we employed a variety of electrophysiological, optogenetic, and gene knock-out strategies to test the relative contributions of glutamate, GABA, and acetylcholine (ACh) input that are known to drive DSGCs, in male and female mouse retinas. Extracellular spike recordings revealed that DSGCs respond preferentially to either vertical or horizontal bars, those that are perpendicular to their preferred-null motion axes. By contrast, the glutamate input to all four DSGC types measured using whole-cell patch-clamp techniques was found to be tuned along the vertical axis. Tuned glutamatergic excitation was heavily reliant on type 5A bipolar cells, which appear to be electrically coupled via connexin 36 containing gap junctions to the vertically oriented processes of wide-field amacrine cells. Vertically tuned inputs are transformed by the GABAergic/cholinergic "starburst" amacrine cells (SACs), which are critical components of the direction-selective circuit, into distinct patterns of inhibition and excitation. Feed-forward SAC inhibition appears to "veto" preferred orientation glutamate excitation in dorsal/ventral (but not nasal/temporal) coding DSGCs "flipping" their orientation tuning by 90° and accounts for the apparent mismatch between glutamate input tuning and the DSGC's spiking response. Together, these results reveal how two distinct synaptic motifs interact to generate complex feature selectivity, shedding light on the intricate circuitry that underlies visual processing in the retina.
Collapse
Affiliation(s)
| | - Laura Hanson
- Department of Biology, University of Victoria, Victoria, British Columbia V8W 4A4, Canada
| | - Pavitra Chundekkad
- Department of Biology, University of Victoria, Victoria, British Columbia V8W 4A4, Canada
| | | |
Collapse
|
14
|
Yuan M, Jin S, Tan G, Song S, Liu Y, Wang H, Shen Y. A Non-canonical Excitatory PV RGC-PV SC Visual Pathway for Mediating the Looming-evoked Innate Defensive Response. Neurosci Bull 2024; 40:310-324. [PMID: 37302108 PMCID: PMC10912393 DOI: 10.1007/s12264-023-01076-z] [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: 02/23/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Parvalbumin-positive retinal ganglion cells (PV+ RGCs) are an essential subset of RGCs found in various species. However, their role in transmitting visual information remains unclear. Here, we characterized PV+ RGCs in the retina and explored the functions of the PV+ RGC-mediated visual pathway. By applying multiple viral tracing strategies, we investigated the downstream of PV+ RGCs across the whole brain. Interestingly, we found that the PV+ RGCs provided direct monosynaptic input to PV+ excitatory neurons in the superficial layers of the superior colliculus (SC). Ablation or suppression of SC-projecting PV+ RGCs abolished or severely impaired the flight response to looming visual stimuli in mice without affecting visual acuity. Furthermore, using transcriptome expression profiling of individual cells and immunofluorescence colocalization for RGCs, we found that PV+ RGCs are predominant glutamatergic neurons. Thus, our findings indicate the critical role of PV+ RGCs in an innate defensive response and suggest a non-canonical subcortical visual pathway from excitatory PV+ RGCs to PV+ SC neurons that regulates looming visual stimuli. These results provide a potential target for intervening and treating diseases related to this circuit, such as schizophrenia and autism.
Collapse
Affiliation(s)
- Man Yuan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Sen Jin
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Siyuan Song
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, 77030, USA
| | - Yizong Liu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Huadong Wang
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China.
- Frontier Science Center of Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
15
|
Reinhard J, Mueller-Buehl C, Wiemann S, Roll L, Luft V, Shabani H, Rathbun DL, Gan L, Kuo CC, Franzen J, Joachim SC, Faissner A. Neural extracellular matrix regulates visual sensory motor integration. iScience 2024; 27:108846. [PMID: 38318351 PMCID: PMC10839651 DOI: 10.1016/j.isci.2024.108846] [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: 08/08/2023] [Revised: 10/12/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Visual processing depends on sensitive and balanced synaptic neurotransmission. Extracellular matrix proteins in the environment of cells are key modulators in synaptogenesis and synaptic plasticity. In the present study, we provide evidence that the combined loss of the four extracellular matrix components, brevican, neurocan, tenascin-C, and tenascin-R, in quadruple knockout mice leads to severe retinal dysfunction and diminished visual motion processing in vivo. Remarkably, impaired visual motion processing was accompanied by a developmental loss of cholinergic direction-selective starburst amacrine cells. Additionally, we noted imbalance of inhibitory and excitatory synaptic signaling in the quadruple knockout retina. Collectively, the study offers insights into the functional importance of four key extracellular matrix proteins for retinal function, visual motion processing, and synaptic signaling.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Veronika Luft
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Hamed Shabani
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard-Karls-University Tuebingen, 72076 Tuebingen, Germany
| | - Daniel L. Rathbun
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard-Karls-University Tuebingen, 72076 Tuebingen, Germany
| | - Lin Gan
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Chao-Chung Kuo
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Julia Franzen
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr University Bochum, 44892 Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| |
Collapse
|
16
|
Kerschensteiner D, Feller MB. Mapping the Retina onto the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041512. [PMID: 38052498 PMCID: PMC10835620 DOI: 10.1101/cshperspect.a041512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Vision begins in the retina, which extracts salient features from the environment and encodes them in the spike trains of retinal ganglion cells (RGCs), the output neurons of the eye. RGC axons innervate diverse brain areas (>50 in mice) to support perception, guide behavior, and mediate influences of light on physiology and internal states. In recent years, complete lists of RGC types (∼45 in mice) have been compiled, detailed maps of their dendritic connections drawn, and their light responses surveyed at scale. We know less about the RGCs' axonal projection patterns, which map retinal information onto the brain. However, some organizing principles have emerged. Here, we review the strategies and mechanisms that govern developing RGC axons and organize their innervation of retinorecipient brain areas.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences
- Department of Neuroscience
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marla B Feller
- Department of Molecular and Cell Biology
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720, USA
| |
Collapse
|
17
|
Li J, Choi J, Cheng X, Ma J, Pema S, Sanes JR, Mardon G, Frankfort BJ, Tran NM, Li Y, Chen R. Comprehensive single-cell atlas of the mouse retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577060. [PMID: 38328114 PMCID: PMC10849744 DOI: 10.1101/2024.01.24.577060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has advanced our understanding of cellular heterogeneity at the single-cell resolution by classifying and characterizing cell types in multiple tissues and species. While several mouse retinal scRNA-seq reference datasets have been published, each dataset either has a relatively small number of cells or is focused on specific cell classes, and thus is suboptimal for assessing gene expression patterns across all retina types at the same time. To establish a unified and comprehensive reference for the mouse retina, we first generated the largest retinal scRNA-seq dataset to date, comprising approximately 190,000 single cells from C57BL/6J mouse whole retinas. This dataset was generated through the targeted enrichment of rare population cells via antibody-based magnetic cell sorting. By integrating this new dataset with public datasets, we conducted an integrated analysis to construct the Mouse Retina Cell Atlas (MRCA) for wild-type mice, which encompasses over 330,000 single cells. The MRCA characterizes 12 major classes and 138 cell types. It captured consensus cell type characterization from public datasets and identified additional new cell types. To facilitate the public use of the MRCA, we have deposited it in CELLxGENE, UCSC Cell Browser, and the Broad Single Cell Portal for visualization and gene expression exploration. The comprehensive MRCA serves as an easy-to-use, one-stop data resource for the mouse retina communities.
Collapse
Affiliation(s)
- Jin Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jongsu Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xuesen Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Justin Ma
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Shahil Pema
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02130, USA
| | - Graeme Mardon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
- Departments of Ophthalmology and Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Benjamin J. Frankfort
- Departments of Ophthalmology and Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Nicholas M. Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
18
|
Matsumoto A, Yonehara K. Emerging computational motifs: Lessons from the retina. Neurosci Res 2023; 196:11-22. [PMID: 37352934 DOI: 10.1016/j.neures.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
The retinal neuronal circuit is the first stage of visual processing in the central nervous system. The efforts of scientists over the last few decades indicate that the retina is not merely an array of photosensitive cells, but also a processor that performs various computations. Within a thickness of only ∼200 µm, the retina consists of diverse forms of neuronal circuits, each of which encodes different visual features. Since the discovery of direction-selective cells by Horace Barlow and Richard Hill, the mechanisms that generate direction selectivity in the retina have remained a fascinating research topic. This review provides an overview of recent advances in our understanding of direction-selectivity circuits. Beyond the conventional wisdom of direction selectivity, emerging findings indicate that the retina utilizes complicated and sophisticated mechanisms in which excitatory and inhibitory pathways are involved in the efficient encoding of motion information. As will become evident, the discovery of computational motifs in the retina facilitates an understanding of how sensory systems establish feature selectivity.
Collapse
Affiliation(s)
- Akihiro Matsumoto
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan; Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan.
| | - Keisuke Yonehara
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan; Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan
| |
Collapse
|
19
|
McCracken S, Fitzpatrick MJ, Hall AL, Wang Z, Kerschensteiner D, Morgan JL, Williams PR. Diversity in homeostatic calcium set points predicts retinal ganglion cell survival following optic nerve injury in vivo. Cell Rep 2023; 42:113165. [PMID: 37751356 PMCID: PMC10947246 DOI: 10.1016/j.celrep.2023.113165] [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: 10/17/2022] [Revised: 06/29/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Retinal ganglion cell (RGC) degeneration drives vision loss in blinding conditions. RGC death is often triggered by axon degeneration in the optic nerve. Here, we study the contributions of dynamic and homeostatic Ca2+ levels to RGC death from axon injury. We find that axonal Ca2+ elevations from optic nerve injury do not propagate over distance or reach RGC somas, and acute and chronic Ca2+ dynamics do not affect RGC survival. Instead, we discover that baseline Ca2+ levels vary widely between RGCs and predict their survival after axon injury, and that lowering these levels reduces RGC survival. Further, we find that well-surviving RGC types have higher baseline Ca2+ levels than poorly surviving types. Finally, we observe considerable variation in the baseline Ca2+ levels of different RGCs of the same type, which are predictive of within-type differences in survival.
Collapse
Affiliation(s)
- Sean McCracken
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael J Fitzpatrick
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Allison L Hall
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Postbaccalaureate Program in Developmental Biology & Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zelun Wang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- John F. Hardesty, MD 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; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Josh L Morgan
- John F. Hardesty, MD 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
| | - Philip R Williams
- John F. Hardesty, MD 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; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
20
|
Prigge CL, Dembla M, Sharma A, El-Quessny M, Kozlowski C, Paisley CE, Miltner AM, Johnson TM, Della Santina L, Feller MB, Kay JN. Rejection of inappropriate synaptic partners in mouse retina mediated by transcellular FLRT2-UNC5 signaling. Dev Cell 2023; 58:2080-2096.e7. [PMID: 37557174 PMCID: PMC10615732 DOI: 10.1016/j.devcel.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/26/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
During nervous system development, neurons choose synaptic partners with remarkable specificity; however, the cell-cell recognition mechanisms governing rejection of inappropriate partners remain enigmatic. Here, we show that mouse retinal neurons avoid inappropriate partners by using the FLRT2-uncoordinated-5 (UNC5) receptor-ligand system. Within the inner plexiform layer (IPL), FLRT2 is expressed by direction-selective (DS) circuit neurons, whereas UNC5C/D are expressed by non-DS neurons projecting to adjacent IPL sublayers. In vivo gain- and loss-of-function experiments demonstrate that FLRT2-UNC5 binding eliminates growing DS dendrites that have strayed from the DS circuit IPL sublayers. Abrogation of FLRT2-UNC5 binding allows mistargeted arbors to persist, elaborate, and acquire synapses from inappropriate partners. Conversely, UNC5C misexpression within DS circuit sublayers inhibits dendrite growth and drives arbors into adjacent sublayers. Mechanistically, UNC5s promote dendrite elimination by interfering with FLRT2-mediated adhesion. Based on their broad expression, FLRT-UNC5 recognition is poised to exert widespread effects upon synaptic partner choices across the nervous system.
Collapse
Affiliation(s)
- Cameron L Prigge
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Mayur Dembla
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Arsha Sharma
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Malak El-Quessny
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher Kozlowski
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Caitlin E Paisley
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Adam M Miltner
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Tyler M Johnson
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Luca Della Santina
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX 77204, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeremy N Kay
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA.
| |
Collapse
|
21
|
Benowitz LI, Xie L, Yin Y. Inflammatory Mediators of Axon Regeneration in the Central and Peripheral Nervous Systems. Int J Mol Sci 2023; 24:15359. [PMID: 37895039 PMCID: PMC10607492 DOI: 10.3390/ijms242015359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Although most pathways in the mature central nervous system cannot regenerate when injured, research beginning in the late 20th century has led to discoveries that may help reverse this situation. Here, we highlight research in recent years from our laboratory identifying oncomodulin (Ocm), stromal cell-derived factor (SDF)-1, and chemokine CCL5 as growth factors expressed by cells of the innate immune system that promote axon regeneration in the injured optic nerve and elsewhere in the central and peripheral nervous systems. We also review the role of ArmC10, a newly discovered Ocm receptor, in mediating many of these effects, and the synergy between inflammation-derived growth factors and complementary strategies to promote regeneration, including deleting genes encoding cell-intrinsic suppressors of axon growth, manipulating transcription factors that suppress or promote the expression of growth-related genes, and manipulating cell-extrinsic suppressors of axon growth. In some cases, combinatorial strategies have led to unprecedented levels of nerve regeneration. The identification of some similar mechanisms in human neurons offers hope that key discoveries made in animal models may eventually lead to treatments to improve outcomes after neurological damage in patients.
Collapse
Affiliation(s)
- Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Wang B, Zhang Y. Asymmetric connections with starburst amacrine cells underlie the upward motion selectivity of J-type retinal ganglion cells. PLoS Biol 2023; 21:e3002301. [PMID: 37721959 PMCID: PMC10538761 DOI: 10.1371/journal.pbio.3002301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/28/2023] [Accepted: 08/17/2023] [Indexed: 09/20/2023] Open
Abstract
Motion is an important aspect of visual information. The directions of visual motion are encoded in the retina by direction-selective ganglion cells (DSGCs). ON-OFF DSGCs and ON DSGCs co-stratify with starburst amacrine cells (SACs) in the inner plexiform layer and depend on SACs for their direction selectivity. J-type retinal ganglion cells (J-RGCs), a type of OFF DSGCs in the mouse retina, on the other hand, do not co-stratify with SACs, and how direction selectivity in J-RGCs emerges has not been understood. Here, we report that both the excitatory and inhibitory synaptic inputs to J-RGCs are direction-selective (DS), with the inhibitory inputs playing a more important role for direction selectivity. The DS inhibitory inputs come from SACs, and the functional connections between J-RGCs and SACs are spatially asymmetric. Thus, J-RGCs and SACs form functionally important synaptic contacts even though their dendritic arbors show little overlap. These findings underscore the need to look beyond the neurons' stratification patterns in retinal circuit studies. Our results also highlight the critical role of SACs for retinal direction selectivity.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yifeng Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Mani A, Yang X, Zhao TA, Leyrer ML, Schreck D, Berson DM. A circuit suppressing retinal drive to the optokinetic system during fast image motion. Nat Commun 2023; 14:5142. [PMID: 37612305 PMCID: PMC10447436 DOI: 10.1038/s41467-023-40527-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Optokinetic nystagmus (OKN) assists stabilization of the retinal image during head rotation. OKN is driven by ON direction selective retinal ganglion cells (ON DSGCs), which encode both the direction and speed of global retinal slip. The synaptic circuits responsible for the direction selectivity of ON DSGCs are well understood, but those sculpting their slow-speed preference remain enigmatic. Here, we probe this mechanism in mouse retina through patch clamp recordings, functional imaging, genetic manipulation, and electron microscopic reconstructions. We confirm earlier evidence that feedforward glycinergic inhibition is the main suppressor of ON DSGC responses to fast motion, and reveal the source for this inhibition-the VGluT3 amacrine cell, a dual neurotransmitter, excitatory/inhibitory interneuron. Together, our results identify a role for VGluT3 cells in limiting the speed range of OKN. More broadly, they suggest VGluT3 cells shape the response of many retinal cell types to fast motion, suppressing it in some while enhancing it in others.
Collapse
Affiliation(s)
- Adam Mani
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Xinzhu Yang
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Tiffany A Zhao
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Megan L Leyrer
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Daniel Schreck
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - David M Berson
- Department of Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
24
|
Montijn JS, Riguccini V, Levelt CN, Heimel JA. Impaired Direction Selectivity in the Nucleus of the Optic Tract of Albino Mice. Invest Ophthalmol Vis Sci 2023; 64:9. [PMID: 37548962 PMCID: PMC10411648 DOI: 10.1167/iovs.64.11.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Purpose Human albinos have a low visual acuity. This is partially due to the presence of spontaneous erroneous eye movements called pendular nystagmus. This nystagmus is present in other albino vertebrates and has been hypothesized to be caused by aberrant wiring of retinal ganglion axons to the nucleus of the optic tract (NOT), a part of the accessory optic system involved in the optokinetic response to visual motion. The NOT in pigmented rodents is preferentially responsive to ipsiversive motion (i.e., motion in the contralateral visual field in the temporonasal direction). We compared the response to visual motion in the NOT of albino and pigmented mice to understand if motion coding and preference are impaired in the NOT of albino mice. Methods We recorded neuronal spiking activity with Neuropixels probes in the visual cortex and NOT in C57BL/6JRj mice (pigmented) and DBA/1JRj mice with oculocutaneous albinism (albino). Results We found that in pigmented mice, NOT is retinotopically organized, and neurons are direction tuned, whereas in albino mice, neuronal tuning is severely impaired. Neurons in the NOT of albino mice do not have a preference for ipsiversive movement. In contrast, neuronal tuning in visual cortex was preserved in albino mice and did not differ significantly from the tuning in pigmented mice. Conclusions We propose that excessive interhemispheric crossing of retinal projections in albinos may cause the disrupted left/right direction encoding we found in NOT. This, in turn, impairs the normal horizontal optokinetic reflex and leads to pendular albino nystagmus.
Collapse
Affiliation(s)
- Jorrit S. Montijn
- Department of Circuits, Structure & Function, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Valentina Riguccini
- Department of Molecular Visual Plasticity, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Christiaan N. Levelt
- Department of Molecular Visual Plasticity, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, the Netherlands
| | - J. Alexander Heimel
- Department of Circuits, Structure & Function, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Peng YR. Cell-type specification in the retina: Recent discoveries from transcriptomic approaches. Curr Opin Neurobiol 2023; 81:102752. [PMID: 37499619 DOI: 10.1016/j.conb.2023.102752] [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: 05/16/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
Understanding the formation of the complex nervous system hinges on decoding the mechanism that specifies a vast array of neuronal types, each endowed with a unique morphology, physiology, and connectivity. As a pivotal step towards addressing this problem, seminal work has been devoted to characterizing distinct neuronal types. In recent years, high-throughput, single-cell transcriptomic methods have enabled a rapid inventory of cell types in various regions of the nervous system, with the retina exhibiting complete molecular characterization across many vertebrate species. This invaluable resource has furnished a fresh perspective for investigating the molecular principles of cell-type specification, thereby advancing our understanding of retinal development. Accordingly, this review focuses on the most recent transcriptomic characterizations of retinal cells, with a particular focus on amacrine cells and retinal ganglion cells. These investigations have unearthed new insights into their cell-type specification.
Collapse
Affiliation(s)
- Yi-Rong Peng
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Szalak R, Matysek M, Mozel S, Arciszewski MB. Cocaine- and Amphetamine-Regulated Transcript (CART) Peptide Is Co-Expressed with Parvalbumin, Neuropeptide Y and Somatostatin in the Claustrum of the Chinchilla. Animals (Basel) 2023; 13:2177. [PMID: 37443975 DOI: 10.3390/ani13132177] [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: 05/19/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Although for many years, researchers have been working on understanding the function of the cocaine- and amphetamine-regulated transcript (CART) peptide at the central- and peripheral-nervous-system level, data describing the presence of CART in the claustrum are still missing. Therefore, the aim of the present study was to immunohistochemically investigate the CART expression in the claustrum neurons in chinchillas as well as the CART co-localization with somatostatin (SOM), parvalbumin (PV), and neuropeptide Y (NPY) using double-immunohistochemical staining. The claustrum is divided into two main parts: the dorsal segment (CL), which is located above the rhinal fissure, and the ventral segment (EN), located below the rhinal fissure. The presence of HU C/D-IR CART-IR-positive neurons was detected in both the insular claustrum (CL) and the endopiriform nucleus (EN). The vast majority of CART-IR neurons were predominantly small and medium in size and were evenly scattered throughout the claustrum. CART co-localization with selected neurotransmitters/neuromodulators (SOM, NPY, and PV) showed the presence of a CART-IR reaction only in the neurons, while the nerve fibers were, in all cases, devoid of the CART-IR response. Our research supplements missing knowledge about the distribution and co-localization pattern of CART with SOM, NPY, and PV in the chinchilla claustrum, and also provides a better understanding of the similarities and differences compared to other species of rodents and other mammals.
Collapse
Affiliation(s)
- Radosław Szalak
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, 12 Akademicka St., 20-950 Lublin, Poland
| | - Małgorzata Matysek
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, 12 Akademicka St., 20-950 Lublin, Poland
| | - Sylwia Mozel
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, 12 Akademicka St., 20-950 Lublin, Poland
| | - Marcin B Arciszewski
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, 12 Akademicka St., 20-950 Lublin, Poland
| |
Collapse
|
27
|
Liu Y, Savier EL, DePiero VJ, Chen C, Schwalbe DC, Abraham-Fan RJ, Chen H, Campbell JN, Cang J. Mapping visual functions onto molecular cell types in the mouse superior colliculus. Neuron 2023; 111:1876-1886.e5. [PMID: 37086721 PMCID: PMC10330256 DOI: 10.1016/j.neuron.2023.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023]
Abstract
The superficial superior colliculus (sSC) carries out diverse roles in visual processing and behaviors, but how these functions are delegated among collicular neurons remains unclear. Here, using single-cell transcriptomics, we identified 28 neuron subtypes and subtype-enriched marker genes from tens of thousands of adult mouse sSC neurons. We then asked whether the sSC's molecular subtypes are tuned to different visual stimuli. Specifically, we imaged calcium dynamics in single sSC neurons in vivo during visual stimulation and then mapped marker gene transcripts onto the same neurons ex vivo. Our results identify a molecular subtype of inhibitory neuron accounting for ∼50% of the sSC's direction-selective cells, suggesting a genetic logic for the functional organization of the sSC. In addition, our studies provide a comprehensive molecular atlas of sSC neuron subtypes and a multimodal mapping method that will facilitate investigation of their respective functions, connectivity, and development.
Collapse
Affiliation(s)
- Yuanming Liu
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elise L Savier
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Victor J DePiero
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA
| | - Dana C Schwalbe
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | - Hui Chen
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
28
|
Harris SC, Dunn FA. Asymmetric retinal direction tuning predicts optokinetic eye movements across stimulus conditions. eLife 2023; 12:e81780. [PMID: 36930180 PMCID: PMC10023158 DOI: 10.7554/elife.81780] [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: 07/11/2022] [Accepted: 02/02/2023] [Indexed: 03/18/2023] Open
Abstract
Across species, the optokinetic reflex (OKR) stabilizes vision during self-motion. OKR occurs when ON direction-selective retinal ganglion cells (oDSGCs) detect slow, global image motion on the retina. How oDSGC activity is integrated centrally to generate behavior remains unknown. Here, we discover mechanisms that contribute to motion encoding in vertically tuned oDSGCs and leverage these findings to empirically define signal transformation between retinal output and vertical OKR behavior. We demonstrate that motion encoding in vertically tuned oDSGCs is contrast-sensitive and asymmetric for oDSGC types that prefer opposite directions. These phenomena arise from the interplay between spike threshold nonlinearities and differences in synaptic input weights, including shifts in the balance of excitation and inhibition. In behaving mice, these neurophysiological observations, along with a central subtraction of oDSGC outputs, accurately predict the trajectories of vertical OKR across stimulus conditions. Thus, asymmetric tuning across competing sensory channels can critically shape behavior.
Collapse
Affiliation(s)
- Scott C Harris
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Neuroscience Graduate Program, University of California, San FranciscoSan FranciscoUnited States
| | - Felice A Dunn
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
29
|
Nadal-Nicolás FM, Galindo-Romero C, Lucas-Ruiz F, Marsh-Amstrong N, Li W, Vidal-Sanz M, Agudo-Barriuso M. Pan-retinal ganglion cell markers in mice, rats, and rhesus macaques. Zool Res 2023; 44:226-248. [PMID: 36594396 PMCID: PMC9841181 DOI: 10.24272/j.issn.2095-8137.2022.308] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Univocal identification of retinal ganglion cells (RGCs) is an essential prerequisite for studying their degeneration and neuroprotection. Before the advent of phenotypic markers, RGCs were normally identified using retrograde tracing of retinorecipient areas. This is an invasive technique, and its use is precluded in higher mammals such as monkeys. In the past decade, several RGC markers have been described. Here, we reviewed and analyzed the specificity of nine markers used to identify all or most RGCs, i.e., pan-RGC markers, in rats, mice, and macaques. The best markers in the three species in terms of specificity, proportion of RGCs labeled, and indicators of viability were BRN3A, expressed by vision-forming RGCs, and RBPMS, expressed by vision- and non-vision-forming RGCs. NEUN, often used to identify RGCs, was expressed by non-RGCs in the ganglion cell layer, and therefore was not RGC-specific. γ-SYN, TUJ1, and NF-L labeled the RGC axons, which impaired the detection of their somas in the central retina but would be good for studying RGC morphology. In rats, TUJ1 and NF-L were also expressed by non-RGCs. BM88, ERRβ, and PGP9.5 are rarely used as markers, but they identified most RGCs in the rats and macaques and ERRβ in mice. However, PGP9.5 was also expressed by non-RGCs in rats and macaques and BM88 and ERRβ were not suitable markers of viability.
Collapse
Affiliation(s)
- Francisco M Nadal-Nicolás
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-2510, USA
| | - Caridad Galindo-Romero
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
| | - Fernando Lucas-Ruiz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain
| | - Nicholas Marsh-Amstrong
- Department of Ophthalmology and Vision Science, University of California, Davis, CA 95817, USA
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-2510, USA
| | - Manuel Vidal-Sanz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain. E-mail:
| | - Marta Agudo-Barriuso
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Murcia 30120, Spain
- Dpto. Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia 30120, Spain. E-mail:
| |
Collapse
|
30
|
Huang W, Xu Q, Su J, Tang L, Hao ZZ, Xu C, Liu R, Shen Y, Sang X, Xu N, Tie X, Miao Z, Liu X, Xu Y, Liu F, Liu Y, Liu S. Linking transcriptomes with morphological and functional phenotypes in mammalian retinal ganglion cells. Cell Rep 2022; 40:111322. [PMID: 36103830 DOI: 10.1016/j.celrep.2022.111322] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/19/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022] Open
Abstract
Retinal ganglion cells (RGCs) are the brain's gateway to the visual world. They can be classified into different types on the basis of their electrophysiological, transcriptomic, or morphological characteristics. Here, we characterize the transcriptomic, morphological, and functional features of 472 high-quality RGCs using Patch sequencing (Patch-seq), providing functional and morphological annotation of many transcriptomic-defined cell types of a previously established RGC atlas. We show a convergence of different modalities in defining the RGC identity and reveal the degree of correspondence for well-characterized cell types across multimodal data. Moreover, we complement some RGC types with detailed morphological and functional properties. We also identify differentially expressed genes among ON, OFF, and ON-OFF RGCs such as Vat1l, Slitrk6, and Lmo7, providing candidate marker genes for functional studies. Our research suggests that the molecularly distinct clusters may also differ in their roles of encoding visual information.
Collapse
Affiliation(s)
- Wanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qiang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jing Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuhui Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xiaoxiu Tie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhichao Miao
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Ying Xu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Feng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing 100085, China.
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou 510080, China.
| |
Collapse
|
31
|
Gao S, Xiang SY, Song ZW, Han YN, Zhang YN, Hao Y. Motion detection and direction recognition in a photonic spiking neural network consisting of VCSELs-SA. OPTICS EXPRESS 2022; 30:31701-31713. [PMID: 36242247 DOI: 10.1364/oe.465653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/04/2022] [Indexed: 06/16/2023]
Abstract
Motion detection and direction recognition are two important fundamental visual functions among the many cognitive functions performed by the human visual system. The retina and visual cortex are indispensable for composing the visual nervous system. The retina is responsible for transmitting electrical signals converted from light signals to the visual cortex of the brain. We propose a photonic spiking neural network (SNN) based on vertical-cavity surface-emitting lasers with an embedding saturable absorber (VCSELs-SA) with temporal integration effects, and demonstrate that the motion detection and direction recognition tasks can be solved by mimicking the visual nervous system. Simulation results reveal that the proposed photonic SNN with a modified supervised algorithm combining the tempotron and the STDP rule can correctly detect the motion and recognize the direction angles, and is robust to time jitter and the current difference between VCSEL-SAs. The proposed approach adopts a low-power photonic neuromorphic system for real-time information processing, which provides theoretical support for the large-scale application of hardware photonic SNN in the future.
Collapse
|
32
|
Tapia ML, Nascimento-Dos-Santos G, Park KK. Subtype-specific survival and regeneration of retinal ganglion cells in response to injury. Front Cell Dev Biol 2022; 10:956279. [PMID: 36035999 PMCID: PMC9411869 DOI: 10.3389/fcell.2022.956279] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons that function synchronously to convey visual information through the optic nerve to retinorecipient target areas in the brain. Injury or disease to the optic nerve results in RGC degeneration and loss of visual function, as few RGCs survive, and even fewer can be provoked to regenerate their axons. Despite causative insults being broadly shared, regeneration studies demonstrate that RGC types exhibit differential resilience to injury and undergo selective survival and regeneration of their axons. While most early studies have identified these RGC types based their morphological and physiological characteristics, recent advances in transgenic and gene sequencing technologies have further enabled type identification based on unique molecular features. In this review, we provide an overview of the well characterized RGC types and identify those shown to preferentially survive and regenerate in various regeneration models. Furthermore, we discuss cellular characteristics of both the resilient and susceptible RGC types including the combinatorial expression of different molecular markers that identify these specific populations. Lastly, we discuss potential molecular mechanisms and genes found to be selectively expressed by specific types that may contribute to their reparative capacity. Together, we describe the studies that lay the important groundwork for identifying factors that promote neural regeneration and help advance the development of targeted therapy for the treatment of RGC degeneration as well as neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Mary L Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gabriel Nascimento-Dos-Santos
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
33
|
Kowal TJ, Dhande OS, Wang B, Wang Q, Ning K, Liu W, Berbari NF, Hu Y, Sun Y. Distribution of prototypical primary cilia markers in subtypes of retinal ganglion cells. J Comp Neurol 2022; 530:2176-2187. [PMID: 35434813 PMCID: PMC9219574 DOI: 10.1002/cne.25326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/27/2022] [Accepted: 03/21/2022] [Indexed: 11/07/2022]
Abstract
Loss of retinal ganglion cells (RGCs) underlies several forms of retinal disease including glaucomatous optic neuropathy, a leading cause of irreversible blindness. Several rare genetic disorders associated with cilia dysfunction have retinal degeneration as a clinical hallmark. Much of the focus of ciliopathy associated blindness is on the connecting cilium of photoreceptors; however, RGCs also possess primary cilia. It is unclear what roles RGC cilia play, what proteins and signaling machinery localize to RGC cilia, or how RGC cilia are differentiated across the subtypes of RGCs. To better understand these questions, we assessed the presence or absence of a prototypical cilia marker Arl13b and a widely distributed neuronal cilia marker AC3 in different subtypes of mouse RGCs. Interestingly, not all RGC subtype cilia are the same and there are significant differences even among these standard cilia markers. Alpha-RGCs positive for osteopontin, calretinin, and SMI32 primarily possess AC3-positive cilia. Directionally selective RGCs that are CART positive or Trhr positive localize either Arl13b or AC3, respectively, in cilia. Intrinsically photosensitive RGCs differentially localize Arl13b and AC3 based on melanopsin expression. Taken together, we characterized the localization of gold standard cilia markers in different subtypes of RGCs and conclude that cilia within RGC subtypes may be differentially organized. Future studies aimed at understanding RGC cilia function will require a fundamental ability to observe the cilia across subtypes as their signaling protein composition is elucidated. A comprehensive understanding of RGC cilia may reveal opportunities to understanding how their dysfunction leads to retinal degeneration.
Collapse
Affiliation(s)
- Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Onkar S. Dhande
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Wendy Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Nicolas F. Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis IN 46202 USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA 94304
| |
Collapse
|
34
|
Jiang Q, Litvina EY, Acarón Ledesma H, Shu G, Sonoda T, Wei W, Chen C. Functional convergence of on-off direction-selective ganglion cells in the visual thalamus. Curr Biol 2022; 32:3110-3120.e6. [PMID: 35793680 PMCID: PMC9438454 DOI: 10.1016/j.cub.2022.06.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 01/21/2023]
Abstract
In the mouse visual system, multiple types of retinal ganglion cells (RGCs) each encode distinct features of the visual space. A clear understanding of how this information is parsed in their downstream target, the dorsal lateral geniculate nucleus (dLGN), remains elusive. Here, we characterized retinogeniculate connectivity in Cart-IRES2-Cre-D and BD-CreER2 mice, which labels subsets of on-off direction-selective ganglion cells (ooDSGCs) tuned to the vertical directions and to only ventral motion, respectively. Our immunohistochemical, electrophysiological, and optogenetic experiments reveal that only a small fraction (<15%) of thalamocortical (TC) neurons in the dLGN receives primary retinal drive from these subtypes of ooDSGCs. The majority of the functionally identifiable ooDSGC inputs in the dLGN are weak and converge together with inputs from other RGC types. Yet our modeling indicates that this mixing is not random: BD-CreER+ ooDSGC inputs converge less frequently with ooDSGCs tuned to the opposite direction than with non-CART-Cre+ RGC types. Taken together, these results indicate that convergence of distinct information lines in dLGN follows specific rules of organization.
Collapse
Affiliation(s)
- Qiufen Jiang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Elizabeth Y Litvina
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA; National Institute of Neurological Disorders and Stroke, 6001 Executive Boulevard Suite 3309, Bethesda, MD 20824, USA
| | - Héctor Acarón Ledesma
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Guanhua Shu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Takuma Sonoda
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Wei Wei
- Department of Neurobiology, The University of Chicago, 947 East 58th Street, Chicago, IL 60637, USA
| | - Chinfei Chen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Huang X, Kim AJ, Acarón Ledesma H, Ding J, Smith RG, Wei W. Visual Stimulation Induces Distinct Forms of Sensitization of On-Off Direction-Selective Ganglion Cell Responses in the Dorsal and Ventral Retina. J Neurosci 2022; 42:4449-4469. [PMID: 35474276 PMCID: PMC9172291 DOI: 10.1523/jneurosci.1391-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Experience-dependent modulation of neuronal responses is a key attribute in sensory processing. In the mammalian retina, the On-Off direction-selective ganglion cell (DSGC) is well known for its robust direction selectivity. However, how the On-Off DSGC light responsiveness dynamically adjusts to the changing visual environment is underexplored. Here, we report that On-Off DSGCs tuned to posterior motion direction [i.e. posterior DSGCs (pDSGCs)] in mice of both sexes can be transiently sensitized by prior stimuli. Notably, distinct sensitization patterns are found in dorsal and ventral pDSGCs. Although responses of both dorsal and ventral pDSGCs to dark stimuli (Off responses) are sensitized, only dorsal cells show the sensitization of responses to bright stimuli (On responses). Visual stimulation to the dorsal retina potentiates a sustained excitatory input from Off bipolar cells, leading to tonic depolarization of pDSGCs. Such tonic depolarization propagates from the Off to the On dendritic arbor of the pDSGC to sensitize its On response. We also identified a previously overlooked feature of DSGC dendritic architecture that can support dendritic integration between On and Off dendritic layers bypassing the soma. By contrast, ventral pDSGCs lack a sensitized tonic depolarization and thus do not exhibit sensitization of their On responses. Our results highlight a topographic difference in Off bipolar cell inputs underlying divergent sensitization patterns of dorsal and ventral pDSGCs. Moreover, substantial crossovers between dendritic layers of On-Off DSGCs suggest an interactive dendritic algorithm for processing On and Off signals before they reach the soma.SIGNIFICANCE STATEMENT Visual neuronal responses are dynamically influenced by the prior visual experience. This form of plasticity reflects the efficient coding of the naturalistic environment by the visual system. We found that a class of retinal output neurons, On-Off direction-selective ganglion cells, transiently increase their responsiveness after visual stimulation. Cells located in dorsal and ventral retinas exhibit distinct sensitization patterns because of different adaptive properties of Off bipolar cell signaling. A previously overlooked dendritic morphologic feature of the On-Off direction-selective ganglion cell is implicated in the cross talk between On and Off pathways during sensitization. Together, these findings uncover a topographic difference in the adaptive encoding of upper and lower visual fields and the underlying neural mechanism in the dorsal and ventral retinas.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Neurobiology, The University of Chicago, Chicago, Illinois 60637
- The Committee on Neurobiology Graduate Program, The University of Chicago, Chicago, Illinois 60637
| | - Alan Jaehyun Kim
- Department of Neurobiology, The University of Chicago, Chicago, Illinois 60637
| | - Héctor Acarón Ledesma
- Department of Neurobiology, The University of Chicago, Chicago, Illinois 60637
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, Illinois 60637
| | - Jennifer Ding
- Department of Neurobiology, The University of Chicago, Chicago, Illinois 60637
- The Committee on Neurobiology Graduate Program, The University of Chicago, Chicago, Illinois 60637
| | - Robert G Smith
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Wei Wei
- Department of Neurobiology, The University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
36
|
Huang X, Qiao H, Li H, Jiang Z. Bioinspired approach-sensitive neural network for collision detection in cluttered and dynamic backgrounds. Appl Soft Comput 2022. [DOI: 10.1016/j.asoc.2022.108782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
37
|
Noailles A, Kutsyr O, Mayordomo-Febrer A, Lax P, López-Murcia M, Sanz-González SM, Pinazo-Durán MD, Cuenca N. Sodium Hyaluronate-Induced Ocular Hypertension in Rats Damages the Direction-Selective Circuit and Inner/Outer Retinal Plexiform Layers. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 35503230 PMCID: PMC9078050 DOI: 10.1167/iovs.63.5.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To assess the changes in retinal morphology in a rat model of chronic glaucoma induced by ocular hypertension. Methods Intraocular pressure (IOP) was surgically increased through weekly injections of sodium hyaluronate (HYA) in the anterior eye chamber of the left eye of male Wistar rats, whereas the right eyes were sham operated (salt solution). During the 10-week experimental period, IOP was measured weekly with a rebound tonometer. Retinal cryosections were prepared for histological/immunohistochemical analysis and morphometry. Results IOP was higher in HYA-treated eyes than in sham-operated eyes along the 10-week period, which was significant from the fourth to the nineth week. Ocular hypertension in HYA-treated eyes was associated with morphologic and morphometric changes in bipolar cells, ON-OFF direction-selective ganglion cells, ON/OFF starburst amacrine cells, and inner plexiform layer sublamina. Conclusions Serial HYA treatment in the rat anterior eye chamber results in mild-to-moderate elevated and sustained IOP and ganglion cell death, which mimics most human open-angle glaucoma hallmarks. The reduced number of direction-selective ganglion cells and starburst amacrine cells accompanied by a deteriorated ON/OFF plexus in this glaucoma model could lend insight to the abnormalities in motion perception observed in patients with glaucoma.
Collapse
Affiliation(s)
- Agustina Noailles
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - Oksana Kutsyr
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - Aloma Mayordomo-Febrer
- Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universidad CEU Cardenal Herrera, Valencia, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Mixed Research Unit for Visual Health and Veterinary Ophthalmology CEU/FISABIO, Valencia, Spain
| | - Pedro Lax
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| | - María López-Murcia
- Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universidad CEU Cardenal Herrera, Valencia, Spain.,Mixed Research Unit for Visual Health and Veterinary Ophthalmology CEU/FISABIO, Valencia, Spain
| | - Silvia M Sanz-González
- OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Cellular and Molecular Ophthalmo-biology Research Group, Department of Surgery, University of Valencia, Valencia, Spain.,Ophthalmic Research Unit "Santiago Grisolía"/FISABIO, Valencia, Spain
| | - María Dolores Pinazo-Durán
- OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain.,Cellular and Molecular Ophthalmo-biology Research Group, Department of Surgery, University of Valencia, Valencia, Spain.,Ophthalmic Research Unit "Santiago Grisolía"/FISABIO, Valencia, Spain
| | - Nicolás Cuenca
- Physiology, Genetics and Microbiology, University of Alicante, Spain.,OFTARED. Spanish Net of Ophthalmic Research. Institute of health Carlos III, Madrid, Spain
| |
Collapse
|
38
|
Sokhadze G, Campbell PW, Charalambakis N, Govindaiah G, Guido W, McGee AW. Cre driver mouse lines for thalamocortical circuit mapping. J Comp Neurol 2022; 530:1049-1063. [PMID: 34545582 PMCID: PMC9891227 DOI: 10.1002/cne.25248] [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: 05/23/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Subpopulations of neurons and associated neural circuits can be targeted in mice with genetic tools in a highly selective manner for visualization and manipulation. However, there are not well-defined Cre "driver" lines that target the expression of Cre recombinase to thalamocortical (TC) neurons. Here, we characterize three Cre driver lines for the nuclei of the dorsal thalamus: Oligodendrocyte transcription factor 3 (Olig3)-Cre, histidine decarboxylase (HDC)-Cre, and corticotropin-releasing hormone (CRH)-Cre. We examined the postnatal distribution of Cre expression for each of these lines with the Cre-dependent reporter CAG-tdTomato (Ai9). Cre-dependent expression of tdTomato reveals that Olig3-Cre expresses broadly within the thalamus, including TC neurons and interneurons, while HDC-Cre and CRH-Cre each have unique patterns of expression restricted to TC neurons within and across the sensory relay nuclei of the dorsal thalamus. Cre expression is present by the time of natural birth in all three lines, underscoring their utility for developmental studies. To demonstrate the utility of these Cre drivers for studying sensory TC circuitry, we targeted the expression of channelrhodopsin-2 to thalamus from the CAG-COP4*H134R/EYFP (Ai32) allele with either HDC-Cre or CRH-Cre. Optogenetic activation of TC afferents in primary visual cortex was sufficient to measure frequency-dependent depression. Thus, these Cre drivers provide selective Cre-dependent gene expression in thalamus suitable for both anatomical and functional studies.
Collapse
Affiliation(s)
- Guela Sokhadze
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Peter W Campbell
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Naomi Charalambakis
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Gubbi Govindaiah
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Aaron W McGee
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
39
|
Tsai NY, Wang F, Toma K, Yin C, Takatoh J, Pai EL, Wu K, Matcham AC, Yin L, Dang EJ, Marciano DK, Rubenstein JL, Wang F, Ullian EM, Duan X. Trans-Seq maps a selective mammalian retinotectal synapse instructed by Nephronectin. Nat Neurosci 2022; 25:659-674. [PMID: 35524141 PMCID: PMC9172271 DOI: 10.1038/s41593-022-01068-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 03/30/2022] [Indexed: 12/21/2022]
Abstract
The mouse visual system serves as an accessible model to understand mammalian circuit wiring. Despite rich knowledge in retinal circuits, the long-range connectivity map from distinct retinal ganglion cell (RGC) types to diverse brain neuron types remains unknown. In this study, we developed an integrated approach, called Trans-Seq, to map RGCs to superior collicular (SC) circuits. Trans-Seq combines a fluorescent anterograde trans-synaptic tracer, consisting of codon-optimized wheat germ agglutinin fused to mCherry, with single-cell RNA sequencing. We used Trans-Seq to classify SC neuron types innervated by genetically defined RGC types and predicted a neuronal pair from αRGCs to Nephronectin-positive wide-field neurons (NPWFs). We validated this connection using genetic labeling, electrophysiology and retrograde tracing. We then used transcriptomic data from Trans-Seq to identify Nephronectin as a determinant for selective synaptic choice from αRGC to NPWFs via binding to Integrin α8β1. The Trans-Seq approach can be broadly applied for post-synaptic circuit discovery from genetically defined pre-synaptic neurons.
Collapse
Affiliation(s)
- Nicole Y Tsai
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program and Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Fei Wang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Kenichi Toma
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Chen Yin
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Jun Takatoh
- McGovern Institute for Brain Research, MIT Brain and Cognitive Sciences, Cambridge, MA, USA
| | - Emily L Pai
- Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Kongyan Wu
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Angela C Matcham
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Luping Yin
- McGovern Institute for Brain Research, MIT Brain and Cognitive Sciences, Cambridge, MA, USA
| | - Eric J Dang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Denise K Marciano
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John L Rubenstein
- Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Fan Wang
- McGovern Institute for Brain Research, MIT Brain and Cognitive Sciences, Cambridge, MA, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
40
|
Sokhadze G, Whyland KL, Bickford ME, Guido W. The organization of cholinergic projections in the visual thalamus of the mouse. J Comp Neurol 2022; 530:1081-1098. [PMID: 34448209 DOI: 10.1002/cne.25235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 11/11/2022]
Abstract
Cholinergic projections from the brainstem serve as important modulators of activity in visual thalamic nuclei such as the dorsal lateral geniculate nucleus (dLGN). While these projections have been studied in several mammals, a comprehensive examination of their organization in the mouse is lacking. We used the retrograde transport of viruses or cholera toxin subunit B (CTB) injected in the dLGN, immunocytochemical labeling with antibodies against choline acetyltransferase (ChAT), brain nitric oxide synthase (BNOS), and vesicular acetylcholine transporter (VAChT), ChAT-Cre mice crossed with a reporter line (Ai9), as well as brainstem virus injections in ChAT-Cre mice to examine the pattern of thalamic innervation from cholinergic neurons in the pedunculopontine tegmental nucleus (PPTg), laterodorsal tegmental nucleus (LDTg), and the parabigeminal nucleus (PBG). Retrograde tracing demonstrated that the dLGN receives input from the PPTg, LDTg, and PBG. Viral tracing in ChAT-Cre mice and retrograde tracing combined with immunocytochemistry revealed that many of these inputs originate from cholinergic neurons in the PBG and PPTg. Most notable was an extensive cholinergic projection from the PBG which innervated most of the contralateral dLGN, with an especially dense concentration in the dorsolateral shell, as well as a small region in the dorsomedial pole of the ipsilateral dLGN. The PPTg was found to provide a sparse somewhat diffuse innervation of the ipsilateral dLGN. Neurons in the PPTg co-expressed ChAT, BNOS, and VAChT, whereas PBG neurons expressed ChAT, but not BNOS or VAChT. These results highlight the presence of distinct cholinergic populations that innervate the mouse dLGN.
Collapse
Affiliation(s)
- Guela Sokhadze
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Kyle L Whyland
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Martha E Bickford
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
41
|
Tools and Biomarkers for the Study of Retinal Ganglion Cell Degeneration. Int J Mol Sci 2022; 23:ijms23084287. [PMID: 35457104 PMCID: PMC9025234 DOI: 10.3390/ijms23084287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The retina is part of the central nervous system, its analysis may provide an idea of the health and functionality, not only of the retina, but also of the entire central nervous system, as has been shown in Alzheimer’s or Parkinson’s diseases. Within the retina, the ganglion cells (RGC) are the neurons in charge of processing and sending light information to higher brain centers. Diverse insults and pathological states cause degeneration of RGC, leading to irreversible blindness or impaired vision. RGCs are the measurable endpoints in current research into experimental therapies and diagnosis in multiple ocular pathologies, like glaucoma. RGC subtype classifications are based on morphological, functional, genetical, and immunohistochemical aspects. Although great efforts are being made, there is still no classification accepted by consensus. Moreover, it has been observed that each RGC subtype has a different susceptibility to injury. Characterizing these subtypes together with cell death pathway identification will help to understand the degenerative process in the different injury and pathological models, and therefore prevent it. Here we review the known RGC subtypes, as well as the diagnostic techniques, probes, and biomarkers for programmed and unprogrammed cell death in RGC.
Collapse
|
42
|
Abstract
Retinal circuits transform the pixel representation of photoreceptors into the feature representations of ganglion cells, whose axons transmit these representations to the brain. Functional, morphological, and transcriptomic surveys have identified more than 40 retinal ganglion cell (RGC) types in mice. RGCs extract features of varying complexity; some simply signal local differences in brightness (i.e., luminance contrast), whereas others detect specific motion trajectories. To understand the retina, we need to know how retinal circuits give rise to the diverse RGC feature representations. A catalog of the RGC feature set, in turn, is fundamental to understanding visual processing in the brain. Anterograde tracing indicates that RGCs innervate more than 50 areas in the mouse brain. Current maps connecting RGC types to brain areas are rudimentary, as is our understanding of how retinal signals are transformed downstream to guide behavior. In this article, I review the feature selectivities of mouse RGCs, how they arise, and how they are utilized downstream. Not only is knowledge of the behavioral purpose of RGC signals critical for understanding the retinal contributions to vision; it can also guide us to the most relevant areas of visual feature space. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences; Department of Neuroscience; Department of Biomedical Engineering; and Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, Missouri, USA;
| |
Collapse
|
43
|
Nasir-Ahmad S, Vanstone KA, Novelli M, Lee SCS, Do MTH, Martin PR, Grünert U. Satb1 expression in retinal ganglion cells of marmosets, macaques, and humans. J Comp Neurol 2022; 530:923-940. [PMID: 34622958 PMCID: PMC8831458 DOI: 10.1002/cne.25258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022]
Abstract
Recent advances in single-cell RNA sequencing have enabled the molecular distinction of ganglion cell populations in mammalian retinas. Here we used antibodies against the transcription factor special AT-rich binding protein 1 (Satb1, a protein which is expressed by on-off direction-selective ganglion cells in mouse retina) to study Satb1 expression in the retina of marmosets (Callithrix jacchus), macaques (Macaca fascicularis), and humans. In all species, Satb1 was exclusively expressed in retinal ganglion cells. The Satb1 cells made up ∼2% of the ganglion cell population in the central retina of all species, rising to a maximum ∼7% in peripheral marmoset retina. Intracellular injections in marmoset and macaque retinas revealed that most Satb1 expressing ganglion cells are widefield ganglion cells. In marmoset, Satb1 cells have a densely branching dendritic tree and include broad and narrow thorny, recursive bistratified, and parasol cells, all of which show some costratification with the outer or inner cholinergic amacrine cells. The recursive bistratified cells showed the strongest costratification but did not show extensive cofasciculation as reported for on-off direction-selective ganglion cells in rabbit and rodent retinas. In macaque, Satb1 was not expressed in recursive bistratified cells, but in large sparsely branching cells. Our findings further support the idea that the expression of transcription factors in retinal ganglion cells is not conserved across Old World (human and macaque) and New World (marmoset) primates and provides a further step to link a molecular marker with specific cell types.
Collapse
Affiliation(s)
- Subha Nasir-Ahmad
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, Australia
| | - Kurt A Vanstone
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia
| | - Mario Novelli
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia
| | - Sammy C S Lee
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, Australia
| | - Michael Tri H Do
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Paul R Martin
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, Australia
| | - Ulrike Grünert
- Faculty of Medicine and Health, Save Sight Institute, and Discipline of Ophthalmology, The University of Sydney, Sydney, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, Australia
| |
Collapse
|
44
|
Shekhar K, Whitney IE, Butrus S, Peng YR, Sanes JR. Diversification of multipotential postmitotic mouse retinal ganglion cell precursors into discrete types. eLife 2022; 11:e73809. [PMID: 35191836 PMCID: PMC8956290 DOI: 10.7554/elife.73809] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
The genesis of broad neuronal classes from multipotential neural progenitor cells has been extensively studied, but less is known about the diversification of a single neuronal class into multiple types. We used single-cell RNA-seq to study how newly born (postmitotic) mouse retinal ganglion cell (RGC) precursors diversify into ~45 discrete types. Computational analysis provides evidence that RGC transcriptomic type identity is not specified at mitotic exit, but acquired by gradual, asynchronous restriction of postmitotic multipotential precursors. Some types are not identifiable until a week after they are generated. Immature RGCs may be specified to project ipsilaterally or contralaterally to the rest of the brain before their type identity emerges. Optimal transport inference identifies groups of RGC precursors with largely nonoverlapping fates, distinguished by selectively expressed transcription factors that could act as fate determinants. Our study provides a framework for investigating the molecular diversification of discrete types within a neuronal class.
Collapse
Affiliation(s)
- Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Center for Computational Biology; California Institute for Quantitative Biosciences, QB3, University of California, BerkeleyBerkeleyUnited States
- Biological Systems and Engineering Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; Center for Computational Biology; California Institute for Quantitative Biosciences, QB3, University of California, BerkeleyBerkeleyUnited States
| | - Yi-Rong Peng
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
- Department of Ophthalmology, Stein Eye Institute, UCLA David Geffen School of MedicineLos AngelesUnited States
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| |
Collapse
|
45
|
Jain V, Hanson L, Sethuramanujam S, Michaels T, Gawley J, Gregg RG, Pyle I, Zhang C, Smith RG, Berson D, McCall MA, Awatramani GB. Gain control by sparse, ultra-slow glycinergic synapses. Cell Rep 2022; 38:110410. [PMID: 35196487 DOI: 10.1016/j.celrep.2022.110410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/21/2021] [Accepted: 01/28/2022] [Indexed: 12/23/2022] Open
Abstract
In the retina, ON starburst amacrine cells (SACs) play a crucial role in the direction-selective circuit, but the sources of inhibition that shape their response properties remain unclear. Previous studies demonstrate that ∼95% of their inhibitory synapses are GABAergic, yet we find that the light-evoked inhibitory currents measured in SACs are predominantly glycinergic. Glycinergic inhibition is extremely slow, relying on non-canonical glycine receptors containing α4 subunits, and is driven by both the ON and OFF retinal pathways. These attributes enable glycine inputs to summate and effectively control the output gain of SACs, expanding the range over which they compute direction. Serial electron microscopic reconstructions reveal three specific types of ON and OFF narrow-field amacrine cells as the presumptive sources of glycinergic inhibition. Together, these results establish an unexpected role for specific glycinergic amacrine cells in the retinal computation of stimulus direction by SACs.
Collapse
Affiliation(s)
- Varsha Jain
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5 Canada
| | - Laura Hanson
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5 Canada
| | | | - Tracy Michaels
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5 Canada
| | - Jerram Gawley
- Department of Biology, University of Victoria, Victoria, BC V8W 3N5 Canada
| | - Ronald G Gregg
- Department of Ophthalmology & Visual Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Ian Pyle
- Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Chi Zhang
- Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Robert G Smith
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Berson
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Maureen A McCall
- Department of Ophthalmology & Visual Sciences, University of Louisville, Louisville, KY 40202, USA; Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY 40202, USA.
| | | |
Collapse
|
46
|
Niu F, Han P, Zhang J, She Y, Yang L, Yu J, Zhuang M, Tang K, Shi Y, Yang B, Liu C, Peng B, Ji SJ. The m 6A reader YTHDF2 is a negative regulator for dendrite development and maintenance of retinal ganglion cells. eLife 2022; 11:75827. [PMID: 35179492 PMCID: PMC8906807 DOI: 10.7554/elife.75827] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/16/2022] [Indexed: 11/29/2022] Open
Abstract
The precise control of growth and maintenance of the retinal ganglion cell (RGC) dendrite arborization is critical for normal visual functions in mammals. However, the underlying mechanisms remain elusive. Here, we find that the N6-methyladenosine (m6A) reader YTHDF2 is highly expressed in the mouse RGCs. Conditional knockout (cKO) of Ythdf2 in the retina leads to increased RGC dendrite branching, resulting in more synapses in the inner plexiform layer. Interestingly, the Ythdf2 cKO mice show improved visual acuity compared with control mice. We further demonstrate that Ythdf2 cKO in the retina protects RGCs from dendrite degeneration caused by the experimental acute glaucoma model. We identify the m6A-modified YTHDF2 target transcripts which mediate these effects. This study reveals mechanisms by which YTHDF2 restricts RGC dendrite development and maintenance. YTHDF2 and its target mRNAs might be valuable in developing new treatment approaches for glaucomatous eyes.
Collapse
Affiliation(s)
- Fugui Niu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Peng Han
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jian Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yuanchu She
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Lixin Yang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jun Yu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Mengru Zhuang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kezhen Tang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuwei Shi
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Baisheng Yang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chunqiao Liu
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bo Peng
- Department of Neurosurgery, Fudan University, Shanghai, China
| | - Sheng-Jian Ji
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
47
|
Differential susceptibility of retinal ganglion cell subtypes against neurodegenerative diseases. Graefes Arch Clin Exp Ophthalmol 2022; 260:1807-1821. [PMID: 35038014 DOI: 10.1007/s00417-022-05556-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/27/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Retinal ganglion cells (RGCs) are essential to propagate external visual information from the retina to the brain. Death of RGCs is speculated to be closely correlated with blinding retinal diseases, such as glaucoma and traumatic optic neuropathy (TON). Emerging innovative technologies have helped refine and standardize the classification of RGCs; at present, they are classified into more than 40 subpopulations in mammals. These RGC subtypes are identified by a combination of anatomical morphologies, electrophysiological functions, and genetic profiles. Increasing evidence suggests that neurodegenerative diseases do not collectively affect the RGCs. In fact, which RGC subtype exhibits the strongest or weakest susceptibility is hotly debated. Although a consensus has not yet been reached, it is certain that assorted RGCs display differential susceptibility against irreversible degeneration. Interestingly, a single RGC subtype can exhibit various vulnerabilities to optic nerve damage in diverse injury models. Thus, elucidating how susceptible RGC subtypes are to various injuries can protect vulnerable RGCs from damage and improve the possibility of preventing and treating visual impairment caused by neurodegenerative diseases. In this review, we summarize in detail the progress and status quo of research on the type-specific susceptibility of RGCs and point out current limitations and the possible directions for future research in this field.
Collapse
|
48
|
Stacy AK, Van Hooser SD. Development of Functional Properties in the Early Visual System: New Appreciations of the Roles of Lateral Geniculate Nucleus. Curr Top Behav Neurosci 2022; 53:3-35. [PMID: 35112333 DOI: 10.1007/7854_2021_297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the years following Hubel and Wiesel's first reports on ocular dominance plasticity and amblyopia, much attention has been focused on understanding the role of cortical circuits in developmental and experience-dependent plasticity. Initial studies found few differences between retinal ganglion cells and neurons in the lateral geniculate nucleus and uncovered little evidence for an impact of altered visual experience on the functional properties of lateral geniculate nucleus neurons. In the last two decades, however, studies have revealed that the connectivity between the retina and lateral geniculate nucleus is much richer than was previously appreciated, even revealing visual plasticity - including ocular dominance plasticity - in lateral geniculate nucleus neurons. Here we review the development of the early visual system and the impact of experience with a distinct focus on recent discoveries about lateral geniculate nucleus, its connectivity, and evidence for its plasticity and rigidity during development.
Collapse
Affiliation(s)
- Andrea K Stacy
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | |
Collapse
|
49
|
Ruff T, Peters C, Matsumoto A, Ihle SJ, Morales PA, Gaitanos L, Yonehara K, Del Toro D, Klein R. FLRT3 Marks Direction-Selective Retinal Ganglion Cells That Project to the Medial Terminal Nucleus. Front Mol Neurosci 2021; 14:790466. [PMID: 34955746 PMCID: PMC8696037 DOI: 10.3389/fnmol.2021.790466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/19/2021] [Indexed: 11/23/2022] Open
Abstract
The mammalian retina extracts a multitude of diverse features from the visual scene such as color, contrast, and direction of motion. These features are transmitted separately to the brain by more than 40 different retinal ganglion cell (RGC) subtypes. However, so far only a few genetic markers exist to fully characterize the different RGC subtypes. Here, we present a novel genetic Flrt3-CreERT2 knock-in mouse that labels a small subpopulation of RGCs. Using single-cell injection of fluorescent dyes in Flrt3 positive RGCs, we distinguished four morphological RGC subtypes. Anterograde tracings using a fluorescent Cre-dependent Adeno-associated virus (AAV) revealed that a subgroup of Flrt3 positive RGCs specifically project to the medial terminal nucleus (MTN), which is part of the accessory optic system (AOS) and is essential in driving reflex eye movements for retinal image stabilization. Functional characterization using ex vivo patch-clamp recordings showed that the MTN-projecting Flrt3 RGCs preferentially respond to downward motion in an ON-fashion. These neurons distribute in a regular pattern and most of them are bistratified at the level of the ON and OFF bands of cholinergic starburst amacrine cells where they express the known ON-OFF direction-selective RGC marker CART. Together, our results indicate that MTN-projecting Flrt3 RGCs represent a new functionally homogeneous AOS projecting direction-selective RGC subpopulation.
Collapse
Affiliation(s)
- Tobias Ruff
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Laboratory of Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Christian Peters
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Akihiro Matsumoto
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Stephan J Ihle
- Laboratory of Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Pilar Alcalá Morales
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Louise Gaitanos
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Keisuke Yonehara
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Daniel Del Toro
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Department of Biological Sciences, Faculty of Medicine, Institute of Neurosciences, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| | - Rüdiger Klein
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
50
|
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: 18] [Impact Index Per Article: 6.0] [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
|