1
|
Olguin AGR, Rochon PL, Theriault C, Brown T, Yao H, Cayouette M, Cook EP, Krishnaswamy A. Cadherin 4 assembles a family of color-preferring retinal circuits that respond to light offset. Curr Biol 2025; 35:1298-1310.e7. [PMID: 40081378 DOI: 10.1016/j.cub.2025.02.008] [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: 05/10/2024] [Revised: 11/22/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025]
Abstract
Retinal interneurons and projection neurons (retinal ganglion cells, RGCs) connect in specific combinations in a specialized neuropil called the inner plexiform layer (IPL). The IPL is divided into multiple sublaminae, with neurites of each neuronal type confined to one or a few layers. This laminar specificity is a major determinant of circuit specificity and circuit function. Using a combination of approaches, we show that RGCs targeting IPL sublaminae 1 and 3a (s1-s3a) express the cell adhesion molecule cadherin 4 (Cdh4). Using calcium imaging and iterative immunostaining, we classified Cdh4 RGCs into nine types that each encode unique aspects of dark visual stimuli. Cdh4 loss selectively disrupted the layer targeting of these RGCs, reduced their synaptic inputs from interneurons, and severely altered their visual responses. Overexpression of Cdh4 in other retinal neurons directed their neurites to s1-s3a through homophilic interactions. Taken together, these results demonstrate that Cdh4 is a novel layer-targeting system for nearly a quarter of all RGCs.
Collapse
Affiliation(s)
| | - Pierre-Luc Rochon
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | | | - Thomas Brown
- Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Houwen Yao
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Michel Cayouette
- Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Erik P Cook
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
2
|
Gupta HP, Azevedo AW, Chen YCHD, Xing K, Sims PA, Varol E, Mann RS. Decoding neuronal wiring by joint inference of cell identity and synaptic connectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.640006. [PMID: 40093165 PMCID: PMC11908227 DOI: 10.1101/2025.03.04.640006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Animal behaviors are executed by motor neurons (MNs), which receive information from complex pre-motor neuron (preMN) circuits and output commands to muscles. How motor circuits are established during development remains an important unsolved problem in neuroscience. Here we focus on the development of the motor circuits that control the movements of the adult legs in Drosophila melanogaster. After generating single-cell RNA sequencing (scRNAseq) datasets for leg MNs at multiple time points, we describe the time course of gene expression for multiple gene families. This analysis reveals that transcription factors (TFs) and cell adhesion molecules (CAMs) appear to drive the molecular diversity between individual MNs. In parallel, we introduce ConnectionMiner, a novel computational tool that integrates scRNAseq data with electron microscopy-derived connectomes. ConnectionMiner probabilistically refines ambiguous cell type annotations by leveraging neural wiring patterns, and, in turn, it identifies combinatorial gene expression signatures that correlate with synaptic connectivity strength. Applied to the Drosophila leg motor system, ConnectionMiner yields a comprehensive transcriptional annotation of both MNs and preMNs and uncovers candidate effector gene combinations that likely orchestrate the assembly of neural circuits from preMNs to MNs and ultimately to muscles.
Collapse
Affiliation(s)
| | - Anthony W. Azevedo
- Department of Neurobiology and Biophysics, University of Washington, WA, USA
| | | | - Kristi Xing
- Barnard College, Columbia University, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Erdem Varol
- Department of Computer Science & Engineering at Tandon School of Engineering, New York University, New York, NY, USA
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Richard S. Mann
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Wang J, Zhang L, Cavallini M, Pahlevan A, Sun J, Morshedian A, Fain GL, Sampath AP, Peng YR. Molecular characterization of the sea lamprey retina illuminates the evolutionary origin of retinal cell types. Nat Commun 2024; 15:10761. [PMID: 39737973 PMCID: PMC11685597 DOI: 10.1038/s41467-024-55019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
The lamprey, a primitive jawless vertebrate whose ancestors diverged from all other vertebrates over 500 million years ago, offers a unique window into the ancient formation of the retina. Using single-cell RNA-sequencing, we characterize retinal cell types in the lamprey and compare them to those in mouse, chicken, and zebrafish. We find six cell classes and 74 distinct cell types, many shared with other vertebrate species. The conservation of cell types indicates their emergence early in vertebrate evolution, highlighting primordial designs of retinal circuits for the rod pathway, ON-OFF discrimination, and direction selectivity. The diversification of amacrine and some ganglion cell types appears, however, to be distinct in the lamprey. We further infer genetic regulators in specifying retinal cell classes and identify ancestral regulatory elements across species, noting decreased conservation in specifying amacrine cells. Altogether, our characterization of the lamprey retina illuminates the evolutionary origin of visual processing in the retina.
Collapse
Affiliation(s)
- Junqiang Wang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lin Zhang
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martina Cavallini
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ali Pahlevan
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Junwei Sun
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ala Morshedian
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Gordon L Fain
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology and Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Shiga Y, Rangel Olguin AG, El Hajji S, Belforte N, Quintero H, Dotigny F, Alarcon-Martinez L, Krishnaswamy A, Di Polo A. Endoplasmic reticulum stress-related deficits in calcium clearance promote neuronal dysfunction that is prevented by SERCA2 gene augmentation. Cell Rep Med 2024; 5:101839. [PMID: 39615485 PMCID: PMC11722116 DOI: 10.1016/j.xcrm.2024.101839] [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: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Disruption of calcium (Ca2+) homeostasis in neurons is a hallmark of neurodegenerative diseases. Here, we investigate the mechanisms leading to Ca2+ dysregulation and ask whether altered Ca2+ dynamics impinge on neuronal stress and circuit dysfunction. Using two-photon microscopy, we show that ocular hypertension, a major risk factor in glaucoma, and optic nerve crush injury disrupt the capacity of retinal neurons to clear cytosolic Ca2+ leading to impaired light-evoked responses. Gene- and protein expression analysis reveal the loss of the sarco-endoplasmic reticulum (ER) Ca2+-ATPase2 pump (SERCA2/ATP2A2) in injured retinal neurons from mice and patients with primary open-angle glaucoma. Pharmacological activation or neuron-specific gene delivery of SERCA2 is sufficient to rescue single-cell Ca2+ dynamics and promote robust survival of damaged neurons. Furthermore, SERCA2 gene supplementation reduces ER stress, reestablishes circuit balance, and restores visual behaviors. Our findings reveal that enhancing the Ca2+ clearance capacity of vulnerable neurons alleviates organelle stress and promotes neurorecovery.
Collapse
Affiliation(s)
- Yukihiro Shiga
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | | | - Sana El Hajji
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada.
| |
Collapse
|
5
|
Rajendran Nair DS, Gupta A, Iseri E, Wei T, Phuong Quach LT, Seiler MJ, Lazzi G, Thomas BB. Extrinsic electric field modulates neuronal development and increases photoreceptor population in retinal organoids. Front Neurosci 2024; 18:1438903. [PMID: 39678532 PMCID: PMC11639233 DOI: 10.3389/fnins.2024.1438903] [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: 05/31/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Considering the significant role played by both intrinsic and extrinsic electric fields in the growth and maturation of the central nervous system, the impact of short exposure to external electric fields on the development and differentiation of retinal organoids was investigated. Methods Retinal organoids derived from human embryonic stem cells were used at day 80, a key stage in their differentiation. A single 60-minute exposure to a biphasic electrical field was administered to assess its influence on retinal cell populations and maturation markers. Immunohistochemistry, qPCR, and RNA sequencing were employed to evaluate cell type development and gene expression changes. Results Electrical stimulation significantly enhanced neuronal development and increased the population of photoreceptors within the organoids. RNA sequencing data showed upregulated expression of genes related to rod photoreceptors, Müller cells, horizontal cells, and amacrine cells, while genes associated with retinal pigment epithelium and retinal ganglion cells were downregulated. Variations in development and maturation were observed depending on the specific parameters of the applied electric field. Discussion These findings highlight the significant impact of extrinsic electrical fields on early retinal development and suggest that optimizing electrical field parameters could effectively address certain limitations in retinal organoid technology, potentially reducing the reliance on chemicals and small molecules.
Collapse
Affiliation(s)
- Deepthi S. Rajendran Nair
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Anika Gupta
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Ege Iseri
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Tianyuan Wei
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Le Tam Phuong Quach
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Magdalene J. Seiler
- Departments of Physical Medicine and Rehabilitation; Ophthalmology; Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | - Gianluca Lazzi
- Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Biju B. Thomas
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
6
|
Kosior-Jarecka E, Grzybowski A. Retinal Ganglion Cell Replacement in Glaucoma Therapy: A Narrative Review. J Clin Med 2024; 13:7204. [PMID: 39685661 DOI: 10.3390/jcm13237204] [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: 06/11/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide. It leads to the progressive degeneration of retinal ganglion cells (RGCs), the axons of which form the optic nerve. Enormous RGC apoptosis causes a lack of transfer of visual information to the brain. The RGC loss typical of the central nervous system is irreversible, and when glaucoma progresses, the total amount of RGCs in the retina enormously diminishes. The successful treatment in glaucoma patients is a direct neuroprotection by decreasing the intraocular pressure, which enables RGC protection but does not revive the lost ones. The intriguing new therapy for advanced glaucoma is the possibility of RGC replacement with new healthy cells. In this review article, the strategies regarding RGC replacement therapy are presented with the latest advances in the technique and the obstacles that it meets.
Collapse
Affiliation(s)
- Ewa Kosior-Jarecka
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University of Lublin, 20-079 Lublin, Poland
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, 60-836 Poznan, Poland
| |
Collapse
|
7
|
Qiao M. Deciphering the genetic code of neuronal type connectivity through bilinear modeling. eLife 2024; 12:RP91532. [PMID: 38857169 PMCID: PMC11164534 DOI: 10.7554/elife.91532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
Understanding how different neuronal types connect and communicate is critical to interpreting brain function and behavior. However, it has remained a formidable challenge to decipher the genetic underpinnings that dictate the specific connections formed between neuronal types. To address this, we propose a novel bilinear modeling approach that leverages the architecture similar to that of recommendation systems. Our model transforms the gene expressions of presynaptic and postsynaptic neuronal types, obtained from single-cell transcriptomics, into a covariance matrix. The objective is to construct this covariance matrix that closely mirrors a connectivity matrix, derived from connectomic data, reflecting the known anatomical connections between these neuronal types. When tested on a dataset of Caenorhabditis elegans, our model achieved a performance comparable to, if slightly better than, the previously proposed spatial connectome model (SCM) in reconstructing electrical synaptic connectivity based on gene expressions. Through a comparative analysis, our model not only captured all genetic interactions identified by the SCM but also inferred additional ones. Applied to a mouse retinal neuronal dataset, the bilinear model successfully recapitulated recognized connectivity motifs between bipolar cells and retinal ganglion cells, and provided interpretable insights into genetic interactions shaping the connectivity. Specifically, it identified unique genetic signatures associated with different connectivity motifs, including genes important to cell-cell adhesion and synapse formation, highlighting their role in orchestrating specific synaptic connections between these neurons. Our work establishes an innovative computational strategy for decoding the genetic programming of neuronal type connectivity. It not only sets a new benchmark for single-cell transcriptomic analysis of synaptic connections but also paves the way for mechanistic studies of neural circuit assembly and genetic manipulation of circuit wiring.
Collapse
Affiliation(s)
- Mu Qiao
- LinkedInMountain ViewUnited States
| |
Collapse
|
8
|
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
|
9
|
Vlasits AL, Syeda M, Wickman A, Guzman P, Schmidt TM. Atypical retinal function in a mouse model of Fragile X syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585283. [PMID: 38559003 PMCID: PMC10980068 DOI: 10.1101/2024.03.15.585283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Altered function of peripheral sensory neurons is an emerging mechanism for symptoms of autism spectrum disorders. Visual sensitivities are common in autism, but whether differences in the retina might underlie these sensitivities is not well-understood. We explored retinal function in the Fmr1 knockout model of Fragile X syndrome, focusing on a specific type of retinal neuron, the "sustained On alpha" retinal ganglion cell. We found that these cells exhibit changes in dendritic structure and dampened responses to light in the Fmr1 knockout. We show that decreased light sensitivity is due to increased inhibitory input and reduced E-I balance. The change in E-I balance supports maintenance of circuit excitability similar to what has been observed in cortex. These results show that loss of Fmr1 in the mouse retina affects sensory function of one retinal neuron type. Our findings suggest that the retina may be relevant for understanding visual function in Fragile X syndrome.
Collapse
Affiliation(s)
- Anna L Vlasits
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Ophthalmology, University of Illinois, Chicago, IL, USA
- Lead contact
| | - Maria Syeda
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Annelise Wickman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Pedro Guzman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
10
|
Woodworth MB, Greig LC, Goldberg JL. Intrinsic and Induced Neuronal Regeneration in the Mammalian Retina. Antioxid Redox Signal 2023; 39:1039-1052. [PMID: 37276181 PMCID: PMC10715439 DOI: 10.1089/ars.2023.0309] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Accepted: 05/14/2023] [Indexed: 06/07/2023]
Abstract
Significance: Retinal neurons are vulnerable to disease and injury, which can result in neuronal death and degeneration leading to irreversible vision loss. The human retina does not regenerate to replace neurons lost to disease or injury. However, cells within the retina of other animals are capable of regenerating neurons, and homologous cells within the mammalian retina could potentially be prompted to do the same. Activating evolutionarily silenced intrinsic regenerative capacity of the mammalian retina could slow, or even reverse, vision loss, leading to an improved quality of life for millions of people. Recent Advances: During development, neurons in the retina are generated progressively by retinal progenitor cells, with distinct neuron types born over developmental time. Many genes function in this process to specify the identity of newly generated neuron types, and these appropriate states of gene expression inform recent regenerative work. When regeneration is initiated in other vertebrates, including birds and fish, specific signaling pathways control the efficiency of regeneration, and these conserved pathways are likely to be important in mammals as well. Critical Issues: Using insights from development and from other animals, limited regeneration from intrinsic cell types has been demonstrated in the mammalian retina, but it is able only to generate a subset of partially differentiated retinal neuron types. Future Directions: Future studies should aim at increasing the efficiency of regeneration, activating regeneration in a targeted fashion across the retina, and improving the ability to generate specific types of retinal neurons to replace those lost to disease or injury. Antioxid. Redox Signal. 39, 1039-1052.
Collapse
Affiliation(s)
- Mollie B. Woodworth
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, USA
| | - Luciano C. Greig
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, USA
| | - Jeffrey L. Goldberg
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, USA
| |
Collapse
|
11
|
Hahn J, Monavarfeshani A, Qiao M, Kao AH, Kölsch Y, Kumar A, Kunze VP, Rasys AM, Richardson R, Wekselblatt JB, Baier H, Lucas RJ, Li W, Meister M, Trachtenberg JT, Yan W, Peng YR, Sanes JR, Shekhar K. Evolution of neuronal cell classes and types in the vertebrate retina. Nature 2023; 624:415-424. [PMID: 38092908 PMCID: PMC10719112 DOI: 10.1038/s41586-023-06638-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/13/2023] [Indexed: 12/17/2023]
Abstract
The basic plan of the retina is conserved across vertebrates, yet species differ profoundly in their visual needs1. Retinal cell types may have evolved to accommodate these varied needs, but this has not been systematically studied. Here we generated and integrated single-cell transcriptomic atlases of the retina from 17 species: humans, two non-human primates, four rodents, three ungulates, opossum, ferret, tree shrew, a bird, a reptile, a teleost fish and a lamprey. We found high molecular conservation of the six retinal cell classes (photoreceptors, horizontal cells, bipolar cells, amacrine cells, retinal ganglion cells (RGCs) and Müller glia), with transcriptomic variation across species related to evolutionary distance. Major subclasses were also conserved, whereas variation among cell types within classes or subclasses was more pronounced. However, an integrative analysis revealed that numerous cell types are shared across species, based on conserved gene expression programmes that are likely to trace back to an early ancestral vertebrate. The degree of variation among cell types increased from the outer retina (photoreceptors) to the inner retina (RGCs), suggesting that evolution acts preferentially to shape the retinal output. Finally, we identified rodent orthologues of midget RGCs, which comprise more than 80% of RGCs in the human retina, subserve high-acuity vision, and were previously believed to be restricted to primates2. By contrast, the mouse orthologues have large receptive fields and comprise around 2% of mouse RGCs. Projections of both primate and mouse orthologous types are overrepresented in the thalamus, which supplies the primary visual cortex. We suggest that midget RGCs are not primate innovations, but are descendants of evolutionarily ancient types that decreased in size and increased in number as primates evolved, thereby facilitating high visual acuity and increased cortical processing of visual information.
Collapse
Affiliation(s)
- Joshua Hahn
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Aboozar Monavarfeshani
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mu Qiao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- LinkedIn, Mountain View, CA, USA
| | - Allison H Kao
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yvonne Kölsch
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Ayush Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Vincent P Kunze
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ashley M Rasys
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Rose Richardson
- Division of Neuroscience and Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Joseph B Wekselblatt
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Robert J Lucas
- Division of Neuroscience and Centre for Biological Timing, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Markus Meister
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Joshua T Trachtenberg
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Wenjun Yan
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yi-Rong Peng
- Department of Ophthalmology, Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Joshua R Sanes
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA.
- Helen Wills Neuroscience Institute,Vision Science Graduate Group, University of California, Berkeley, Berkeley, CA, USA.
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Center for Computational Biology, Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA, USA.
- California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
12
|
Kerstein PC, Agreda YS, Curran BM, Ma L, Wright KM. Gbx2 controls amacrine cell dendrite stratification through Robo1/2 receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551861. [PMID: 37577554 PMCID: PMC10418232 DOI: 10.1101/2023.08.03.551861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Within the neuronal classes of the retina, amacrine cells (ACs) exhibit the greatest neuronal diversity in morphology and function. We show that the selective expression of the transcription factor Gbx2 is required for cell fate specification and dendritic stratification of an individual AC subtype in the mouse retina. We identify Robo1 and Robo2 as downstream effectors that when deleted, phenocopy the dendritic misprojections seen in Gbx2 mutants. Slit1 and Slit2, the ligands of Robo receptors, are localized to the OFF layers of the inner plexiform layer where we observe the dendritic misprojections in both Gbx2 and Robo1/2 mutants. We show that Robo receptors also are required for the proper dendritic stratification of additional AC subtypes, such as Vglut3+ ACs. These results show both that Gbx2 functions as a terminal selector in a single AC subtype and identify Slit-Robo signaling as a developmental mechanism for ON-OFF pathway segregation in the retina.
Collapse
|
13
|
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: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
14
|
Kiyama T, Altay HY, Badea TC, Mao CA. Pou4f1-Tbr1 transcriptional cascade controls the formation of Jam2-expressing retinal ganglion cells. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1175568. [PMID: 38469155 PMCID: PMC10926710 DOI: 10.3389/fopht.2023.1175568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/14/2023] [Indexed: 03/13/2024]
Abstract
More than 40 retinal ganglion cell (RGC) subtypes have been categorized in mouse based on their morphologies, functions, and molecular features. Among these diverse subtypes, orientation-selective Jam2-expressing RGCs (J-RGCs) has two unique morphologic characteristics: the ventral-facing dendritic arbor and the OFF-sublaminae stratified terminal dendrites in the inner plexiform layer. Previously, we have discovered that T-box transcription factor T-brain 1 (Tbr1) is expressed in J-RGCs. We further found that Tbr1 is essential for the expression of Jam2, and Tbr1 regulates the formation and the dendritic morphogenesis of J-RGCs. However, Tbr1 begins to express in terminally differentiated RGCs around perinatal stage, suggesting that it is unlikely involved in the initial fate determination for J-RGC and other upstream transcription factors must control Tbr1 expression and J-RGC formation. Using the Cleavage Under Targets and Tagmentation technique, we discovered that Pou4f1 binds to Tbr1 on the evolutionary conserved exon 6 and an intergenic region downstream of the 3'UTR, and on a region flanking the promoter and the first exon of Jam2. We showed that Pou4f1 is required for the expression of Tbr1 and Jam2, indicating Pou4f1 as a direct upstream regulator of Tbr1 and Jam2. Most interestingly, the Pou4f1-bound element in exon 6 of Tbr1 possesses high-level enhancer activity, capable of directing reporter gene expression in J-RGCs. Together, these data revealed a Pou4f1-Tbr1-Jam2 genetic hierarchy as a critical pathway in the formation of J-RGC subtype.
Collapse
Affiliation(s)
- Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Halit Y. Altay
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Tudor C. Badea
- Research and Development Institute, Transilvania University of Brasov, School of Medicine, Brasov, Romania
- National Center for Brain Research, Research Institute for Artificial Intelligence, Romanian Academy, Bucharest, Romania
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
15
|
Hahn J, Monavarfeshani A, Qiao M, Kao A, Kölsch Y, Kumar A, Kunze VP, Rasys AM, Richardson R, Baier H, Lucas RJ, Li W, Meister M, Trachtenberg JT, Yan W, Peng YR, Sanes JR, Shekhar K. Evolution of neuronal cell classes and types in the vertebrate retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536039. [PMID: 37066415 PMCID: PMC10104162 DOI: 10.1101/2023.04.07.536039] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The basic plan of the retina is conserved across vertebrates, yet species differ profoundly in their visual needs (Baden et al., 2020). One might expect that retinal cell types evolved to accommodate these varied needs, but this has not been systematically studied. Here, we generated and integrated single-cell transcriptomic atlases of the retina from 17 species: humans, two non-human primates, four rodents, three ungulates, opossum, ferret, tree shrew, a teleost fish, a bird, a reptile and a lamprey. Molecular conservation of the six retinal cell classes (photoreceptors, horizontal cells, bipolar cells, amacrine cells, retinal ganglion cells [RGCs] and Muller glia) is striking, with transcriptomic differences across species correlated with evolutionary distance. Major subclasses are also conserved, whereas variation among types within classes or subclasses is more pronounced. However, an integrative analysis revealed that numerous types are shared across species based on conserved gene expression programs that likely trace back to the common ancestor of jawed vertebrates. The degree of variation among types increases from the outer retina (photoreceptors) to the inner retina (RGCs), suggesting that evolution acts preferentially to shape the retinal output. Finally, we identified mammalian orthologs of midget RGCs, which comprise >80% of RGCs in the human retina, subserve high-acuity vision, and were believed to be primate-specific (Berson, 2008); in contrast, the mouse orthologs comprise <2% of mouse RGCs. Projections both primate and mouse orthologous types are overrepresented in the thalamus, which supplies the primary visual cortex. We suggest that midget RGCs are not primate innovations, but descendants of evolutionarily ancient types that decreased in size and increased in number as primates evolved, thereby facilitating high visual acuity and increased cortical processing of visual information.
Collapse
Affiliation(s)
- Joshua Hahn
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aboozar Monavarfeshani
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA 02138, USA
| | - Mu Qiao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Allison Kao
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA 02138, USA
| | - Yvonne Kölsch
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Ayush Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vincent P Kunze
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ashley M. Rasys
- Department of Cellular Biology, University of Georgia, Athens, GA 30602
| | - Rose Richardson
- Division of Neuroscience and Centre for Biological Timing, Faculty of Biology Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Robert J. Lucas
- Division of Neuroscience and Centre for Biological Timing, Faculty of Biology Medicine & Health, University of Manchester, Upper Brook Street, Manchester M13 9PT, UK
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Markus Meister
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Joshua T. Trachtenberg
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Wenjun Yan
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA 02138, USA
| | - Yi-Rong Peng
- Department of Ophthalmology, Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA 90095 United States
| | - Joshua R. Sanes
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA 02138, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, Vision Science Graduate Group, Center for Computational Biology, Biophysics Graduate Group, California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley CA 94720, USA
- Faculty Scientist, Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
16
|
Georges A, Lavergne A, Mandai M, Lepiemme F, Karim L, Demeulenaere L, Aguilar D, Schyns M, Nguyen L, Rakic JM, Takahashi M, Georges M, Takeda H. Comparing the transcriptome of developing native and iPSC-derived mouse retinae by single cell RNA sequencing. Sci Rep 2023; 13:1223. [PMID: 36681719 PMCID: PMC9867755 DOI: 10.1038/s41598-023-28429-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
We report the generation and analysis of single-cell RNA-Seq data (> 38,000 cells) from mouse native retinae and induced pluripotent stem cell (iPSC)-derived retinal organoids at four matched stages of development spanning the emergence of the major retinal cell types. We combine information from temporal sampling, visualization of 3D UMAP manifolds, pseudo-time and RNA velocity analyses, to show that iPSC-derived 3D retinal organoids broadly recapitulate the native developmental trajectories. However, we observe relaxation of spatial and temporal transcriptome control, premature emergence and dominance of photoreceptor precursor cells, and susceptibility of dynamically regulated pathways and transcription factors to culture conditions in retinal organoids. We demonstrate that genes causing human retinopathies are enriched in cell-type specifying genes and identify a subset of disease-causing genes with expression profiles that are highly conserved between human retinae and murine retinal organoids. This study provides a resource to the community that will be useful to assess and further improve protocols for ex vivo recapitulation and study of retinal development.
Collapse
Affiliation(s)
- Anouk Georges
- GIGA Stem Cells, GIGA Institute, University of Liège, Liège, Belgium
- Department of Ophthalmology, Faculty of Medicine and CHU University Hospital, University of Liège, Liège, Belgium
| | - Arnaud Lavergne
- GIGA Bioinformatics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan
| | - Fanny Lepiemme
- GIGA Stem Cells, GIGA Institute, University of Liège, Liège, Belgium
| | - Latifa Karim
- GIGA Genomics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Loic Demeulenaere
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Diego Aguilar
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Michael Schyns
- Digital Business, HEC Management School, University of Liège, Liège, Belgium
| | - Laurent Nguyen
- GIGA Stem Cells, GIGA Institute, University of Liège, Liège, Belgium
| | - Jean-Marie Rakic
- Department of Ophthalmology, Faculty of Medicine and CHU University Hospital, University of Liège, Liège, Belgium
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe, Japan
| | - Michel Georges
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium.
| | - Haruko Takeda
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| |
Collapse
|
17
|
Whitney IE, Butrus S, Dyer MA, Rieke F, Sanes JR, Shekhar K. Vision-Dependent and -Independent Molecular Maturation of Mouse Retinal Ganglion Cells. Neuroscience 2023; 508:153-173. [PMID: 35870562 PMCID: PMC10809145 DOI: 10.1016/j.neuroscience.2022.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
The development and connectivity of retinal ganglion cells (RGCs), the retina's sole output neurons, are patterned by activity-independent transcriptional programs and activity-dependent remodeling. To inventory the molecular correlates of these influences, we applied high-throughput single-cell RNA sequencing (scRNA-seq) to mouse RGCs at six embryonic and postnatal ages. We identified temporally regulated modules of genes that correlate with, and likely regulate, multiple phases of RGC development, ranging from differentiation and axon guidance to synaptic recognition and refinement. Some of these genes are expressed broadly while others, including key transcription factors and recognition molecules, are selectively expressed by one or a few of the 45 transcriptomically distinct types defined previously in adult mice. Next, we used these results as a foundation to analyze the transcriptomes of RGCs in mice lacking visual experience due to dark rearing from birth or to mutations that ablate either bipolar or photoreceptor cells. 98.5% of visually deprived (VD) RGCs could be unequivocally assigned to a single RGC type based on their transcriptional profiles, demonstrating that visual activity is dispensable for acquisition and maintenance of RGC type identity. However, visual deprivation significantly reduced the transcriptomic distinctions among RGC types, implying that activity is required for complete RGC maturation or maintenance. Consistent with this notion, transcriptomic alternations in VD RGCs significantly overlapped with gene modules found in developing RGCs. Our results provide a resource for mechanistic analyses of RGC differentiation and maturation, and for investigating the role of activity in these processes.
Collapse
Affiliation(s)
- Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Karthik Shekhar
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Helen Wills Neuroscience Institute, California Institute for Quantitative Biosciences, QB3, Center for Computational Biology, University of California, Berkeley, CA 94720, USA; Biological Systems Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
18
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
19
|
Al-Khindi T, Sherman MB, Kodama T, Gopal P, Pan Z, Kiraly JK, Zhang H, Goff LA, du Lac S, Kolodkin AL. The transcription factor Tbx5 regulates direction-selective retinal ganglion cell development and image stabilization. Curr Biol 2022; 32:4286-4298.e5. [PMID: 35998637 PMCID: PMC9560999 DOI: 10.1016/j.cub.2022.07.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/05/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022]
Abstract
The diversity of visual input processed by the mammalian visual system requires the generation of many distinct retinal ganglion cell (RGC) types, each tuned to a particular feature. The molecular code needed to generate this cell-type diversity is poorly understood. Here, we focus on the molecules needed to specify one type of retinal cell: the upward-preferring ON direction-selective ganglion cell (up-oDSGC) of the mouse visual system. Single-cell transcriptomic profiling of up- and down-oDSGCs shows that the transcription factor Tbx5 is selectively expressed in up-oDSGCs. The loss of Tbx5 in up-oDSGCs results in a selective defect in the formation of up-oDSGCs and a corresponding inability to detect vertical motion. A downstream effector of Tbx5, Sfrp1, is also critical for vertical motion detection but not up-oDSGC formation. These results advance our understanding of the molecular mechanisms that specify a rare retinal cell type and show how disrupting this specification leads to a corresponding defect in neural circuitry and behavior.
Collapse
Affiliation(s)
- Timour Al-Khindi
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael B Sherman
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Kodama
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Otolaryngology & Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Preethi Gopal
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhiwei Pan
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James K Kiraly
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hao Zhang
- Department of Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Loyal A Goff
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sascha du Lac
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Otolaryngology & Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex L Kolodkin
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
20
|
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: 20] [Impact Index Per Article: 6.7] [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
|
21
|
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: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons that function synchronously to convey visual information through the optic nerve to retinorecipient target areas in the brain. Injury or disease to the optic nerve results in RGC degeneration and loss of visual function, as few RGCs survive, and even fewer can be provoked to regenerate their axons. Despite causative insults being broadly shared, regeneration studies demonstrate that RGC types exhibit differential resilience to injury and undergo selective survival and regeneration of their axons. While most early studies have identified these RGC types based their morphological and physiological characteristics, recent advances in transgenic and gene sequencing technologies have further enabled type identification based on unique molecular features. In this review, we provide an overview of the well characterized RGC types and identify those shown to preferentially survive and regenerate in various regeneration models. Furthermore, we discuss cellular characteristics of both the resilient and susceptible RGC types including the combinatorial expression of different molecular markers that identify these specific populations. Lastly, we discuss potential molecular mechanisms and genes found to be selectively expressed by specific types that may contribute to their reparative capacity. Together, we describe the studies that lay the important groundwork for identifying factors that promote neural regeneration and help advance the development of targeted therapy for the treatment of RGC degeneration as well as neurodegenerative diseases in general.
Collapse
Affiliation(s)
- Mary L Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gabriel Nascimento-Dos-Santos
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
22
|
Goetz J, Jessen ZF, Jacobi A, Mani A, Cooler S, Greer D, Kadri S, Segal J, Shekhar K, Sanes JR, Schwartz GW. Unified classification of mouse retinal ganglion cells using function, morphology, and gene expression. Cell Rep 2022; 40:111040. [PMID: 35830791 PMCID: PMC9364428 DOI: 10.1016/j.celrep.2022.111040] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/27/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Classification and characterization of neuronal types are critical for understanding their function and dysfunction. Neuronal classification schemes typically rely on measurements of electrophysiological, morphological, and molecular features, but aligning such datasets has been challenging. Here, we present a unified classification of mouse retinal ganglion cells (RGCs), the sole retinal output neurons. We use visually evoked responses to classify 1,859 mouse RGCs into 42 types. We also obtain morphological or transcriptomic data from subsets and use these measurements to align the functional classification to publicly available morphological and transcriptomic datasets. We create an online database that allows users to browse or download the data and to classify RGCs from their light responses using a machine learning algorithm. This work provides a resource for studies of RGCs, their upstream circuits in the retina, and their projections in the brain, and establishes a framework for future efforts in neuronal classification and open data distribution.
Collapse
Affiliation(s)
- Jillian Goetz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zachary F Jessen
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA; Medical Scientist Training Program, Northwestern University, Chicago, IL, USA
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Adam Mani
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sam Cooler
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Devon Greer
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Sabah Kadri
- Department of Pathology, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Jeremy Segal
- Department of Pathology, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Gregory W Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
23
|
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: 37] [Impact Index Per Article: 12.3] [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
|
24
|
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: 5.3] [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
|
25
|
Goel M, Aponte AM, Wistow G, Badea TC. Molecular studies into cell biological role of Copine-4 in Retinal Ganglion Cells. PLoS One 2021; 16:e0255860. [PMID: 34847148 PMCID: PMC8631636 DOI: 10.1371/journal.pone.0255860] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/12/2021] [Indexed: 11/19/2022] Open
Abstract
The molecular mechanisms underlying morphological diversity in retinal cell types are poorly understood. We have previously reported that several members of the Copine family of Ca-dependent membrane adaptors are expressed in Retinal Ganglion Cells and transcriptionally regulated by Brn3 transcription factors. Several Copines are enriched in the retina and their over-expression leads to morphological changes -formation of elongated processes-, reminiscent of neurites, in HEK293 cells. However, the role of Copines in the retina is largely unknown. We now investigate Cpne4, a Copine whose expression is restricted to Retinal Ganglion Cells. Over-expression of Cpne4 in RGCs in vivo led to formation of large varicosities on the dendrites but did not otherwise visibly affect dendrite or axon formation. Protein interactions studies using yeast two hybrid analysis from whole retina cDNA revealed two Cpne4 interacting proteins-Host Cell Factor 1 and Morn2. Mass Spectrometry analysis of retina lysate pulled down using Cpne4 or its vonWillebrand A domain showed 207 interacting proteins. A Gene Ontology analysis of the discovered proteins suggests that Cpne4 is involved in several metabolic and signaling pathways in the retina.
Collapse
Affiliation(s)
- Manvi Goel
- Retinal Circuit Development & Genetics Unit, Neurobiology Neurodegeneration & Repair Laboratory, NEI, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Angel M. Aponte
- Proteomics Core, NHLBI, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, NEI, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tudor C. Badea
- Retinal Circuit Development & Genetics Unit, Neurobiology Neurodegeneration & Repair Laboratory, NEI, National Institutes of Health, Bethesda, Maryland, United States of America
- Faculty of Medicine, Research and Development Institute, Transilvania University of Brasov, Brasov, Romania
| |
Collapse
|
26
|
Rochon PL, Theriault C, Rangel Olguin AG, Krishnaswamy A. The cell adhesion molecule Sdk1 shapes assembly of a retinal circuit that detects localized edges. eLife 2021; 10:e70870. [PMID: 34545809 PMCID: PMC8514235 DOI: 10.7554/elife.70870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/11/2021] [Indexed: 01/10/2023] Open
Abstract
Nearly 50 different mouse retinal ganglion cell (RGC) types sample the visual scene for distinct features. RGC feature selectivity arises from their synapses with a specific subset of amacrine (AC) and bipolar cell (BC) types, but how RGC dendrites arborize and collect input from these specific subsets remains poorly understood. Here we examine the hypothesis that RGCs employ molecular recognition systems to meet this challenge. By combining calcium imaging and type-specific histological stains, we define a family of circuits that express the recognition molecule Sidekick-1 (Sdk1), which include a novel RGC type (S1-RGC) that responds to local edges. Genetic and physiological studies revealed that Sdk1 loss selectively disrupts S1-RGC visual responses, which result from a loss of excitatory and inhibitory inputs and selective dendritic deficits on this neuron. We conclude that Sdk1 shapes dendrite growth and wiring to help S1-RGCs become feature selective.
Collapse
|
27
|
Zhang R, He H, Yuan B, Wu Z, Wang X, Du Y, Chen Y, Qiu Z. An Intronic Variant of CHD7 Identified in Autism Patients Interferes with Neuronal Differentiation and Development. Neurosci Bull 2021; 37:1091-1106. [PMID: 33948885 PMCID: PMC8353028 DOI: 10.1007/s12264-021-00685-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/27/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic composition plays critical roles in the pathogenesis of autism spectrum disorder (ASD). Especially, inherited and de novo intronic variants are often seen in patients with ASD. However, the biological significance of intronic variants is difficult to address. Here, among a Chinese ASD cohort, we identified a recurrent inherited intronic variant in the CHD7 gene, which is specifically enriched in East Asian populations. CHD7 has been implicated in numerous developmental disorders including CHARGE syndrome and ASD. To investigate whether the ASD-associated CHD7 intronic variant affects neural development, we established human embryonic stem cells carrying this variant using CRISPR/Cas9 methods and found that the level of CHD7 mRNA significantly decreased compared to control. Upon differentiation towards the forebrain neuronal lineage, we found that neural cells carrying the CHD7 intronic variant exhibited developmental delay and maturity defects. Importantly, we found that TBR1, a gene also implicated in ASD, was significantly increased in neurons carrying the CHD7 intronic variant, suggesting the intrinsic relevance among ASD genes. Furthermore, the morphological defects found in neurons carrying CHD7 intronic mutations were rescued by knocking down TBR1, indicating that TBR1 may be responsible for the defects in CHD7-related disorders. Finally, the CHD7 intronic variant generated three abnormal forms of transcripts through alternative splicing, which all exhibited loss-of-function in functional assays. Our study provides crucial evidence supporting the notion that the intronic variant of CHD7 is potentially an autism susceptibility site, shedding new light on identifying the functions of intronic variants in genetic studies of autism.
Collapse
Affiliation(s)
- Ran Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Ziyan Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiuzhen Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yasong Du
- Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yuejun Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zilong Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
28
|
Chen CK, Kiyama T, Weber N, Whitaker CM, Pan P, Badea TC, Massey SC, Mao CA. Characterization of Tbr2-expressing retinal ganglion cells. J Comp Neurol 2021; 529:3513-3532. [PMID: 34245014 DOI: 10.1002/cne.25208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/27/2021] [Accepted: 06/28/2021] [Indexed: 12/19/2022]
Abstract
The mammalian retina contains more than 40 retinal ganglion cell (RGC) subtypes based on their unique morphologies, functions, and molecular profiles. Among them, intrinsically photosensitive RGCs (ipRGCs) are the first specified RGC type emerging from a common retinal progenitor pool during development. Previous work has shown that T-box transcription factor T-brain 2 (Tbr2) is essential for the formation and maintenance of ipRGCs, and that Tbr2-expressing RGCs activate Opn4 expression upon native ipRGC ablation, suggesting that Tbr2+ RGCs contain a reservoir for ipRGCs. However, the identity of Tbr2+ RGCs has not been fully vetted. Here, using genetic sparse labeling and single cell recording, we showed that Tbr2-expressing retinal neurons include RGCs and a subset of GABAergic displaced amacrine cells (dACs). Most Tbr2+ RGCs are intrinsically photosensitive and morphologically resemble native ipRGCs with identical retinofugal projections. Tbr2+ RGCs also include a unique and rare Pou4f1-expressing OFF RGC subtype. Using a loss-of-function strategy, we have further demonstrated that Tbr2 is essential for the survival of these RGCs and dACs, as well as maintaining the expression of Opn4. These data set a strong foundation to study how Tbr2 regulates ipRGC development and survival, as well as the expression of molecular machinery regulating intrinsic photosensitivity.
Collapse
Affiliation(s)
- Ching-Kang Chen
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Nicole Weber
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Christopher M Whitaker
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Ping Pan
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Tudor C Badea
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA.,Research and Development Institute, Transilvania University of Brasov, School of Medicine, Brasov, Romania
| | - Stephen C Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA.,The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA.,The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
29
|
Abstract
It has been known for over a century that the basic organization of the retina is conserved across vertebrates. It has been equally clear that retinal cells can be classified into numerous types, but only recently have methods been devised to explore this diversity in unbiased, scalable, and comprehensive ways. Advances in high-throughput single-cell RNA-sequencing (scRNA-seq) have played a pivotal role in this effort. In this article, we outline the experimental and computational components of scRNA-seq and review studies that have used them to generate retinal atlases of cell types in several vertebrate species. These atlases have enabled studies of retinal development, responses of retinal cells to injury, expression patterns of genes implicated in retinal disease, and the evolution of cell types. Recently, the inquiry has expanded to include the entire eye and visual centers in the brain. These studies have enhanced our understanding of retinal function and dysfunction and provided tools and insights for exploring neural diversity throughout the brain. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Karthik Shekhar
- Department of Chemical and Biomolecular Engineering; Helen Wills Neuroscience Institute; and California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, California 94720, USA;
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cell Biology, Harvard University, Cambridge, Massachusetts 02138, USA;
| |
Collapse
|
30
|
Retinal Ganglion Cell Transplantation: Approaches for Overcoming Challenges to Functional Integration. Cells 2021; 10:cells10061426. [PMID: 34200991 PMCID: PMC8228580 DOI: 10.3390/cells10061426] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
As part of the central nervous system, mammalian retinal ganglion cells (RGCs) lack significant regenerative capacity. Glaucoma causes progressive and irreversible vision loss by damaging RGCs and their axons, which compose the optic nerve. To functionally restore vision, lost RGCs must be replaced. Despite tremendous advancements in experimental models of optic neuropathy that have elucidated pathways to induce endogenous RGC neuroprotection and axon regeneration, obstacles to achieving functional visual recovery through exogenous RGC transplantation remain. Key challenges include poor graft survival, low donor neuron localization to the host retina, and inadequate dendritogenesis and synaptogenesis with afferent amacrine and bipolar cells. In this review, we summarize the current state of experimental RGC transplantation, and we propose a set of standard approaches to quantifying and reporting experimental outcomes in order to guide a collective effort to advance the field toward functional RGC replacement and optic nerve regeneration.
Collapse
|
31
|
Transcription factor encoding of neuron subtype: Strategies that specify arbor pattern. Curr Opin Neurobiol 2021; 69:149-158. [PMID: 33895620 DOI: 10.1016/j.conb.2021.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023]
Abstract
Dendrite and axon arbors form scaffolds that connect a neuron to its partners; they are patterned to support the specific connectivity and computational requirements of each neuron subtype. Transcription factor networks control the specification of neuron subtypes, and the consequent diversification of their stereotyped arbor patterns during differentiation. We outline how the differentiation trajectories of stereotyped arbors are shaped by hierarchical deployment of precursor cell and postmitotic transcription factors. These transcription factors exert modular control over the dendrite and axon features of a single neuron, create spatial and functional compartmentalization of an arbor, instruct implementation of developmental patterning rules, and exert operational control over the cell biological processes that construct an arbor.
Collapse
|
32
|
Lyu J, Mu X. Genetic control of retinal ganglion cell genesis. Cell Mol Life Sci 2021; 78:4417-4433. [PMID: 33782712 DOI: 10.1007/s00018-021-03814-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/27/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Retinal ganglion cells (RGCs) are the only projection neurons in the neural retina. They receive and integrate visual signals from upstream retinal neurons in the visual circuitry and transmit them to the brain. The function of RGCs is performed by the approximately 40 RGC types projecting to various central brain targets. RGCs are the first cell type to form during retinogenesis. The specification and differentiation of the RGC lineage is a stepwise process; a hierarchical gene regulatory network controlling the RGC lineage has been identified and continues to be elaborated. Recent studies with single-cell transcriptomics have led to unprecedented new insights into their types and developmental trajectory. In this review, we summarize our current understanding of the functions and relationships of the many regulators of the specification and differentiation of the RGC lineage. We emphasize the roles of these key transcription factors and pathways in different developmental steps, including the transition from retinal progenitor cells (RPCs) to RGCs, RGC differentiation, generation of diverse RGC types, and central projection of the RGC axons. We discuss critical issues that remain to be addressed for a comprehensive understanding of these different aspects of RGC genesis and emerging technologies, including single-cell techniques, novel genetic tools and resources, and high-throughput genome editing and screening assays, which can be leveraged in future studies.
Collapse
Affiliation(s)
- Jianyi Lyu
- Department of Ophthalmology/Ross Eye Institute, State University of New York At Buffalo, Buffalo, NY, 14203, USA
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, State University of New York At Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
33
|
Abstract
Neurons develop dendritic morphologies that bear cell type-specific features in dendritic field size and geometry, branch placement and density, and the types and distributions of synaptic contacts. Dendritic patterns influence the types and numbers of inputs a neuron receives, and the ways in which neural information is processed and transmitted in the circuitry. Even subtle alterations in dendritic structures can have profound consequences on neuronal function and are implicated in neurodevelopmental disorders. In this chapter, I review how growing dendrites acquire their exquisite patterns by drawing examples from diverse neuronal cell types in vertebrate and invertebrate model systems. Dendrite morphogenesis is shaped by intrinsic and extrinsic factors such as transcriptional regulators, guidance and adhesion molecules, neighboring cells and synaptic partners. I discuss molecular mechanisms that regulate dendrite morphogenesis with a focus on five aspects of dendrite patterning: (1) Dendritic cytoskeleton and cellular machineries that build the arbor; (2) Gene regulatory mechanisms; (3) Afferent cues that regulate dendritic arbor growth; (4) Space-filling strategies that optimize dendritic coverage; and (5) Molecular cues that specify dendrite wiring. Cell type-specific implementation of these patterning mechanisms produces the diversity of dendrite morphologies that wire the nervous system.
Collapse
|
34
|
Kölsch Y, Hahn J, Sappington A, Stemmer M, Fernandes AM, Helmbrecht TO, Lele S, Butrus S, Laurell E, Arnold-Ammer I, Shekhar K, Sanes JR, Baier H. Molecular classification of zebrafish retinal ganglion cells links genes to cell types to behavior. Neuron 2021; 109:645-662.e9. [PMID: 33357413 PMCID: PMC7897282 DOI: 10.1016/j.neuron.2020.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/09/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
Retinal ganglion cells (RGCs) form an array of feature detectors, which convey visual information to central brain regions. Characterizing RGC diversity is required to understand the logic of the underlying functional segregation. Using single-cell transcriptomics, we systematically classified RGCs in adult and larval zebrafish, thereby identifying marker genes for >30 mature types and several developmental intermediates. We used this dataset to engineer transgenic driver lines, enabling specific experimental access to a subset of RGC types. Expression of one or few transcription factors often predicts dendrite morphologies and axonal projections to specific tectal layers and extratectal targets. In vivo calcium imaging revealed that molecularly defined RGCs exhibit specific functional tuning. Finally, chemogenetic ablation of eomesa+ RGCs, which comprise melanopsin-expressing types with projections to a small subset of central targets, selectively impaired phototaxis. Together, our study establishes a framework for systematically studying the functional architecture of the visual system.
Collapse
Affiliation(s)
- Yvonne Kölsch
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany; Graduate School of Systemic Neurosciences, Ludwig Maximilian University, 82152 Martinsried, Germany
| | - Joshua Hahn
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94720, USA
| | - Anna Sappington
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Manuel Stemmer
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - António M Fernandes
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - Thomas O Helmbrecht
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - Shriya Lele
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94720, USA
| | - Eva Laurell
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - Irene Arnold-Ammer
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, California Institute for Quantitative Biosciences, QB3, Center for Computational Biology, UC Berkeley, Berkeley, CA 94720, USA.
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cell Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Herwig Baier
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, 82152 Martinsried, Germany.
| |
Collapse
|
35
|
Role of the Internal Limiting Membrane in Structural Engraftment and Topographic Spacing of Transplanted Human Stem Cell-Derived Retinal Ganglion Cells. Stem Cell Reports 2020; 16:149-167. [PMID: 33382979 PMCID: PMC7897583 DOI: 10.1016/j.stemcr.2020.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Retinal ganglion cell (RGC) replacement holds potential for restoring vision lost to optic neuropathy. Transplanted RGCs must undergo neuroretinal integration to receive afferent visual signals for processing and efferent transmission. To date, retinal integration following RGC transplantation has been limited. We sought to overcome key barriers to transplanted human stem cell-derived RGC integration. Following co-culture ex vivo on organotypic mouse retinal explants, human RGCs cluster and extend bundled neurites that remain superficial to the neuroretina, hindering afferent synaptogenesis. To enhance integration, we increased the cellular permeability of the internal limiting membrane (ILM). Extracellular matrix digestion using proteolytic enzymes achieved ILM disruption while minimizing retinal toxicity and preserving glial reactivity. ILM disruption is associated with dispersion rather than clustering of co-cultured RGC bodies and neurites, and increased parenchymal neurite ingrowth. The ILM represents a significant obstacle to transplanted RGC connectivity and its circumvention may be necessary for functional RGC replacement.
Collapse
|
36
|
Kerstein PC, Leffler J, Sivyer B, Taylor WR, Wright KM. Gbx2 Identifies Two Amacrine Cell Subtypes with Distinct Molecular, Morphological, and Physiological Properties. Cell Rep 2020; 33:108382. [PMID: 33207201 PMCID: PMC7713908 DOI: 10.1016/j.celrep.2020.108382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/21/2020] [Accepted: 10/22/2020] [Indexed: 01/21/2023] Open
Abstract
Our understanding of nervous system function is limited by our ability to identify and manipulate neuronal subtypes within intact circuits. We show that the Gbx2CreERT2-IRES-EGFP mouse line labels two amacrine cell (AC) subtypes in the mouse retina that have distinct morphological, physiological, and molecular properties. Using a combination of RNA-seq, genetic labeling, and patch clamp recordings, we show that one subtype is GABAergic that receives excitatory input from On bipolar cells. The other population is a non-GABAergic, non-glycinergic (nGnG) AC subtype that lacks the expression of standard neurotransmitter markers. Gbx2+ nGnG ACs have smaller, asymmetric dendritic arbors that receive excitatory input from both On and Off bipolar cells. Gbx2+ nGnG ACs also exhibit spatially restricted tracer coupling to bipolar cells (BCs) through gap junctions. This study identifies a genetic tool for investigating the two distinct AC subtypes, and it provides a model for studying synaptic communication and visual circuit function. Investigations into neural circuit development and function are limited by the lack of genetic tools to label and perturb individual neuronal subtypes. Using the Gbx2CreERT2 mouse line, Kerstein et al. identify two amacrine cell subtypes in the mouse retina and explore their distinct molecular, morphological, and physiological characteristics.
Collapse
Affiliation(s)
- Patrick C Kerstein
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joseph Leffler
- School of Optometry and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Neuroscience Graduate Program, Oregon Health and Science University, Portland, OR 97239, USA
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
| | - W Rowland Taylor
- School of Optometry and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
37
|
Kurmangaliyev YZ, Yoo J, Valdes-Aleman J, Sanfilippo P, Zipursky SL. Transcriptional Programs of Circuit Assembly in the Drosophila Visual System. Neuron 2020; 108:1045-1057.e6. [PMID: 33125872 DOI: 10.1016/j.neuron.2020.10.006] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/31/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022]
Abstract
Precise patterns of synaptic connections between neurons are encoded in their genetic programs. Here, we use single-cell RNA sequencing to profile neuronal transcriptomes at multiple stages in the developing Drosophila visual system. We devise an efficient strategy for profiling neurons at multiple time points in a single pool, thereby minimizing batch effects and maximizing the reliability of time-course data. A transcriptional atlas spanning multiple stages is generated, including more than 150 distinct neuronal populations; of these, 88 are followed through synaptogenesis. This analysis reveals a common (pan-neuronal) program unfolding in highly coordinated fashion in all neurons, including genes encoding proteins comprising the core synaptic machinery and membrane excitability. This program is overlaid by cell-type-specific programs with diverse cell recognition molecules expressed in different combinations and at different times. We propose that a pan-neuronal program endows neurons with the competence to form synapses and that cell-type-specific programs control synaptic specificity.
Collapse
Affiliation(s)
- Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Juyoun Yoo
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Javier Valdes-Aleman
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Piero Sanfilippo
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - S Lawrence Zipursky
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
38
|
Monti G, Jensen ML, Mehmedbasic A, Jørgensen MM, Holm IE, Barkholt P, Zole E, Vægter CB, Vorum H, Nyengaard JR, Andersen OM. SORLA Expression in Synaptic Plexiform Layers of Mouse Retina. Mol Neurobiol 2020; 57:3106-3117. [PMID: 32472518 DOI: 10.1007/s12035-020-01946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/13/2020] [Indexed: 11/25/2022]
Abstract
Sorting protein-related receptor containing LDLR class A repeats (SORLA; also known as LR11) exerts intraneuronal trafficking functions in the central nervous system. Recently, involvement of SORLA in retinogenesis was proposed, but no studies have examined yet in detail the expression pattern of this sorting receptor in the retina. Here, we provide a spatio-temporal characterization of SORL1 mRNA and its translational product SORLA in the postnatal mouse retina. Using stereological analysis, we confirmed previous studies showing that receptor depletion in knockout mice significantly reduces the number of cells in the inner nuclear layer (INL), suggesting that functional SORLA expression is essential for the development of this retinal strata. qPCR and Western blot analyses showed that SORL1/SORLA expression peaks at postnatal day 15, just after eye opening. Interestingly, we found that transcripts are somatically located in several neuronal populations residing in the INL and the ganglion cell layer, whereas SORLA protein is also present in the synaptic plexiform layers. In line with receptor expression in dendritic terminals, we found delayed stratification of the inner plexiform layer in knockout mice, indicating an involvement of SORLA in neuronal connectivity. Altogether, these data suggest a novel role of SORLA in synaptogenesis. Receptor dysfunctions may be implicated in morphological and functional impairments of retinal inner layer formation associated with eye disorders.
Collapse
Affiliation(s)
- Giulia Monti
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Marianne L Jensen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Arnela Mehmedbasic
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Margarita Melnikova Jørgensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Ida E Holm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Pernille Barkholt
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Egija Zole
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Christian B Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Henrik Vorum
- Department of Ophthalmology, Aalborg University Hospital, Hobrovej 18-22, DK-9000, Aalborg, Denmark
| | - Jens R Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
39
|
Graham HK, Duan X. Molecular mechanisms regulating synaptic specificity and retinal circuit formation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e379. [PMID: 32267095 DOI: 10.1002/wdev.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/28/2022]
Abstract
The central nervous system (CNS) is composed of precisely assembled circuits which support a variety of physiological functions and behaviors. These circuits include multiple subtypes of neurons with unique morphologies, electrical properties, and molecular identities. How these component parts are precisely wired-up has been a topic of great interest to the field of developmental neurobiology and has implications for our understanding of the etiology of many neurological disorders and mental illnesses. To date, many molecules involved in synaptic choice and specificity have been identified, including members of several families of cell-adhesion molecules (CAMs), which are cell-surface molecules that mediate cell-cell contacts and subsequent intracellular signaling. One favored hypothesis is that unique expression patterns of CAMs define specific neuronal subtype populations and determine compatible pre- and postsynaptic neuronal partners based on the expression of these unique CAMs. The mouse retina has served as a beautiful model for investigations into mammalian CAM interactions due to its well-defined neuronal subtypes and distinct circuits. Moreover, the retina is readily amenable to visualization of circuit organization and electrophysiological measurement of circuit function. The advent of recent genetic, genomic, and imaging technologies has opened the field up to large-scale, unbiased approaches for identification of new molecular determinants of synaptic specificity. Thus, building on the foundation of work reviewed here, we can expect rapid expansion of the field, harnessing the mouse retina as a model to understand the molecular basis for synaptic specificity and functional circuit assembly. This article is categorized under: Nervous System Development > Vertebrates: General Principles Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Hannah K Graham
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA.,Neuroscience Graduate Program, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA.,Department of Physiology, University of California San Francisco, San Francisco, California, USA.,Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
40
|
Sanes JR, Zipursky SL. Synaptic Specificity, Recognition Molecules, and Assembly of Neural Circuits. Cell 2020; 181:536-556. [DOI: 10.1016/j.cell.2020.04.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 01/02/2023]
|
41
|
Tran NM, Shekhar K, Whitney IE, Jacobi A, Benhar I, Hong G, Yan W, Adiconis X, Arnold ME, Lee JM, Levin JZ, Lin D, Wang C, Lieber CM, Regev A, He Z, Sanes JR. Single-Cell Profiles of Retinal Ganglion Cells Differing in Resilience to Injury Reveal Neuroprotective Genes. Neuron 2019; 104:1039-1055.e12. [PMID: 31784286 PMCID: PMC6923571 DOI: 10.1016/j.neuron.2019.11.006] [Citation(s) in RCA: 387] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/25/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023]
Abstract
Neuronal types in the central nervous system differ dramatically in their resilience to injury or other insults. Here we studied the selective resilience of mouse retinal ganglion cells (RGCs) following optic nerve crush (ONC), which severs their axons and leads to death of ∼80% of RGCs within 2 weeks. To identify expression programs associated with differential resilience, we first used single-cell RNA-seq (scRNA-seq) to generate a comprehensive molecular atlas of 46 RGC types in adult retina. We then tracked their survival after ONC; characterized transcriptomic, physiological, and morphological changes that preceded degeneration; and identified genes selectively expressed by each type. Finally, using loss- and gain-of-function assays in vivo, we showed that manipulating some of these genes improved neuronal survival and axon regeneration following ONC. This study provides a systematic framework for parsing type-specific responses to injury and demonstrates that differential gene expression can be used to reveal molecular targets for intervention.
Collapse
Affiliation(s)
- Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Karthik Shekhar
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Inbal Benhar
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Guosong Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Department of Material Science and Engineering and Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Xian Adiconis
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - McKinzie E Arnold
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jung Min Lee
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua Z Levin
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Dingchang Lin
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Chen Wang
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles M Lieber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Aviv Regev
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA and Department of Biology and Koch Institute, MIT, Cambridge, MA 02139, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
42
|
Yook C, Kim K, Kim D, Kang H, Kim SG, Kim E, Kim SY. A TBR1-K228E Mutation Induces Tbr1 Upregulation, Altered Cortical Distribution of Interneurons, Increased Inhibitory Synaptic Transmission, and Autistic-Like Behavioral Deficits in Mice. Front Mol Neurosci 2019; 12:241. [PMID: 31680851 PMCID: PMC6797848 DOI: 10.3389/fnmol.2019.00241] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Mutations in Tbr1, a high-confidence ASD (autism spectrum disorder)-risk gene encoding the transcriptional regulator TBR1, have been shown to induce diverse ASD-related molecular, synaptic, neuronal, and behavioral dysfunctions in mice. However, whether Tbr1 mutations derived from autistic individuals cause similar dysfunctions in mice remains unclear. Here we generated and characterized mice carrying the TBR1-K228E de novo mutation identified in human ASD and identified various ASD-related phenotypes. In heterozygous mice carrying this mutation (Tbr1+/K228E mice), levels of the TBR1-K228E protein, which is unable to bind target DNA, were strongly increased. RNA-Seq analysis of the Tbr1+/K228E embryonic brain indicated significant changes in the expression of genes associated with neurons, astrocytes, ribosomes, neuronal synapses, and ASD risk. The Tbr1+/K228E neocortex also displayed an abnormal distribution of parvalbumin-positive interneurons, with a lower density in superficial layers but a higher density in deep layers. These changes were associated with an increase in inhibitory synaptic transmission in layer 6 pyramidal neurons that was resistant to compensation by network activity. Behaviorally, Tbr1+/K228E mice showed decreased social interaction, increased self-grooming, and modestly increased anxiety-like behaviors. These results suggest that the human heterozygous TBR1-K228E mutation induces ASD-related transcriptomic, protein, neuronal, synaptic, and behavioral dysfunctions in mice.
Collapse
Affiliation(s)
- Chaehyun Yook
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Sun-Gyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Soo Young Kim
- College of Pharmacy, Yeongnam University, Gyeongsan, South Korea
| |
Collapse
|
43
|
Kiyama T, Long Y, Chen CK, Whitaker CM, Shay A, Wu H, Badea TC, Mohsenin A, Parker-Thornburg J, Klein WH, Mills SL, Massey SC, Mao CA. Essential Roles of Tbr1 in the Formation and Maintenance of the Orientation-Selective J-RGCs and a Group of OFF-Sustained RGCs in Mouse. Cell Rep 2019; 27:900-915.e5. [PMID: 30995485 PMCID: PMC6542366 DOI: 10.1016/j.celrep.2019.03.077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 02/10/2019] [Accepted: 03/20/2019] [Indexed: 01/27/2023] Open
Abstract
In the mouse retina, more than 30 retinal ganglion cell (RGC) subtypes have been classified based on a combined metric of morphological and functional characteristics. RGCs arise from a common pool of retinal progenitor cells during embryonic stages and differentiate into mature subtypes in adult retinas. However, the cellular and molecular mechanisms controlling formation and maturation of such remarkable cellular diversity remain unknown. Here, we demonstrate that T-box transcription factor T-brain 1 (Tbr1) is expressed in two groups of morphologically and functionally distinct RGCs: the orientation-selective J-RGCs and a group of OFF-sustained RGCs with symmetrical dendritic arbors. When Tbr1 is genetically ablated during retinal development, these two RGC groups cannot develop. Ectopically expressing Tbr1 in M4 ipRGCs during development alters dendritic branching and density but not the inner plexiform layer stratification level. Our data indicate that Tbr1 plays critical roles in regulating the formation and dendritic morphogenesis of specific RGC types.
Collapse
Affiliation(s)
- Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ye Long
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ching-Kang Chen
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher M Whitaker
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Allison Shay
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongyu Wu
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Tudor C Badea
- National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Amir Mohsenin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Robert Cizik Eye Clinic, Houston, TX 77030, USA
| | - Jan Parker-Thornburg
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William H Klein
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen L Mills
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Stephen C Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA.
| |
Collapse
|
44
|
Goel M, Li T, Badea TC. Differential expression and subcellular localization of Copines in mouse retina. J Comp Neurol 2019; 527:2245-2262. [PMID: 30866042 DOI: 10.1002/cne.24684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/04/2019] [Accepted: 03/11/2019] [Indexed: 01/06/2023]
Abstract
Combinatorial expression of Brn3 transcription factors is required for the development of cell-specific morphologies in retinal ganglion cells (RGCs). The molecular mechanisms by which Brn3s regulate RGC type specific features are largely unexplored. We previously identified several members of the Copine (Cpne) family of molecules as potential targets of Brn3 transcription factors in the retina. We now use in situ hybridization and immunohistochemistry to characterize Copine expression in the postnatal and adult mouse retina. We find that Cpne5, 6, and 9 are expressed in the ganglion cell layer (GCL) and inner nuclear layer (INL) in both amacrine cells and RGCs. Cpne4 expression is restricted to one amacrine cell population of the INL, but is specifically expressed in RGCs in the GCL. Cpne4 expression in RGCs is regulated by Brn3b both cell autonomously (in Brn3b+ RGCs) and cell nonautonomously (in Brn3b- RGCs). Copines exhibit a variety of subcellular distributions when overexpressed in tissue culture cells (HEK293), and can induce the formation of elongated processes reminiscent of neurites in these non-neuronal cells. Our results suggest that Copines might be involved in a combinatorial fashion in Brn3b-dependent specification of RGC types. Given their expression profile and previously proven role as Ca2+ sensors, they may participate in the morphogenetic processes that shape RGC dendrite and axon formation at early postnatal ages.
Collapse
Affiliation(s)
- Manvi Goel
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| | - Tiansen Li
- Retinal Cell Biology & Degeneration Section, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| | - Tudor C Badea
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, Maryland
| |
Collapse
|
45
|
Molecular Classification and Comparative Taxonomics of Foveal and Peripheral Cells in Primate Retina. Cell 2019; 176:1222-1237.e22. [PMID: 30712875 DOI: 10.1016/j.cell.2019.01.004] [Citation(s) in RCA: 313] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/08/2018] [Accepted: 12/31/2018] [Indexed: 01/03/2023]
Abstract
High-acuity vision in primates, including humans, is mediated by a small central retinal region called the fovea. As more accessible organisms lack a fovea, its specialized function and its dysfunction in ocular diseases remain poorly understood. We used 165,000 single-cell RNA-seq profiles to generate comprehensive cellular taxonomies of macaque fovea and peripheral retina. More than 80% of >60 cell types match between the two regions but exhibit substantial differences in proportions and gene expression, some of which we relate to functional differences. Comparison of macaque retinal types with those of mice reveals that interneuron types are tightly conserved. In contrast, projection neuron types and programs diverge, despite exhibiting conserved transcription factor codes. Key macaque types are conserved in humans, allowing mapping of cell-type and region-specific expression of >190 genes associated with 7 human retinal diseases. Our work provides a framework for comparative single-cell analysis across tissue regions and species.
Collapse
|
46
|
Yamagata M, Sanes JR. Expression and Roles of the Immunoglobulin Superfamily Recognition Molecule Sidekick1 in Mouse Retina. Front Mol Neurosci 2019; 11:485. [PMID: 30687002 PMCID: PMC6333872 DOI: 10.3389/fnmol.2018.00485] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022] Open
Abstract
Processes of >100 types of interneurons (bipolar and amacrine cells) and projection neurons (retinal ganglion cells, RGCs) form specific and stereotyped patterns of connections in the inner plexiform layer (IPL) of the mouse retina. Four closely related homophilic immunoglobulin superfamily recognition molecules (Sidekick [Sdk] 1, Sdk 2, Dscam, and DscamL1) have been shown to play roles in patterning neuronal arbors and connections in chick retina, and all but Sdk1 have been shown to play related roles in mice. Here, we compare patterns of Sdk1 and Sdk2 expression in mouse retina and use genetic methods to assess roles of Sdk1. In adult retina, 3 neuronal types express sdk1 but not sdk2 at detectable levels, 5 express sdk2 but not sdk1 and 3 express both. Patterns of gene expression and protein localization at or near synapses are established during the first postnatal week. Dendrites of amacrine cells and RGCs that express sdk1 but not sdk2 arborize in the same narrow stratum in the center of the IPL. In the absence of Sdk1, this laminar restriction is degraded. Overexpression of sdk1 in developing cells that normally express sdk2 reorients their dendrites to resemble those of endogenously Sdk1-positive cells, indicating that Sdk1 plays an instructive role in patterning the IPL. Sdk1 fails to affect arbors when introduced after they are mature, suggesting that it is required to form but not maintain laminar restrictions. The effect of ectopically expressed sdk1 requires the presence of endogenous Sdk1, suggesting that the effect requires homophilic interactions among Sdk1-positive neurites. Together with previous results on Sdk2, Dscam, DscamL1, as well as the related Contactins, our results support the idea that an elaborate immunoglobulin superfamily code plays a prominent role in establishing neural circuits in the retina by means of tightly regulated cell type-specific expression and homophilically restricted intercellular interactions.
Collapse
Affiliation(s)
- Masahito Yamagata
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, United States
| |
Collapse
|
47
|
Fazel Darbandi S, Robinson Schwartz SE, Qi Q, Catta-Preta R, Pai ELL, Mandell JD, Everitt A, Rubin A, Krasnoff RA, Katzman S, Tastad D, Nord AS, Willsey AJ, Chen B, State MW, Sohal VS, Rubenstein JLR. Neonatal Tbr1 Dosage Controls Cortical Layer 6 Connectivity. Neuron 2018; 100:831-845.e7. [PMID: 30318412 PMCID: PMC6250594 DOI: 10.1016/j.neuron.2018.09.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 08/03/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Abstract
An understanding of how heterozygous loss-of-function mutations in autism spectrum disorder (ASD) risk genes, such as TBR1, contribute to ASD remains elusive. Conditional Tbr1 deletion during late mouse gestation in cortical layer 6 neurons (Tbr1layer6 mutants) provides novel insights into its function, including dendritic patterning, synaptogenesis, and cell-intrinsic physiology. These phenotypes occur in heterozygotes, providing insights into mechanisms that may underlie ASD pathophysiology. Restoring expression of Wnt7b largely rescues the synaptic deficit in Tbr1layer6 mutant neurons. Furthermore, Tbr1layer6 heterozygotes have increased anxiety-like behavior, a phenotype seen ASD. Integrating TBR1 chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) data from layer 6 neurons and activity of TBR1-bound candidate enhancers provides evidence for how TBR1 regulates layer 6 properties. Moreover, several putative TBR1 targets are ASD risk genes, placing TBR1 in a central position both for ASD risk and for regulating transcriptional circuits that control multiple steps in layer 6 development essential for the assembly of neural circuits.
Collapse
Affiliation(s)
- Siavash Fazel Darbandi
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sarah E Robinson Schwartz
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Qihao Qi
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rinaldo Catta-Preta
- Department of Neurobiology, Physiology, and Behavior and Department of Psychiatry and Behavioral Sciences, Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Emily Ling-Lin Pai
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey D Mandell
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amanda Everitt
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anna Rubin
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rebecca A Krasnoff
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sol Katzman
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - David Tastad
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Alex S Nord
- Department of Neurobiology, Physiology, and Behavior and Department of Psychiatry and Behavioral Sciences, Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - A Jeremy Willsey
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bin Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Matthew W State
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Vikaas S Sohal
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Kavli Institute for Fundamental Neuroscience and Sloan-Swartz Center for Theoretical Neurobiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John L R Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
48
|
Varadarajan SG, Huberman AD. Assembly and repair of eye-to-brain connections. Curr Opin Neurobiol 2018; 53:198-209. [PMID: 30339988 DOI: 10.1016/j.conb.2018.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/24/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022]
Abstract
Vision is the sense humans rely on most to navigate the world and survive. A tremendous amount of research has focused on understanding the neural circuits for vision and the developmental mechanisms that establish them. The eye-to-brain, or 'retinofugal' pathway remains a particularly important model in these contexts because it is essential for sight, its overt anatomical features relate to distinct functional attributes and those features develop in a tractable sequence. Much progress has been made in understanding the growth of retinal axons out of the eye, their selection of targets in the brain, the development of laminar and cell type-specific connectivity within those targets, and also dendritic connectivity within the retina itself. Moreover, because the retinofugal pathway is prone to degeneration in many common blinding diseases, understanding the cellular and molecular mechanisms that establish connectivity early in life stands to provide valuable insights into approaches that re-wire this pathway after damage or loss. Here we review recent progress in understanding the development of retinofugal pathways and how this information is important for improving visual circuit regeneration.
Collapse
Affiliation(s)
- Supraja G Varadarajan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States; Department of Ophthalmology, Stanford University School of Medicine, Stanford, United States; BioX, Stanford University School of Medicine, Stanford, United States; Neurosciences Institute, Stanford University School of Medicine, Stanford, United States.
| |
Collapse
|
49
|
Duan X, Krishnaswamy A, Laboulaye MA, Liu J, Peng YR, Yamagata M, Toma K, Sanes JR. Cadherin Combinations Recruit Dendrites of Distinct Retinal Neurons to a Shared Interneuronal Scaffold. Neuron 2018; 99:1145-1154.e6. [PMID: 30197236 PMCID: PMC6284407 DOI: 10.1016/j.neuron.2018.08.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
Distinct neuronal types connect in complex ways to generate functional neural circuits. The molecular diversity required to specify this connectivity could be supplied by multigene families of synaptic recognition molecules, but most studies to date have assessed just one or a few members at a time. Here, we analyze roles of cadherins (Cdhs) in formation of retinal circuits comprising eight neuronal types that inform the brain about motion in four directions. We show that at least 15 classical Cdhs are expressed by neurons in these circuits and at least 6 (Cdh6-10 and 18) act individually or in combinations to promote specific connectivity among the cells. They act in part by directing the processes of output neurons and excitatory interneurons to a cellular scaffold formed by inhibitory interneurons. Because Cdhs are expressed combinatorially by many central neurons, similar interactions could be involved in patterning circuits throughout the brain.
Collapse
Affiliation(s)
- Xin Duan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Krishnaswamy
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jinyue Liu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yi-Rong Peng
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Masahito Yamagata
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Kenichi Toma
- Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|